MXPA00005325A - Hiv-1 tat, or derivatives thereof for prophylactic and therapeutic vaccination - Google Patents

Hiv-1 tat, or derivatives thereof for prophylactic and therapeutic vaccination

Info

Publication number
MXPA00005325A
MXPA00005325A MXPA/A/2000/005325A MXPA00005325A MXPA00005325A MX PA00005325 A MXPA00005325 A MX PA00005325A MX PA00005325 A MXPA00005325 A MX PA00005325A MX PA00005325 A MXPA00005325 A MX PA00005325A
Authority
MX
Mexico
Prior art keywords
tat
vaccine
cells
dna
protein
Prior art date
Application number
MXPA/A/2000/005325A
Other languages
Spanish (es)
Inventor
Ensoli Barbara
Original Assignee
Ensoli Barbara
Istituto Superiore Di Sanita'
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ensoli Barbara, Istituto Superiore Di Sanita' filed Critical Ensoli Barbara
Publication of MXPA00005325A publication Critical patent/MXPA00005325A/en

Links

Abstract

The present invention refers to Tat as the active principale for a prophylactic and/or therapeutic vaccine against HIV infection, the progression towards AIDS and the development of tumors and other syndromes and symptoms in subjects infected by HIV. Tat is in biologically active form either as recombinant protein or peptide or as DNA. More particularly, the invention refers to a vaccine based on HIV-1 Tat as immunogen, inoculated as DNA and/or recombinant protein or as peptides, alone or in combination with other genes or viral gene products (Nef, Rev, Gag) or parts thereof, or in combination with various immuno modulant cytokines (IL-12, IL-15) or with the gene coding for an immuno modulant cytokine or part thereof. Tat, Nef, Rev, Gag and the immuno modulant cytokines are administrated both as a mixture of recombinant proteins, peptides or fusion proteins (Tat/Nef, Tat/Rev, Tat/Gag, Tat/IL-12, Tat/IL-15) or as plasmid DNA.

Description

TAT DEVHI-10 DERIVED FROM THEM FOR PROPHYLACTIC AND THERAPEUTIC VACCINATION FIELD OF THE INVENTION The present invention relates to a prophylactic and / or therapeutic vaccine against HIV, against AIDS and against tumors and syndromes associated with HIV infection, which is used as immunogens, proteins, peptides or wild-type or mutated DNA of HIV Tat, only or associated with proteins, peptides or DNA of other viral products (Nef, Rev, Gag) or cytokines that enhance the antiviral immune response. The invention also relates to immunization with Tat or its derivatives by the use of autologous dendritic cells, mucosal immunization or ex vivo immunization of peripheral blood cells by co-stimulation with monoclonal antibodies against CD3 and CD28, and with the delivery of the immunogens mentioned above using erythrocytes or nanoparticles.
BACKGROUND OF THE INVENTION AIDS (acquired immunodeficiency syndrome) is caused by HIV (human immunodeficiency virus) and is characterized by immunodeficiency, tumors such as REF .: 120226 Kaposi's sarcoma (KS) and B-cell lymphomas, opportunistic infections and central nervous system disorders. Since AIDS is highly disseminated in the world and has a high mortality rate, one of the most important public health objectives is to develop a prophylactic and / or therapeutic vaccine against HIV or AIDS, most of the past and current strategies have used the viral envelope or its subunits as immunogens, but with unsatisfactory results due to the high variability of the viral envelope (ref 162, 112 - through this specification, reference will be made to several references in parentheses to describe more completely the state of the art to which the present invention belongs The complete bibliographic information for each citation is at the end of the specification, immediately before the claims). Therefore, as an alternative to sterilizing immunity, it is a common opinion that it should be sufficient to blog the progress of infection and the onset of the disease. In addition, immunoproive responses can be obtained using regions of HIV DNA as immunogens (reference 91, 17). Due to the published experimental data, the inventor believes that it is necessary to produce a vaccine that uses viral products other than env. In particular, the viral proteins to be used as immunogens must be more conserved among the HIV isolates, they must be able to induce an effective immune response, both humoral and cellular, and must have a vital function for the virus. Such products should be examined in the model in non-human primates (because their immune system is more similar to that of humans compared to phylogenetically more distant animals) and in which AIDS can develop after virus infection. The Tat protein of HIV-l has all the characteristics of being a good immunogen for vaccine purposes: it is conserved, it is immunogenic and essential in the early stages of viral infection. In addition, -Tat plays a key role not only in the replication, transmission and progress of viral infection, but also in the initiation and progression of tumors associated with AIDS, for example KS, which is the tumor most frequently associated with AIDS. , • and of the other syndromes and symptoms that develop after HIV infection. Tat is a protein of 86-102 amino acids, depending on the viral strain, encoded by two exons. Tat is produced rapidly after infection, localized in the nucleus and transactivates the expression of all viral genes by interaction with the "element that responds to Tat" (TAR) present in the LTR (Ref. 25). Tat also has a role in the virulence of HIV (Ref. 63, 113, 60, 84). The product of the first exon (amino acids 1-72) is conserved between different viral isolates (Ref. 112) and is sufficient for the transactivation of the HIV-1 products (Ref. 25). It contains 4 domains. The acid domain (amino acids 1-21) is important for Tat interaction with cellular proteins; the cysteine-rich region (amino acids 22-37) represents the transactivation domain. This region is the most conserved among the primary isolates (Ref. 108) of cysteine 22 with a glycine that suppresses the ability of Tat to transactivate the HIV-LTR (Ref. 166) the nucleus domain (amino acids 38-48) is also preserved and it is important for the function. Substitution of lysine 41 with a threonine inactivates the transactivating activity of Tat on HIV LTR (Ref. 70) and the basic domain (amino acids 49-57), rich in arginine and lysine, is necessary for the nuclear localization of Tat and binds specifically to its RNA target (TAR) (Ref. 25). In addition, the basic region is responsible for the binding of extracellular Tat to heparin and heparansulfate proteoglycans (HSPG) (Ref. 26). Mutations in the base region suppress such interactions. The carboxy terminal portion of Tat is not necessary for the transactivation of LTR, but contains an arginine-glycine-aspartic acid sequence), usually present in extracellular matrix (ECM) proteins, which is responsible for the binding of Tat to the integrin receptors a5ßx and avß3. These interactions mediate the effects of Tat on tumors associated with AIDS and on the immune, vascular and nervous system (Ref. 11, 42, 170, 25). During acute infection of T cells with HIV-1, or after transfection of the tat gene in COS-1 cells, the Tat protein is released in the absence of cell death in the extracellular environment (Ref. 40, 41, 25). The release of Tat from infected cells also occurs in vivo since extracellular Tat is present in the serum of infected subjects (Ref. 164) and in AIDS-KS lesions (Ref. 42). After release, part of the protein remains in a soluble form and part binds to the HSPG of the ECM. Tat binds to HSPG and can be recovered in soluble form by the addition of heparin. The binding with heparin is due to the basic Tat region, avoids the effects of extracellular Tat and protects the protein from oxidation. This characteristic has been used by us to purify Tat with a high biological activity (Ref. 26). Extracellular Tat can be internalized by cells, can migrate to the nucleus and transactivate viral gene expression (Ref. 49, 98, 100, 41). The internalization of Tat is produced by endocytosis mediated by the binding of the RGD region of Tat to C-sß-L and avß3 (Ref. 10, 42, Ensoli et al., Unpublished data) and / or by the basic region which joins HSPG. Tat can activate viral replication and virus transmission also through indirect mechanisms that involve the modulation of the expression of cellular genes which play a key role in the control of cell survival, and in the expression of inflammatory cytokines (IC ) with an effect on viral replication (Ref. 25). Beyond its importance in viral replication, Tat plays an important role in the pathogenesis of AIDS. Tat is able to modulate the survival and proliferation of infected or uninfected cells by causing activation or repression of cytokines, such as IL-2 (Ref. 123, 163, 31), or of genes with a key role in the cell cycle ( Ref. 145, 169, 164, 173). Its anti-or pro-apoptotic effects of Tat depend on numerous factors such as cell type, the fact that Tat is expressed by the cell or is added to the cell and on its concentration (Ref. 40, 41, 171). Tat is the factor responsible for the increased frequency and aggressiveness of KS in subjects infected with HIV-1 (Ref. 43, 33). KS is a tumor of vascular origin and is the most frequent neoplasm in HIV infected individuals. Tat induces KS cells and endothelial cells activated by IC to migrate, so that they express type IV collagenase, so that they invade ECM and proliferate, such mechanisms are necessary for angiogenesis and tumor invasion (Ref. 40, 41, 42, 2, 46). Such Tat effects are induced by IC, since they stimulate the expression of Tat receptors, tts? and avß3 (Ref. 10). Tat mimics the effect of ECM proteins, such as fibronectin and vitronectin, and both the RGD region and the basic region are necessary for the effects of extracellular Tat on KS cells, in angiogenesis and in the progress of KS. The ability of extracellular Tat to bind to its receptors in vivo in AIDS-KS lesions (Ref. 40) supports the idea that Tat is involved in the initiation and maintenance of KS associated with AIDS. In addition, mice transgenic for the Tat gene develop KS or other phenotypes depending on the expression level of the transgene (Ref. 160, 34). It has been suggested that Tat plays a role in hyperproliferative phenomena and in the pathogenesis of B lymphomas, which are frequently observed in seropositive subjects and in transgenic mice with Tat f-Ref. 157), through mechanisms that involve the increase of bel-2 and the expression of cytokines (Ref. 122). Other evidence confirms a probable role of Tat in oncogenesis (Ref. 72). Tat can also activate the expression of viral promoters, such as herpes virus and other viruses which reactivate in individuals with AIDS, promote the onset and progress of opportunistic infections (Ref. 25). Tat also seems capable of exerting neurotoxic effects both directly (through basic and RGD regions) and indirectly, through the induction of IC that has a toxic effect on neurons in the central nervous system or on the blood-brain barrier (Ref 25). Regarding the immune response to Tat, numerous studies suggest that antibodies against Tat play a protective role in controlling the evolution of the disease in vivo (Ref. 130, 135, 136, 149, 127). Furthermore, in vitro, antibodies against Tat not only suppress internalization, transcellular activation of Tat and viral infection (Ref. 41, 127), but also inhibit Tat-induced proliferation and migration of KS cells and formation of lesions similar to KS in mice (Ref. 40, 41, 42). Finally, our preliminary results show that antibodies against Tat are absent in subjects with AIDS-KS, which suggests that such subjects can not blog extracellular Tat activity. The development of a response against Tat mediated by cells in the initial phase of the invention is important for the control of the infection itself (Ref. 161, 133, 59) and there is an inverse correlation between the presence of CTL against specific Tat and the progress of the disease (Ref. 156). Such results are obtained in studies in macaques inoculated with SlVmac (Ref. 91, 158). In addition, recent data in mice of different species in which Tat has been inoculated either as a plasmid or as a protein show that it is possible to induce both a humoral and cellular response to the protein.
(Ref. 61). However, variability among several species of mice has been observed and such results have not been reproduced with the same immunogens in non-human primates (Ref. 124). The lack of reproductive capacity in the non-human primate model of the results of vaccination experiments performed on mice is frequent and possibly due to the immune system different from these two species which can lead to different immune responses with the same immunogen, as demonstrated for HIV Env protein. Therefore, candidate vaccines for humans should be tested in non-human primates and not only in lower species. The inventor considers that other viral proteins, or parts of them, must be associated with Tat to improve the specific immune response against HIV and could also be of benefit in vaccination against the onset of tumors and other pathologies and symptoms associated with infection. for HIV. Such products are the HIV Nef, Ref and Gag proteins. Nef is another important viral regulatory protein for the development of the disease (Ref 3, 48, 58). Nef occurs early after infection and is released in the extracellular environment (Ensoli, unpublished data). In the SlVmac / macaco system, the presence of Nef correlates with high viral replication and progress to AIDS (Ref. 71). Nef is more variable than Tat (Ref. 112). Nef is an immunogenic protein (Ref. 53, 32, 35, 151) and is capable of inducing CTL (Ref. 16, 36). In particular, an immunodominant region of Nef (region 73-144) has been identified, which is recognized by CTL in the majority of patients infected with HIV. Rev is a viral regulatory protein produced early during infection (Ref. 51, 119) and is released in the extracellular environment (Ensoli et al., Unpublished data). ReV is essential for replication of HIV and for the progress of the disease, and is encoded by two exons, which partially overlap the regions that code for Tat. ReV is a nuclear protein (Ref. 44) necessary for the expression of viral messenger RNAs that code for late proteins (Ref. 97). ReV is a highly conserved protein among the various viral isolates of HIV-1 (Ref. 111) and is immunogenic. In fact, it induces the production of specific antibodies directed against two functional domains of the protein (Ref. 120) during natural infection in man (Ref. 131) and subsequent inoculation in mice (Ref. 61). Lower levels of antibodies against Rev in the sera of infected individuals seem to correlate with progress to AIDS (Ref. 131). Rev can induce CTL in both man and monkey (Ref. 156, 158) and it has been reported that a specific response of CTL against Rev, early during infection, correlates inversely with the progress of the disease ( Ref. 156, 158). Another viral target is the gaa gene, which is expressed late during infection and codes for a group of highly immunogenic structural proteins of the capsid (Ref. 147). The antibody titers against Gag are elevated and stable during the asymptomatic phase of the infection, and reach very low levels when the infection progresses to fully expanded AIDS, in combination with the fall of the CD4 + lymphocytes and the presence of virus in the peripheral blood. (Ref. 174, 73). Gag proteins induce CTL activity early during infection, both in man and in primates (Ref. 103, 168), and their presence is significantly related to the control of the initial viremia and to the progress of the disease ( Ref. -175, 6, 134, 167, 92). Finally, the pl7 and p24 proteins contain immunodominant epitopes which are maintained in different HIV-1 and HIV-2 isolates, and are recognized by CTL (Ref. 89, 19, 114, 155, 115). The inventor considers that the cytokines or parts of them, such as IL-12 and IL-15 or other immunomodulatory cytokines such as IFNa IFNβ or other proteins that enhance the immunogenic effect of Tat, can be used as adjuvants in the formulation of a Tat vaccine. IL-12 is a strong immunoregulatory cytokine produced by antigen-presenting cells (APCs) such as B cells and dendritic cells (Ref. 154). IL-12 occurs early after HIV infection and has a proinflammatory action inducing NK cells and T lymphocytes to produce IFN? which activates phagocytes and promotes the induction of Thl lymphocytes. IL-12 plays a fundamental role in resistance to numerous infections caused by bacteria, fungi, viruses and shows a high antitumor activity. Several evidences suggest that viruses which induce immunosuppression, such as HIV and les virus, also act through mechanisms which suppress the production of IL-12 (Ref. 57, 50, 144). IL-15 is a pleiotropic cytokine expressed by non-lymphoid tissues, by activated monocytes / macrophages and by dendritic cells (DC) (Ref. 125, 66), IL-15 has an important role in the regulation of NK activity, in the proliferation of T lymphocytes and in CTL activity (Ref. 67, 24). IL-15 induces the expression of CTL against HIV antigens in the absence of IL-2 and functional CD4 + T lymphocytes (Ref. 68, 1). Also, similar to IL-2. IL-15 induces the expression of lymphocytes with cytotoxic activity ("lymphokine-disrupted" LAKs) and stimulates the production of IFN? in PBMC of seropositive patients (Ref. 93). UL-15 activates monocytes to produce guimokines, playing a role in the onset of inflammatory processes (Ref. 8). Recent studies have shown that the co-stimulation of CD4 + lymphocytes with paramagnetic spheres, coated with monoclonal antibodies against CD3 and against CD28 determines a logarithmic and polyclonal expansion of lymphocytes from HIV-infected patients (Ref. 82) without activating the replication and transmission of the virus. Such antiviral activity is a consequence of both the negative modulation of CCR5 expression, the co-receptor of HIV-1 monocitotropic strains (Ref. 23), and, to a lesser extent, the high levels of guimokins induced by antibody co-stimulation. monoclonal antibodies against CD3 and against CD28 (Ref. 132). The inventor considers that the possibility of expanding autologous lymphocytes from subjects infected with HIV in the absence of viral replication / transmission makes it possible to obtain an effective immunization ex vivo, described in the Examples, which can be highly useful in the development of vaccines against Tat. Within the different systems aimed at the generation of effective antiviral and antitumor vaccines, the inventor considers that the use of dendritic cells may be the key in the induction of an immune response to Tat. This is due to the fact that these cells are more efficient to present the antigen and the only ability to stimulate lymphocytes not previously exposed (naive), in the absence of adjuvants (Ref. 150). The use of dendritic cells replaces the function of several adjuvants that consist of the induction of a non-specific immune response (natural immunity) which, in turn, generates a strong primary specific response in the presence of an antigen.
Since the transmission of HIV infection occurs mainly at the mucosal level (genital and rectal in the adult, oral in the newborn), the inventor considers that the induction of protective immunity at the mucosal level is the primary objective. Recently many studies have shown the possibility of inducing mucous, local and systemic immunization. Particularly, it has been shown that the nasal and oral routes are more efficient in inducing an effective mucosal immune response even in distant sites, such as the genital mucosa (Ref. 138, 118). In particular, the inventor considers that the use of S. gordonii and Lactobacillus bacteria, modified to express the viral antigens mentioned above, may be a useful strategy to induce or potentiate a specific immune response at the mucosal level in monkeys and man. In fact, these bacteria are able to colonize the oral and vaginal mucosa of the mouse, and induce a specific and local systemic antibody response against heterologous antigens expressed on the surface of recombinant bacteria (Ref. 116, 104, 106, 121, 117, 139, 105, 107). Finally, these bacteria act as living vectors and can induce a prolonged stimulation of the immune system. In addition, the inventor considers that non-replicating and non-pathogenic recombinant viral vectors, such as herpes simplex virus type 1 (HSV-1) (Ref. 99), can be used to express viral proteins for systemic (intradermal) immunization and mucosal (oral, vaginal and nasal). In fact, these vectors can harbor large exogenous sequences (Ref. 52, 64), such as several HIV genes (regulators, accessories and structural). In addition, herpes vectors can be administered via the oral, nasal or vaginal route (Ref. 176, 75). The inventor considers that Tat (either as protein or DNA), alone or in combination with other immunogens described above, can also be inoculated by the use of new delivery systems, such as erythrocytes or nanoparticles. In particular, the inventor considers that it is possible to supply antigens bound to the membrane of autologous erythrocytes (Ref. 95, 96). Since these erythrocytes are removed from the blood by macrophages, professional antigen presenting cells, only after 120 days, this characteristic can be used for vaccine purposes. Finally, another supply strategy is the use of nanoparticles that can transport proteins and DNA (Ref. 172). The nanospheres are polymeric colloidal particles of varying guiding composition, varying from 10-1000 nm. Different substances (oligonucleotides, drugs, proteins, peptides, DNA) can be loaded on their surface or they can be absorbed in the particle and delivered in the cytoplasm or the nucleus of the cells from where they are released slowly. This allows the use of very similar amounts of the substance to be supplied. Based on the results described above, the inventor considers that immunization with Tat, either alone or in combination with other viral products or immunomodulatory cytokines, or parts thereof, in the presence or absence of adjuvants, can blog viral replication in subjects exposed after vaccination and in infected subjects, maintaining the infection in an abortive phase, which can be controlled more easily by the immune system. Therefore, the inventor considers that a vaccine based on Tat may be able to induce an immune response, both humoral and cellular, sufficient to blog or reduce the replication or transmission of the virus and therefore able to control the replication of the virus and of blogging T productive infection, progress to disease and the onset of tumors and other syndromes and symptoms associated with AIDS. Therefore, it is possible to use the Tat vaccine for both preventive and therapeutic purposes. In fact, a humoral response against Tat can neutralize the effects of extracellular Tat by reducing and limiting infection, while the cell-induced response against Tat as against other viral proteins enclosed in the vaccine formulation, can destroy cells infected with virus, which leads to control of the infection. This allows the necessary period of time - to the immune system to develop a complete response against all the viral components of the infectious virus in the absence of irreversible damage due to viral replication. The use of Tat as an immunogen has been described (WO 95/31999). However, the use of a biologically inactive protein is described; in addition, no evidence of the biological activity of the "native" Tat protein is shown in the same patent application. W09415634 refers to synthetic oligopeptides homologous to the signal sequences of Tat and Rev proteins, which can be used in the treatment of HIV infections. However, no preparation indications or tests are provided to demonstrate the effectiveness of the proteins described. Hinula et al., Vaccine, Vol. 15, 8, 874-878 (1997) describe vaccination tests in mice, based on vaccines containing Tat, however, the results obtained there, in lower species, do not they can be extrapolated directly to non-human primates. In addition, there is a strong technical prejudice against the use of Tat protein, biologically active, which is considered to increase viral replication in infected subjects and / or provide immunosuppression in seronegative or seropositive individuals (A. Tonelli: Aids, a vaccino per sperare . "The Republic", page 10, October 24, 1998).
As is clear from the above, despite the efforts made, an effective vaccine against HIV, based on Tat, has not yet been developed.
BRIEF DESCRIPTION OF THE INVENTION An object of the present invention is to design a Tat protein or Tat peptides or Tat DNA for use as a vaccine wherein Tat must be in its biologically active form. Another object of the invention is a protein or a peptide vaccine to be used in humans, prophylactically or therapeutically against AIDS, AIDS-associated tumors and syndromes and symptoms associated with HIV, and constituted by recombinant wild-type Tat protein or its mutants (Sections 1-5), expressed and purified as described, or with wild-type or mutated Tat peptides (Sections 1-7) administered alone or conjugated with T-tetanus T-toxoid epitope or other T-helper epitopes. Another objective of the invention is a vaccine as described above, in combination with Nef, Rev and / or recombinant HIV Gag, with proteins or peptides of Nef, Rev and Gag administered as Tat / Nef, Tat / fusion proteins. Rev and Tat / Gag or as part of these proteins.
Another objective of the invention is a vaccine as described above, in combination with recombinant proteins of immunomodulatory cytokines such as IL-12, IL-15 or other molecules or parts thereof capable of increasing the antiviral immune response, or a vaccine constituted by Tat / lL-12, Tat / IL-15 or Tat / other fusion proteins or parts of these, capable of increasing the antiviral immune response. Another object of the invention is a DNA vaccine, to be administered in humans, prophylactically or therapeutically, against AIDS, AIDS-associated tumors and HIV-related syndromes and symptoms, consisting of vectors which code for wild-type Tat or their mutants (Seq. 1-5) or parts thereof, inserted into the pCVO expression plasmid vector or other vectors. Another object of the invention is a DNA vaccine, as described in 4, in combination with the HIV Rev, Nef or Gag genes, or part of these, inserted into the pCVO vector, or a DNA vaccine administered as a vector. which coexpress Tat / Rev, Tat / Nef, Tat / Gag genes or parts of these. Another object of the invention is a DNA vaccine as described above, in combination with DNA that codes for IL-12 and IL-15 or other genes that code for immunomodulatory cytokines or parts of these, inserted in pCVO or other vectors, or a DNA vaccine administered as a vector that coexpresses Tat / IL-12, Tat / IL-15 or Tat / other molecules or parts thereof, capable of increasing the antiviral immune response. Another object of the invention is a Tat vaccine, such as a protein, peptide and / or DNA, alone or in combination as described above, for immunization with autologous dendritic cells by ex vivo treatment. Another object of the invention is a Tat vaccine, such as a protein, peptide and / or DNA, alone or in combination as described above, for mucosal immunization (nasal, oral, vaginal or rectal). Another object of the invention is a Tat vaccine, such as a protein, peptide and / or DNA, alone or combined as described above, for ex vivo immunization of peripheral blood cells from infected subjects, expanded through costimulation with monoclonal antibodies. against CD3 and against CD28 conjugated with paramagnetic spheres and re-infused into the host. Another object of the invention is a vaccine against Tat, such as a protein, peptide and / or DNA, as described above, combined with inhibitors of viral replication. Another object of the invention is a Tat vaccine, as already described, in combination with adjuvants which increases the immune response. Another object of the invention is a Tat vaccine, alone or in combination as already described, administered by specific delivery systems such as nanoparticles, herpes vectors, red blood cells, bacteria or any other delivery system by which can administer the vaccine described above, in all its combinations. Additional objects will be apparent from the detailed description of the invention.
BRIEF DESCRIPTION OF THE FIGURES Figure IA. Inhibition of the uptake of 10 ng / ml of rhodamine Tat proteins by preincubation of cytokine-activated endothelial cells with antibodies against integrin. Figure IA. Panel A, cells preincubated with buffer, incubated with BSA. Figure 1A. Panel B, cells preincubated with buffer, incubated with Tat. Figure IA. Panel C, cells preincubated with monoclonal antibodies CD 49e and CD29, incubated with Tat. Figure IA. Panel D, cells preincubated with monoclonal antibodies CD51 and CD61, incubated with Tat. Figure IA. Panel E, cells preincubated with antibodies against factor VIII (control antibodies), incubated with Tat.
Figure IB. Capacity of the Tat-cys22 protein (Tat22) purified to complete the transactivating activity of the wild type Tat protein, monitored by cat assays. Figure 2A. Production of specific IgG against Tat in monkeys vaccinated with Tat protein, determined by enzyme-linked immunosorbent assay (ELISA). The results obtained in two monkeys inoculated subcutaneously with 10 or 100 μg of recombinant Tat protein resuspended in 250 μl of autologous serum and 250 μl of RIBI. Figure 2B. Production of specific IgG against Tat in monkeys vaccinated with the Tat protein, determined by enzyme-linked immunosorbent assay (ELISA). Results for the control monkey (M3). Figure 3. Titration of antibodies against Tat in plasma of monkeys inoculated with 100- (MI) and 10 (M2) μg of recombinant Tat protein, described in Figure 2A and Figure 2B. Figure 4A. Mapping of Tat epitopes recognized by IgG against Tat from monkeys injected with 100 (Ml) and 10 (M2) μg of recombinant Tat protein, described in Figure 2A and Figure 2B. The average plasma results are shown diluted 1:50 for each peptide tested in duplicate. Figure 4B. Mapping according to figure 4A. The antibody titers in plasma are shown, expressed as the inverse of the highest dilution at which the test is still positive. Figure 5. Analysis of the specific humoral IgM response against Tat in monkeys inoculated with Tat protein, determined by ELISA. Figure 6. Analysis of the production of specific IgG against Tat in monkeys inoculated with Tat protein, tested by ELISA. Figure 7. Titration of antibodies against Tat in monkeys inoculated with recombinant Tat (10 μg) in the presence of RIBI (Ml-3) or alumina (M4-6) described in figure 6. Figure 8A. Epitopes of Tat recognized by IgG against Tat of inoculated monkeys as described in figure 6. The results refer to diluted samples 1:50 and are the average of wells in duplicate. Figure 8B. Tat epitopes, according to the figure 8A. The results refer to the plasma titration shown in Figure 8A and are expressed as the highest inverse dilution of plasma at which the test is still positive. Figure 9. Specific CTL analysis for Tat. Figure 10. Analysis of the delayed hypersensitivity response to Tat by skin test. Figure HA. Humoral IgG response to Tat in monkeys vaccinated with Tat DNA. The results obtained from two monkeys vaccinated with 200 (Ml) and 500 (M2) μg plasmid pCV-Tat are shown. Figure 11B. Tat humoral IgG response in monkeys vaccinated with DNA for Ta. The results are for the monocontrol (M3). Figure 12. Titration of antibodies against Tat in plasma of the inoculated M2 monkey i.d. with 200 μg of pCV-Tat. Figure 13. Analysis. of production of IgG against Tat in three monkeys (M9 to Mil) inoculated with 1 mg of pCV-Tat and in a control monkey (M12) inoculated with 1 mg of the control vector pCV-0. Figure 14. Kinetics of the proliferative response of PBMC from Macaca fascicularis to co-stimulation with monoclonal antibodies against CD3 and against CD28 in paramagnetic spheres (anti-CD3 / 28 spheres). Figure 15A. Antiviral effect of co-stimulation with anti-CD3 / 28 spheres in PBMC of Macaca fascicularis monkey MK 193. Figure 15B. Antiviral effect of co-stimulation with anti-CD3 / 28 spheres in PBMC of Macaca fascicularis monkey MK D91. Figure 15C. Antiviral effect of co-stimulation with anti-CD3 / 28 spheres in PBMC of Macaca fascicularis monkey MK 9301. Figure 15D. Antiviral effect of co-stimulation with anti-CD3 / 28 spheres in PBMC of Macaca fascicularis monkey MK 9401.Figure 16A. Functional characterization of dendritic cells (DC) obtained from peripheral blood of monkeys. Incorporation of 3H-thymidine on day 4 of haemogenic mixed leukocyte culture (AMLR). Figure 16B. Functional characterization of dendritic cells (DC) obtained from peripheral blood of monkeys. APC, such as DC and Mf, obtained as reported in Figure 16A are exposed with T lymphocytes from another monkey. DETAILED DESCRIPTION OF THE INVENTION The present invention relates to Tat as the active principle for the prophylactic and / or therapeutic vaccine against HIV infection, the progress towards AIDS and the development of tumors and other syndromes and symptoms in subjects infected with HIV. Tat, or Tat wild type, is in its active form or, more correctly, in its biologically active form (as explained below) either as a recombinant protein or peptide, or as DNA. More particularly, the invention relates to a vaccine based on Tat of HIV-l as an immunogen, inoculated as DNA and / or recombinant protein or as peptides, alone or in combination with other genes or other products of viral genes (Nef, Rev, Gag) or parts thereof, or in combination with several immunomodulatory cytokines (IL-12, IL-15) or with genes that code for an immunomodulatory cytokine or part thereof. Tat, Nev, Rev, Gag and immunomodulatory cytokines are administered either as a mixture of recombinant proteins, peptides or fusion proteins (Tat / Nef, Tat / Rev, Tat / Gag, Tat / IL-12, Tat / IL-15 ) or as plasmid DNA. In the present description, "Tat wild type" and "Tat in its active form" should be considered synonymous with "biologically active Tat". According to the present invention, the term "biologically active Tat" is used to mean a protein which is either picomolar or nanomolar (from 10 ng / ml or less to 1 μg / ml, preferably 0.1 ng / ml at 100 ng / ml). ml), is capable of: (i) entering and localizing in the nucleus of activated endothelial cells or dendritic cells, as measured in example IA; (ii) activate the proliferation, migration and invasion of Kaposi sarcoma (KS) cells and cytokine-activated endothelial cell protein (Ref. 40, 2); (iii) activate virus replication when added to infected cells, as measured by a) by the rescue of defective provirus without Tat in HLM-1 cells after the addition of exogenous protein (Ref. 41); b) by the transactivation of the gene expression for HIV-1 in cells transfected with an HIV-1 promoter-indicating plasmid protein (Ref. 41); (iv) induce in mice the development of KS-like lesions in the presence of angiogenic factors or inflammatory cytokines (Ref. 42). The inventor considers that it is fundamental for biologically active Tat, that one of points (i) or (ii) is verified, preferably both must be verified, and more preferably point (i) or point (ii) or both , with point (iii) and / or (iii) b) must be verified. The best results will be obtained when all points (i) to (iv) are verified. A Tat protein or Tat fragments with these characteristics are able to induce a cytotoxic and antiviral immune response in vivo. In fact, a biologically active Tat with the aforementioned characteristics is capable of binding specific cell surface receptors and of capturing them via these receptors. Tat uptake is essential to induce a cytotoxic response. Previous or developing studies are related to the development of a Tat-based vaccine, and have not used biologically active Tat protein with the characteristics mentioned above. A method for obtaining and managing biologically active Tat according to the present invention is described in Example 1. An immunization method using autologous dendritic cells treated ex vivo with recombinant Tat protein, or peptides (Tat, Nef, Rev. Gag) or with Tat protein and one or more immunomodulatory cytokines, or parts thereof, or transduced with eukaryotic vectors containing the tat gene alone or in combination with viral genes encoding Nef, Gag or Rev, or tat and the gene encoding an immunomodulatory cytokine or part thereof. Strategies to induce an immune response at the sal level have also been described. Tat or its peptides, alone or in combination with viral proteins and / or cytokines, is inoculated at the sal level to increase and induce the local immune response. The HIV-1 Tat protein or subunits thereof will also be used for the in vivo immunization of CD4 + and CD8 + lymphocytes isolated from peripheral blood of infected subjects. The antigen-specific cells for Tat will then be expanded in vitro via co-stimulation with monoclonal antibodies directed against CD3 and CD28 and reinfused. Finally, the use of Tat mutants, identified in the examples, to be used as immunogens, is also described as an alternative to wild type Tat. Tat mutants are: i) in the cysteine region (cys22) and ii) in the core region (lys41), iii) the mutant deleted in the RGD sequence; iv) the double mutant deleted in lysine 31 and RGD.
Alternatively, to the use of Tat mutants or Tat peptides (wild-type or mutated as the protein) in the case of therapeutic vaccination, inhibitors of viral replication will be used in conjunction with the immunogen. In this regard, the term "viral replication inhibitors" is understood to mean all molecules known to date, or aguelas which will be subsequently discovered (nucleosides and inhibitors which are not reverse transcriptase nucleosides, protease inhibitors, antisense RNAs). and, in general, all the molecules capable of blogging the expression of the gene for HIV) capable of reducing the blog of HIV replication. As previously stated, different immunization methods are described which use the Tatr protein, Tat peptides and DNA, in association with other genes or viral proteins, or parts thereof, or immunomodulatory cytokines, or genes that code for cytokines immunomodulators or parts thereof. By the term "parts thereof" is meant segments of genes or proteins, described above, whose efficacy in inducing the same immunogenic effects of the entire gene or protein is not demonstrated. In addition, since the efficacy of adjuvants in vaccine strategies is known, the present invention relates to the use of known adjuvants and those which will be subsequently discovered, administered together with Tat (protein or DNA) and combinations of Tat and the other genes or viral or cellular proteins. Similarly, the hypothesis is established that the efficacy of different Tat (protein or DNA) delivery systems and combinations of Tat and other viral or cellular genes or proteins to induce both systemic and local immune response to Tat (mucosal immunization) ). The results obtained from the inventor (unpublished) indicate that only the Tat protein, in its biologically active form, is capable of binding specific cellular receptors and the entire cell. This characteristic is at the base of the immune response in accessory cells and immune cells more generally and, according to the inventor, is of fundamental importance in inducing a much stronger immune response in comparison with which it is capable of inducing an inactivated protein. In conclusion, unlike the use of Tat activated as immunogen, proposed by some scientists, the inventor tries to use HIV-Tat, or its mutants, in its biologically active form, in order to induce a very strong immune response against HIV, capable to avoid infection or the development of the disease and allow efficient therapeutic strategies in individuals infected with HIV-1. According to the inventor, the vaccine can be delivered through the systemic (intramuscular, and i.d., subcutaneous, etc.) or local (mucosal) route. The last route is preferred when bacteria (see below) are used as delivery systems. The vaccine can be produced as follows.
Tat can be prepared according to example 1, and can be lyophilized and stored. At the time of its use, it can be resuspended in a biologically acceptable fluid such as serum, plasma or its fractions. Transformed cells, comprising a Tat expressing vector, or a vector expressing a Tat mutant, or part thereof, as previously described and cells which are cultured to express proteins, Tat will be isolated for use and will be included in the scope of this patent. It is intended that all Tafe variants (including all types and subtypes of HIV strains), with analogous or greater activity than that described above, are included in this invention. The present invention will now be described by means of its specific illustrative and non-restrictive examples, in which reference will be made to the appended figures.
DETAILED DESCRIPTION OF THE FIGURES Figure IA. Inhibition of the uptake of 10 ng / ml of rhodamine Tat proteins by preincubation of cytokine-activated endothelial cells with antibodies against integrin. The cytokine-activated human umbilical vein cells (HUVE) treated as described in the legend to Table 2A, are pre-incubated in serum-free medium containing buffer or antibodies and then incubated for 15 min at 37 ° C, with 10 ng / ml of Rhominated Tat or Rhominated BSA. Panel A. cells preincubated with buffer, incubated with BSA. Panel B. cells preincubated with buffer, incubated with Tat. Panel C. cells preincubated with the monoclonal antibodies CDw49e (against a5) and CD29 (against ßl), incubated with Tat. Panel D. cells preincubated with the monoclonal antibodies CD51 (anti-av) and CD61 (anti-ß3) incubated with Tat. Panel E. cells preincubated with anti-human factor VIII antibodies (control antibodies) incubated with Tat. Figure IB. It shows the ability of the purified Tat-cys22 protein (Tat22) to complete the transactivating activity of the wild type Tat protein monitored by cat assays. H3T1 cells containing the HIV-1 LTR-CAT reporter gene (Ref. 148) are incubated with 100 ng of wild type Tat protein, alone or in the presence of a molar excess of Tat-cys22 protein (1 μg). The transactivating activity of HIV-1 LTR and Tat as well as the ability of the Tat-cys22 protein to complete wild-type Tat has been determined 48 h after transfection by determining Tat activity in cytoplasmic extracts ( which correspond to 200 μg of protein), as described (Ref. 41), the percentages (%) are indicated in acetylation of 14C-chloramphenicol. Figure 2. Production of specific IgG against Tat in monkeys vaccinated with Tat protein, determined by enzyme-linked immunosorbent assay (ELISA). (A) shows the results obtained in two monkeys inoculated subcutaneously with 10 or 100 μg of recombinant Tat protein, resuspended in 250 μl of autologous serum and 250 μl of RIBI; (B) shows the results of the monocontrol (M3). The monkeys were inoculated at time 0 and after 2, 5, 10, 15, 22, 27, 32 and 37 weeks. Antibodies against Tat were also evaluated at week 41 in monkey M2, inoculated with 10 μg of Tat protein, and for monkey M3. The presence of antibodies against Tat in the plasma of vaccinated animals is evaluated by ELISA prepared and characterized as follows. The Tat protein is absorbed in 96-well PVC plates (100 ng / well in 200 μl of 0.05 M carbonate buffer, pH 9.6) for 12 h at 4 ° C. After 3 washes with PBS lx are Ca ++ and Mg ++ (PBS-A) which contains Tween 20 (0.05%), plasma diluted 1:50 in 200 μl of carbonate buffer are added (in duplicate) and the plates are incubated at 37 ° C. ° C for 90 '. The wells are then washed with PBS-A lx / 0.05% Tween, followed by the addition of 100 μl of the secondary antibody (diluted 1: 1000 in PBS-A lx / 0.1% Tween / 1% BSA) conjugated with horseradish peroxidase, for 90 'at room temperature. After 5 washes from the wells, 100 μl of substrate (ABTS 1 M, Amersham) is added for 30-45 ° at room temperature. The reading is made in the spectrophotometer (405 nm). Each ELISA includes a polyclonal serum against rabbit Tat (positive control) diluted 1: 200 to 1: 6400, and preimmune plasma (negative control) diluted 1:50. The limit value was calculated as the mean of the optical densities (D.O.) of the negative monkey plasma +3 standard deviations (S.D.) obtained in all experiments with preimmune plasma. The results shown are the average of duplicate wells. > 2, 7 indicates that the optical density values went out of scale. Figure 3. Titration of antibodies against Tat in plasma of monkeys inoculated with 100 (Ml) and 10 (M2) μg of recombinant Tat protein, described in figure 2. ELISA was carried out as described in the figure 2 and plasma assays were performed (in duplicate) followed by scalar dilutions from 1:50 to 1: 25,600. The values in the ordinate represent the inverse of the highest plasma dilution at which the test was still positive. The limit value was calculated for each dilution and corresponds to the D.O. average preimmune plasma of all monkeys in all experiments, + 3 S.D. (standard deviations). Figure 4. Mapping of tat epitopes recognized by IgG against Tat of monkeys injected with 100 (Ml) and 10 (M2) μg of recombinant Tat proteins, described in figure 2. For the epitope mapping, ELISA was carried out using 8 synthetic peptides corresponding to the amino acids Tat (* aa) 1-20, 21-40, 36-50, 46-60, 52-72, 56-70, 65-80, 73-86. 100 μl of each peptide (10 μg / ml in PBS-A / 0.1% BSA) are absorbed on a 96-well PVC plate for 12 hours at 4 ° C. The plates are then washed and incubated with 100 μl of PBS-A / 3% BSA for 2 hours at 37 ° C. After incubation, the plates are washed with PBS-A / 0.05% Tween 20 and then with 50 μl of plasma, diluted in PBS-A and 3% BSA, and added to each well. The ELISA test is then continued as described in Figure 2. Plasma is obtained at week 37 after primary immunization. The cut-off values, calculated for each peptide and for each plasma dilution, correspond to the D.O. average preimmune plasma in all exments + 3 S.D. (A) shows average plasma results 1:50 diluted for each peptide tested in duplicate; __ (B) shows the plasma antibody titers shown in (A) _, expressed as the inverse of the highest dilution, at which the test was still positive. Figure 5. Analysis of the specific IgM response against Tat in monkeys inoculated with Tat and determined by ELISA. Three monkeys (Ml-3) were inoculated subcutaneously with 10 μg of recombinant Tat protein resuspended in 250 μl of autologous serum and 250 μl of RIBI; 3 monkeys (M4-6) were inoculated subcutaneously with 10 μg of recombinant Tat protein resuspended in 250 μl of autologous serum and 250 μl of alumina; 2 control monkeys were inoculated subcutaneously with RIBI (250 μl and 250 μl of autologous serum) (M7) or with alumina (250 μl and 250 μl of autologous serum) (M8). The monkeys were inoculated at time 0 and after 2, 6, 11 and 15 weeks. The presence of antibodies was investigated at 2, 6, 11 and 15 weeks. The ELISA method is described in Figure 2. In this case, the plasma of the animals was tested (in duplicate) at a 1: 100 dilution and goat IgM was used against monkey serum (diluted to 1: 1000) conjugate with horseradish peroxidase as the "secondary antibody." The cutoff value was calculated as the average (+2 SD) of the OD values of the preimmune plasma.The results are the average of the D values, 0. (at 405 nm) of Two wells subtracted from the limit value (? OD 405) Figure 6. Analysis of the production of specific IgG against Tat in monkeys inoculated with Tat, tested by ELISA Three monkeys (Ml-3) were inoculated with 10 μg of recombinant Tat protein resuspended in 250 μl of autologous serum and 250 μl of RIBI, 3 monkeys (M4-6) were inoculated with 10 μg of recombinant Tat protein resuspended in 250 μl of autologous serum and 250 μl of alumina, two control monkeys were inoculated with RIBI ( 250 μl and 250 μl of autologous serum) (M7) or with alumina (250 μl and 250 μl of its ero autologous) (M8) Monkeys were inoculated at time O and after 2, 6, 11, 15, 21, 28 and 32 weeks. At week 36, monos Ml to M6 were inoculated with 16 μg of Tat protein resuspended in 200 μl of ISCOM and 300 μl of PBS. Antibodies were also evaluated at week 40 and 44. The ELISA method and the determination of the cut-off value are described in figure 2. The results show reference to samples diluted 1:50. The term > 2.7 indicates that the value of D.O. It's off the scale. Figure 7. Titration of antibodies against Tat in monkeys plasma inoculated with 10 μg of recombinant Tat in the presence of RIBI (Ml-3) or alumina (M4-6) described- in figure 6. The results are shown for each plasma as the inverse of the highest serum dilution at which the test is still positive. Figure 8. Tat epitopes recognized by IgG against Tat of monkeys inoculated with 10 μg of recombinant Tat protein in the presence of RIBI (Ml to M3) or alumina (M4 to M6) described in figure 6. Plasma is obtained at week 21 after primary immunization. The ELISA method and the cutoff determination are described in Figure 4. The results in (A) refer to samples diluted 1:50 and are the average of wells in duplicate. The results in (B) refer to the plasma titration shown in (A) and are expressed as the highest inverse of the plasma dilution at which the test is still positive.
Figure 9. Tat-specific CTL analysis. The test is carried out as described in table 5. An example is shown at week 36 for monkey Ml, injected subcutaneously with 10 μg of Tat and RIBI, as described in figure 6. The tables ( control) correspond to cells incubated with BLCL target cells without pulses; the diamonds correspond to the cells incubated with the BLCL target cells pulsed with Tat (1 μg / 250,000 cells). Figure 10. Analysis of the delayed hypersensitivity response to Tat by skin test. The Tat protein (5, 1 and 0.2 μg), resuspended in 150 μl of PBS containing 0.1% BSA or the buffer in which Tat is resuspended, is inoculated intradermally (i.d.) in a shaved area of the animal's back. The area is photographed at time 0 and after 24, 48 and 72 hours. The control monkeys were only inoculated with shock absorber. An example of the M2 monkey is shown (week 15), inoculated with 10 μg of Tat and RIBI, described in figure 6. The positive reaction to Tat is evident at 48 hours after the skin test. Figure 11. Humoral IgG response to Tat in a mouse (ml) inoculated i.d. with 200 μg of plasmid pCV-Tat resuspended in 150 μl of PBS-A, in two sites close to the axillary lymph nodes; one monkey (M2) was injected with 500 μg of pCV-Tat, resuspended in 250 μl of PBS-A, intramuscular in two sites of the back; the control monkey (M3) was not inoculated with Tat DNA, but received, as a control of specificity, repeated skin tests with Tat. The monkeys were injected with pCV-Tat at a time 0 and after 5, 10, 15, 22, 27, 32 and 37 weeks. Finally, after 42 weeks, reinforcement was applied to the monkeys with 16 μg of recombinant Tat protein resuspended in 200 μl of ISCO and 300 μl of PBS. Antibodies were evaluated at weeks 2, 5, 10, 15, 22, 27, 32, 37, 42, 48 and 58. The antibody response against Tat in plasma (diluted 1:50) was analyzed by ELISA as described in figure 2. The results are the average of the DO of wells in duplicate. (A) shows the results obtained from two monkeys vaccinated with 200 (Ml) and 500 (M2) μg of the pVC-Tat plasmid. (B) shows the results of the control monkey (M3). Figure 12. Titration of antibodies against Tat in monkey plasma M2 inoculated i.d. with 200 μg of pCV-Tat. The ELISA test is described in Figure 2. The results in the ordinate are expressed as the inverse of the highest dilution at which the test is still positive. Figure 13. Analysis of IgG production against Tat in 3 monkeys (M9 to Mil) inoculated with 1 mg of pCV-Tat and a control monkey (M12) inoculated with 1 mg of the control vector pCV-0. DNA is resuspended in 1 ml of PBS-A and injected intramuscularly in two sites on the spine. The monkeys are inoculated at time 0 and after 6, 11, 15, 21, 28 and 32 weeks. At week 36, minuses M9 to Mil received a booster with 16 μg of recombinant Tat protein resuspended in 200 μl of ISCOM and 300 μl of PBS. The presence of antibodies against Tat was evaluated in weeks 2, 6, 11, 15, 21, 28, 32, 36, 40 and 44. The determination of ELISA and the cut is described in figure 2. Figure 14. Kinetics of the proliferative response of PBMS of Macaca fascilularis to co-stimulation with monoclonal antibodies against CD3 and against CD28 in paramagnetic spheres (anti-CD3 / 28 spheres). The PBMC were deleted from the CD8 positive subpopulation by immunomagnetic methods. Subsequently, half of the lymphocytes suppressed against CD8 were stimulated with PHA and IL-2 (40 U / ml) starting "from day 3; the remaining part was allowed to adhere on the antibodies of spheres coated with anti-CD3 / 28, so that a population of lymphocytes lacking CD8 and positive CD3 / 28 is obtained. IL-2 (40. U / ml) is added to this cell fraction starting from day 10 of culture. The cells were counted and their viability determined every 2-3 days. The ratio of spheres: cells remained constant. The number of cells is reported at different time points. Figure 15. Antiviral effect of co-stimulation with anti-CD3 / 28 spheres in PBMC of Macaca fascicularis. Lymphocytes lacking CD8 and lacking CD8 + CD3 + / CD28 + CD8 +, obtained from 4 monkeys (Figures 15A to 15D) by the methods described in Figure 14, were stimulated as described in Example 7. The two fractions were infected in vitro in the day O with 0.1 MOI of SIVmac25l / 63M. The stimulation was performed with PHA and IL-2, added from day 3, and with anti-CD3 / 28 spheres without the addition of IL2-hexogen. Viral production was evaluated by determining p27 levels (ng / ml) in cell supernatants on days 6 and 12 after infection, as described in example 7. (Light gray, PHA + IL-2, in dark gray, anti-CD3 / 28 spheres in PBMC CD8"/ CD3 + / CD28 +) Figure 16. Functional characterization of dendritic cells (DC) obtained from monkey peripheral blood. (A) Incorporation of at 3H-thymidine in the day 4 of a mixed allogeneic leukocyte culture (AMLR) to compare the antigen presenting function (APC, determined as the induction of allogeneic T cell proliferation) of DC and macrophages (Mf) obtained from PBMC of Macaca fascicularis after separation in Percoll gradient and adhesion in plastic.The non-adherent cells are removed and the adherent cells are induced to mature in DC by adding GMCSF (200 ng / ml) and IL-4 (200 units / ml) every 3 days. of the culture medium (RPMI, 10% FCS) is removed and replaced ye with fresh medium every 3 days. After 6-7 days, a morphological change is observed in cytokine-induced cells, which adgoes a typical DC phenotype (loss of adhesion, formation of groups, fingers), also verified when determining typical membrane markers (data not shown) . The monocytes were not induced by cytokine and were cultured in the same medium, which can be replaced every 3 days. The cells maintained the characteristics of monocyte-macrophages, such as adherence. On day 7, both cell populations were exposed with T lymphocytes from a human blood donor, purified by a Ficoll and Percoll gradient and by adherence and then frozen. Cell proliferation assays were carried out in a 48-well plate. 500,000 T lymphocytes were stimulated with 500 DC or M0 (ratio T: APC = 100: 1). The culture was maintained for 4 days and fixed aliquots of the cell suspension were transferred in 96-well plates, in triplicate. Then 1 μC was added; of 3 H-thymidine for 16 hours, and counts per minute (cpm) of the incorporated precursor were determined, with a β counter. (B) The APCs, such as DC and M0, obtained as reported in Figure 16A, were exposed with T lymphocytes from another monkey, obtained as previously formed for the human donor. The greater ability to present the antigen is a typical characteristic of DC compared to macrophages. APC were added in scalar concentrations to T lymphocytes in order to evaluate the proliferative responses obtained at different ratios of T: APC (DC or M0). The following examples can be considered illustrative and not limiting of the scope of the invention.
Example 1. Expression, purification and characterization of wild type Tat protein (IIB isolate), mutated Tat proteins and wild type Tat peptides In the past, many difficulties have been encountered in purifying and maintaining the biological activity of the Tat protein due to its ability to oxidize, aggregate and lose activity. This is due to the high amounts of cysteine residues which can form intramolecular and intermolecular bonds, which therefore modify the conformation of the native protein (Ref. 159, 41). The tat gene cDNA has been cloned (Sec. 1, Example 2) into the pL-syn vector, provided by Dr. JF DeLamarter and B. Allet (Glaxo Institute for Molecular Biology SA, Geneva, Switzerland), and used for the expression of the protein in E. coli. In order to obtain efficient immunization with Tat for vaccine purposes, the inventor considers it essential to obtain a biologically active Tat protein as described in the section "Detailed Description of the Invention". Therefore, the methods of production and purification of Tat, described in this example and in the following examples IB, 2 and 3, describe the necessary procedures and controls to obtain biologically active Tat protein, which is an effective immunogen to protect of HIV infection, AIDS or the development of HIV-related diseases. A first method, which will be used to obtain an active protein, is based on successive steps of high pressure liquefied chromatography and ion exchange and lysis chromatography (Ref: 15, 41). The protein obtained by these methods is more than 95% pure and active (Ref. 41, 42). However, good reproducibility from one batch to another is not obtained due to the oxidation of the protein, which is the main problem in commercial preparations of Tat. Due to our observations that the basic region of the Tat protein has a strong affinity for heparin and that heparin prevents its oxidation, we use affinity chromatography with heparin and define a new Tat purification protocol, as described by Chang et al. al., (Ref. 26). Cells (10 g in weight) of E. coli expressing Tat in 40 ml of lysis buffer (20 mM disodium phosphate, pH 7.8, 2.5% glycerol, 0.2 mM PMSF, 5 mM DTT, 50 mM mannitol) are subjected to sonication. 10 mM ascorbic acid, 500 mM NaCl) using an Ultrasonic Liguid Processor (model XL2020, Heat System Ine). with three downloads, each of 20 seconds. The lysate is centrifuged at 12,000 g for 30 min, and the supernatant is incubated for 1 hour at room temperature with 2 ml of heparin resin Sheparose, prewashed with the lysis buffer. The resin is loaded onto a glass column and washed with the lysis buffer until the protein is undetectable in the washing medium. The bound material is eluted with lysis buffer which contains 2M NaCl and the eluate is collected in 1 ml fractions. The homogeneity of the protein eluted by gel electrophoresis (SDS-PAGE) is analyzed. The purified protein is stored lyophilized at -70 ° C and resuspended in a degassed buffer before use. The biological activity of the purified Tat protein is evaluated, according to the above protocol, by a "rescue" test of viral infection in HLM-1 cells, derived from HeLa-CD4 + cells, which contain defective provirus in the Tat gene, obtained and described by Sadaie et al., (Ref. 1, 40). The "rescue" viral infection assay, described by Ensoli et al., (Ref. 41) consists of supplementing the lack of Tat expression in HLM-1 cells (2 x 105) with the addition of exogenous Tat protein ( 2 μg / ml) and when evaluating the viral replication by the determination of the p24 antigen released in the culture medium 48 hours after the addition of the exogenous Tat protein, by commercial eguipos. The results of the "rescue" experiments described by Chang et al., (Ref. 26), show that the Tat protein purified with this method is active and that this method of purification is better, easier and less expensive both for the purity and the biological activity of Tat, when compared with the previously described methods (Ref. 40, 41, 42). Different preparations of recombinant Tat, purified as described above, were inoculated in the presence of Freund's adjuvant in mice and rabbits, according to standard protocols (Ref. 4). Table 1 shows the results of the antibody response induced by the immunization.
Table 1 Analysis of the response of the specific antibody against Tat in sera from mice and rabbits immunized with the Tat protein recombine DO-ELISA antibody / Tat Western against Tat 1: 500 1: 1000 1: 2000 Blot Rabbit 0.651 0.400 0.175 Mouse 0.502 0.240 0.150 The recombinant Tat protein produced in E. coli was used to immunize mice and rabbits according to standard immunization protocols (Ref. 4). The sera of the immunized animals were analyzed by ELISA to determine the presence of antibodies against Tat by using three dilutions of serum (1: 500 to 1: 2000). The results are the measurement of the readings at 405 nm of two rabbits and three mice. In addition, the sera were subjected to a Western blot with the recombinant Tat protein (100 ng). The results of Table 1 demonstrate that recombinant Tat, prepared by us, is capable of inducing an antibody response in both species of animals, as demonstrated by ELISA and Western blot, which uses the recombinant Tat protein. Such antibodies were able to inhibit the internalization and biological activities of Tat (Ref. 40, 41, 42). The pL-syn vector and the Tat protein purification protocol were used to express and purify the Tat mutants described in example 2. The biological activity of the mutated and purified Tat proteins is measured by the "rescue" assays of the viral infection in HLM-1 cells, proliferation assays of KS cells and in mice in vivo, as described above for the wild type Tat protein. In addition, mutated Tat proteins were tested in the presence of wild-type Tat (at serial concentrations) to verify the naive transdominant effect on viral replication. The vector pL-syn and the purification protocol were used to express and purify fusion proteins of this type: Tat (wild type or mutants thereof) / IL-12 or Tat (wild type or mutants thereof) / I1-15 , or parts thereof, or Tat (wild type or mutants thereof) / other molecules (or parts thereof) capable of enhancing the immune response to Tat alone, or associated with other viral products. Recombinant fusion molecules are manufactured by using sequences and primers described in Examples 2 and 3. As an alternative, synthetic peptides, which correspond to the Tat or other viral or cytokine regions to be used in combination with Tat, they are used as immunogens. The Tat peptide sequences are ": Pep. 1. MEPVDPRLEPWKHPGSQPKT Pep. 2. ACTNCYCKKCCFHCQVCFIT Pep. 3. QVCFITKALGISYGRK Pe. 4. SYGRKKRRQRRRPPQ Pep. 5. RPPQGSQTHQVSLSKQ Pep. 6. HQVSLSKQPTSQSRGD Pep. 7. PTSQSRGDPTGPKE The Tat mutant peptides will contain the same amino acid substitutions of the mutated Tat proteins described in Example 2. The peptides will be used in combination with the peptide which represents the universal auxiliary T-epitope of tetanus toxoid or with other peptides which represent the auxiliary T epitopes (Ref. 77).
Example 1A Uptake of picomolar concentrations (10 to 100 ng / ml) of biologically active Tat by activated endothelial cells, which is mediated by integrin receptors When normal endothelial cells are activated in vitro with inflammatory cytokines, they become capable of responding to the effects of extracellular Tat and ei-fto is due to the induction of integrins a5 ^ 1 and vß3 (Ref. 9, 10). Similarly, inflammatory cytokines (IC) or bFGF increase integrin expression in endothelial cells in vivo, and this leads to a synergistic KS promoter effect when biologically active Tat is inoculated into mice, simultaneously or after bFGF (Ref. ). In addition, activated IC endothelial cells adhere to APC function. In this example, it is shown that the IC-activated endothelial cells capture the biologically active Rho-Tat protein and the Tat protein is more efficient and is mediated by integrin receptors. Due to the difficulty of observing the internalization of very low concentrations of cold Tat, the protein is etched with rhodamine '(Ref. 98). The rhodamine Tat still shows activation of KS cell proliferation in the same concentration range as compared to untagged Tat, which indicates that the ethylation procedure does not compromise its biological function. Capture experiments were carried out as follows: human umbilical vein cells (HUVE) are grown and treated for 5 days with IC, as described (Ref. 9, 46). The cells are then tripzinized, plated on 8-well plates (Nunc Inc., Naperville) at 0.5 x 10 B cells per well, and incubated for 18 hours in medium containing 15% fetal bovine serum (FBS), in the presence of IC. Serum free medium (SF, RMPI, BSA 1%, 0.1% antibiotics, fungizone) were added and the sections were preincubated for 2 h at 4 ° C. Fresh medium, containing serial dilution of rhodamine Tat, was added to the cells and the cells were incubated at 37 ° C for the indicated time. The negative controls were BSA rhodamine in the same buffer as Tat. Cells were fixed in acetone-methanol (1: 1) cooled with ice, and Tat uptake was visualized and photographed using fluorescence microscopy. The results are evaluated by comparison of fluorescence of the samples with the negative control and it is classified from 0 to ++++ based on the amount of uptake, without previous knowledge of the sample codes. To investigate the pathways by which Tat is taken by activated endothelial cells, experiments were performed using HUVE cells activated with a wide range of exogenous Tat concentrations, such as those previously used to induce HUVE or the growth of KS cells ( 10-50 ng / ml), or transactivation of HIV by adding protein to cells transporting the HIV-1 promoter or the provirus (0.5 to 1 μg / ml). In these experiments, for consistency, with uptake inhibition experiments (see below), cells were preincubated at 4 ° C for 2 hours with medium lacking fetal bovine serum. This preincubation does not alter the subsequent uptake of rhodamine Tat. With rhodamine Tat, the uptake and translocation of the protein to the nucleus or nucleolus of HUVE and activated cells begins to be evident in the next 15 minutes of incubation with as little as 10 ng / ml of rhodamine Tat. The density of Tat taken up in the cells is increased in a dose-dependent manner and in a time-dependent manner. Rolled BSA or buffer does not show signals and is commonly used as negative controls. To determine if Tat uptake by cells Activated HUVE is mediated by the same integrins that are expressed on KS cells, inhibition experiments were performed by preincubating IC-activated endothelial cells with Tat f ío (competitor), physiological ligands for these receptors such as fibronectin (FN) or vitronectin (VN), or by preincubation of the cells with monoclonal antibodies directed against RGD binding regions of the a5ßl and avß3 receptors. The experimental procedure is reported briefly. After plating on 8-well plates, HUVE cells are incubated with medium containing 15% FBS for 18 hours and then incubated with SF medium containing unlabeled Tat (cold competitor) (Table IA), FN, VN ( Table IB), or monoclonal antibodies directed against the sequence that binds RGD of the FN or VN receptors (a5ßx and avß3, respectively), or monoclonal antibodies directed against human factor VIII (control antibodies) (Figure IA) for 2 hours to 4 hours. ° C. The cells are then incubated with Rhominated Tat for the indicated periods of time. The control consists of cells treated with SF medium only for 2 hours at 4 ° C, and incubated with rhodamine BSA. The cells are fixed, visualized, photographed and the results are graded as indicated above.
TABLE Inhibition of the uptake of 100 ng / ml and 1 μg / ml of Tat rhodamine by HUVE activated by cytokine by preincubation of the cells with 1 μg / ml of unlabeled Tat1. aHUVE cells are cultured as previously described (Ref. 40). IC from CD4 + cells transformed with type II virus T-limf other human (HTLV-II) or T cells stimulated with phytohemagglutinin are obtained, and supernatants (1: 8) are used to activate HUVE cells (passage 8-14). ) for 5 days, as previously described (Ref. 9, 46). This supernatant contains interleukin-la (IL-la) and -B (IL-lß), tumor necrosis factor-a (TNF-a) and -B (TNF-β), and interferon-? (INF-?) (Ref. 9). The Tat protein is rhodamine in the lysine residues essentially as described (Ref. 98). Briefly, 50 μg of recombinant Tat (2 mg / ml) is brought to pH 9.0 by the addition of 2.5 μl of 1 M Na 2 CO 3. 2.5 μl of TRITC I ng / ml in dimethyl sulfoxide (DMSO) is added and the reaction is allowed to proceed for 8 hours at 4 ° C. TRITCs that did not react are suspended by the addition of 2.5 μl of 0.5M NH4C1, decreases, the pH at 7.0 using 1M HCl, and the rhodamine Tat is dialysed against two changes of 50 M Tris-HCl, pH 7. 0, 1 mM di thiotrei tol (DTT) to remove inactivated TRITC. They are used as negative controls BSA or PBS, ruminated in the same way. Rhominated Tat is tested for AIDS cell growth activity-KS as described, to ensure that biological activity is maintained (Ref. 40). HUVE cells are preincubated for 2 hours with serum-free medium or 1 μg / ml unlabeled Tat in serum-free medium, incubated with 100 ng / ml or 1 μg / ml rhodamine Tat for 60 minutes and visualization of the uptake is visualized. of Tat by fluorescence microscopy. Negative controls (uptake +/-) are preincubated with serum free medium, followed by incubation with rhodamine BSA.
TABLE IB.
Inhibition of the uptake of 10 ng / ml of Tat rhodamine by HUVE activated by cytokine, by preincubation of the cells with an excess of FN or VN3 a HUVE cells are preincubated for 2 hours with serum-free medium or FN or VN in serum-free medium, incubated with 10 ng / ml of rhodamine Tat for 60 minutes and Ta t uptake is visualized by fluorescence microscopy. Negative controls (uptake +/-) are preincubated with serum free medium, followed by incubation with rhodamine BSA. Tat uptake is inhibited by cold Tat (table 1A), by FN or VN (table IB) or by pretreatment of the cells with monoclonal antibodies directed against the RGD binding regions of both the FN, a5ßx receptor and the VN receptor , avß3 (figure A). The intensity of fluorescence in the cells is reduced to levels observed with the negative control and no inhibition is observed before the incubation of the cells with monoclonal antibodies directed against human factor VIII, used as a negative control, indicating that the inhibition is specific ( figure A). The uptake and nuclear localization of 100 ng / ml of Tat is inhibited by preincubation of the cells with the monoclonal antibodies directed against the RGD region of the a5ßx receptor and the avß3 receptor. However, in both cases, the inhibition is not complete. These results indicate that the uptake of picomolar concentrations of Tat is mediated by the same integrins involved in cell adhesion to Tat (Ref. 10). However, at a higher concentration of extracellular Tat (such as> 100 ng / ml), a pathway not mediated by integrin is responsible for the uptake of part of the protein. In contrast to these results, the recruitment of Tat iodinated with lymphocytes and epithelial cell lines is shown to be linear and is dependent on the concentration of Tat in the medium and does not compete or does poorly for an excess of cold Tat, indicative of the lack of relationship or involved receptor (Ref. 98). However, the Tat concentration range in the medium in this study is approximately 1-100 μg / ml (Ref. 98), much higher than that necessary to observe Tat uptake by cells that respond to their biological activity, such as activated primary endothelial cells. In addition, Tat iodization can damage its structure and its uptake by cells and these authors do not show results regarding the biological activity of iodinated Tat. These results, which have not been published, show that Tat uptake occurs for at least two days, depending on the concentration of the protein. At low concentrations of Tat (10-100 ng / ml), Tat uptake is mediated by ace receptors. And av 3 by interacting with the RGD sequence of the protein, while at a higher concentration of extracellular Tat, an integrin-independent pathway is more important. The integrin-mediated uptake of picomolar concentrations of Tat by endothelial cells activated by IC indicates a fully active protein capable of entering antigen-presenting cells, such as activated endothelial cells and dendritic cells, which initiate the iniffune response.
Example 2 Construction and characterization of mutated Tat genes We produced 19 mutants in different Tat regions by means of site-specific mutagenesis or by deletion. The sequence of each mutated DNA is controlled by sequencing. The cDNAs of the mutated tat genes are cloned into the PstI site of the vector pCVO, described in example 3. Each mutant is cotransfected as described by Ensoli et al., (Ref. 41) in COS-1 cells or in the line of Jurkat T cells with the HIV-1 LTR-CAT plasmid, in which the CAT reporter gene is activated by HIV-1 LTR. The results of these experiments, unpublished, are presented in Table 2.
TABLE 2 Effects of Tat mutants on the transactivation of LTR-CAT HIV-1 and blocking effect (negative transdominant) on the wild type activity of Tat MUTANT Transactivating activity Transdominant activity (% inhibition) Mean (times) (min-max values) Mean CYS22 0, .09 (0.021-0.22) 21 THR23 0. .36 (0.16-1) THR 23A 0. .30 (0.16-0.78) ASN 24 0, .34 (0.34-0.82) ASN 24A 0. .42 ( 0.45-0.95) TYR26 0. .14 (0.08-0.19) LYS 28/29 0. .52 (0.19-1.04) CYS 30 0. .30 (0.045-0.65) CYS 31 0. .60 (0.27-1.09) ) PHE 32 0. .31 (0.077-0.097) LYS 33 0. .04 (0.0027-0.068! 46 GLU 35 0. .31 (0.19-0.43) PHE 38 0. .05 (0.043-0.057) 98 LYS 41 0 .04 (0.025-0.061) 97 TYR 47 0.58 (0.31-0.08) 57 A 0.35 (0.26-0.44) TAT-RGD 0.94 (0.73-1.15) TAT-KGE 1.11 (0.67-1.49) TAT type 1 1 wild The results are given as the increase in activation of the CAT activity values induced by wild-type Tat (times = 1). b The results are expressed as percent (%) of inhibition of wild-type Tat activity. From the results presented in Table 2, it is evident that for most mutants, the transactivating effect of HIV-1 LTR is reduced or absent, with the exception of the mutant RGD, which has a similar activity to Tat wild type. We selected 4 mutants (cys22, lys33, phe38, lys41), which have the lowest transactivating activity (almost zero) and we determined the negative transdominant effect on the transactivating activity of wild type Tat. For this purpose, COS-1 cells were cotransfected with each vector containing a Tat mutant, and the pCV-Tat vector (in a molar ratio of 10: 1) in the presence of the HIV-1 LTR-CAT vector. As shown in Table 2, the mutants lys41 and phe38 almost completely inhibit Tat activity, while mutants lys33 and cys22 partially inhibit Tat activity. However, the recombinant protein cys22 (described in the following example 3) competes with the wild-type Tat protein in the transactivation of LTR-CAT of HIV-1 (FIG. IB). A mutant in the cysteine region (cys22), one in the core region (lys41), one deleted from the RGD sequence (RGD?) And a double mutant that contains the mutation in lys41 and the deletion of the RGD sequence (Iys41- RGD?) Were the ones that were selected. The sequence of the Tat insert and of the mutants selected for vaccination is reported in the following. A series of tat mutants are described prepared by: 1) substitution of a base to obtain an amino acid substitution, and 2) deletion of a base to obtain a deletion of the corresponding amino acids. Substitutions and deletions were obtained by site-directed mutagenesis. The sequences of the wild type Tat gene and that of the Tat gene mutants, reported as follows, were inserted into the plasmid vector pCVO, as described above. With Sec. 1, the HIV-l gene sequence is presented, starting with clone BH-10 and its derivative protein. The mutant sequence cys22 (and its derivative protein) is presented with Sec. 2, represented by a substitution of the nucleotide thymine (T) at position 66 from end 51 with the guanine nucleotide (G). This substitution originates, in the sequence derived from amino acids, a substitution of a cysteine (C in the code of a letter) at position 22 at the amino terminus, with a glycine (G in the code of a letter). The mutant sequence lys41 (and its derivative protein) is represented by Sec. 3, represented by a substitution of the thymine (T) nucleotide at position 123 from the 5 'end with the nucleotide cytokine (C). This substitution originates, in the sequence derived from amino acids, a substitution of a lysine (K in the code of a letter) at position 41 from the amino terminus, with a threonine (T in the code of a letter). With Sec. 4, a sequence of the mutant RGD (and its derived protein) represented by the deletion of the nucleotide sequence CGAGGGGAC from nucleotide 232 to nucleotide 240, from the 5 'end of the wild-type tat gene is presented. This provides a deletion of the amino acids arginine-glycine-aspartic acid (RGD in one letter code) at positions 78-80 from the amino terminus. With Sec. 5 shows a sequence of the double mutant Iys41-RGD? (and its derived protein) caused by the combination of the mutants described above.sequence of wild-type tat (Seq. 1) 5 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCMCATTGCCA AGTTTGTTTCATAACAAAAGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTTCTCTAT CAAAGCAGCCCACCTCCCAATCCCGAGGGGACCCGACAGGCCCGAAGGAAT AG 3 * Amino acid sequence NH2-MEPVDPRLEPWKHPGSQPKTACTNCYCKKCCFHCQVCFITKALG ISYGRKKRRQRRRPPQGSQTHQVSLSKQPTSQSRGDPTGPKE-COOH Nucleotidse sequence of the cys22 mutant (Sec. 2 Y 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCGGTACCAATTGCTATTGTAAAAAGTGTTGCTTTCATTGCCA AGTTTGTTTCATAACAAAAGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTTCTCTAT CAAAGCAGCCCACCTCCCAATCCCGAGGGGACCCGACAGGCCCGAAGGAAT AG 3' Amino acid sequence NH2-MEPVDPRLEPWKHPGSQPKTAGTNCYCKKCCFHCQVCFITKA LGISYGRKKRRORRRPPQGSQTHQVSLSKQPTSQSRGDPTGPKE-COOH Nucleotide sequence lys41 (Sec. 3) 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCMCATTGCCA AGTTTGTTTCATAACAAACGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTTCTCTAT C? AAGC? GCCCACCTCCCAATCCCGAGGGGACCCGACAGGCCCGAAGGAAT AG 3' Amino acid sequence NH2-MEPVDPRLEPWKHPGSQPKTACTNCYCKKCCFHCQVCFITTALG ISYGRKKRRORRRPPQGSQTHQVSLSKQPTSQSRGDPTGPKE-COOH Nucleotidic sequence of the mutant RGD? (Sec. 4) 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCTCATTGCCA AGTTTGTTTCATAACAAAAGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTTCTCTAT CAAAGCAGCCCACCTCCCAATCCCCGACAGGCCCGAAGGAATAG_3_' Amino acid sequence NH2 -MEPVDPRLEPWKHPGSQPKTACTNCYCKKCCFHCQVCFITKALG ISYGRKKRRORRRPPQGSQTHQVSLSKQPTSQSPTGPKE-COOH Nucleotide sequence of the mutant Iys41-RGD? (Sec. 5) 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCMCATTGCCA AGTTTGTTTCATAACAAACGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTTCTCTAT CAAAGCAGCCCACCTCCCAATCCCCGACAGGCCCGAAGGAATAG_3_' Amino acid sequence NH2-MEPVDPRLEPWKHPGSQPKTACTNCYCKKCCFHCQVCFITTALG ISYGRKKRRORRRPPQGSQTHQVSLSKQPTSOSPTGPKE-COOH Example 3. Construction characterization of DNA immunogens The DNA molecules for the inoculation of animals are inserted into the plasmid vector of 6.4 kb pCVO (Ref. 5).
• This plasmid comprises two SV40 replication origins, the adenovirus major late promoter AdMLP) and the splice sequences of adenovirus and mouse immunoglobulin genes, the cDNA of the mouse dihydrofolate reductase gene (dhFr) and the signal of polyadenylation SV4.
The site for the restriction enzyme PstI is located in the 3 'part of AdMLP, and represents the site in which the exogenous gene of interest is cloned. The tat gene (261 base pairs) cDNA (Sec. 1, example 2) of HIV-1 is derived from clone HIV-1 BH10 (Ref. 126) and codes for a protein of 86 amino acids long. The vector pCV-Tat (Ref. 5) is obtained by cloning Tat cDNA into the PstI site of pCVO, activated by AdMLP. The choice of this vector is based on the fact that AdMLP induces a higher expression and release of Tat, with respect to the other eukaryotic promoters such as, for example, the promoter of the immediate early region of cytomegalovirus (CMV) as demonstrated by Ensoli et al. to the. (Ref. 41) and which is presented in table 3.
TABLE 3. Expression, subcellular localization, release of Tat activity in C0S-1 cells transfected with pCV-Ta t and CMV-Ta ta a COS-1 cells (5 x 10 6) are transfected by electroporation with 30 μg of pCV-Tat, CMV-Tat or control DNA. 48 hours after transfection, Tat expression is evaluated by immunohistochemistry with monoclonal antibodies against Tat (the values provided are the measurement of the percentage values of positive cells) and by location of nuclear Tat and toplasmatic cy. The presence of intracellular and extracellular Tat is analyzed by radioimmunoprecipitation in the cellular extracts (500 μl) and in the culture medium (4 ml) and the subsequent densiometric reading (Gelscan XL; Pharmacia) of the Tat bands precipitated. The intracellular Tat activity is measured in cell extracts of COS-1 cells cotransfected with vectors expressing Tat, or the control vector, and the HIV-1 LTR-CAT plasmid; The activity of extracellular Tat is measured on the proliferation of AIDS-KS cells (determined by the 3 H-thymidine incorporation assay) in the culture medium (diluted 1: 2 and 1: 4) of the cells transfected with plasmids that express Tat or the control plasmid. The results are the average of five independent experiments. b Density analysis in the immunoprecipitated Tat protein band. The values are expressed on an arbitrary scale, the total detected minimum value (intracellular and extracellular Tat) is 10. c ", negative; +, 50% of positive Tat cells; ++, 50-100% - of positive T cells. dActivity CAT after 20, minutes of incubation with respect to the control vector, the activation value of which is considered 1. The growth of AIDS-KS cells is measured by a 3 [H] -thymidine incorporation assay (standard deviation, SD: 12%) .The supernatants of the cells transfected with the control DNA have an incorporation of 3 [H] - thymidine of 1, 400 cpm (SD: 11.5%). The culture medium derived from activated T-lymphocytes (positive control) has an incorporation of 3 [H] -thymidine of 2.400. cpm (SD: 10%). Table 3 shows that in the cells transfected with pCV-Tat, compared to the cells transfected with CMV-Tat, the percentage of cells positive for Tat and the total content of Tat are higher, the amount of Tat released is much higher and is related to the total and cytoplasmic content of Tat, and biological activity of extracellular Tat in AIDS-KS cells that have grown therefore is greater. Such results show that the vector pCV-Tat codes for the biologically active protein, induces high levels of expression of the tat gene and can release from the cells quantities of Tat much higher than those of the CMV-Tat vector.
The pCVO vector is also used for the expression of the nef, rev and gag genes of HIV-1 and the genes that code for the cytokines IL-12 and IL-15. The nef cDNA (618 base pairs, strain NL43) (Ref. 112), rev 348 base pairs, strain NL43) (Ref. 95) and the gag genes (1500 base pairs, strain NL43) (Ref. 95) of the cDNAs of IL-12 (Ref. 165) or of the IL-15 genes (Ref. 56) are amplified by the polymerase chain reaction (PCR) technique by using specific complementary primers for the first 15 nucleotides of the 5 'region (forward primer) (Sec. Pl, 03, P5, P7, P9) or for the last 15 nucleotides of the 3' region of the gene (reverse primer) (Sec. P2, P4, P6, P8, PIO) . In addition, each primer, both direct and reverse, comprises the sequence for the restriction enzyme PstI to allow the cloning of the amplified product into the pCVO vector. After cloning, the sequence of the inserted genes is controlled by DNA sequencing. The vector pCVO is also used for the coexpression of Tat with other HIV-1 viral genes (rev, nef or gaq) or with the genes that code for cytokine IL-12 or IL-15. For this purpose, the 261 base pair HIV-l Tat cDNA (Seq.1, Example 2) is amplified by PCR with a forward primer which includes the sequence for the restriction enzyme PstI (Seq. Pll) and a reverse primer complementary to the last 15 nucleotides of the tat gene (Sec. P12). Viral genes (nef, rev or gag) or genes that code for the cytokines IL-12 or IL-15 are amplified by a direct primer which also includes a sequence of 15 bases complementary to the 3 'region of tat, that allows the gene to be in frame with the tat gene (Sec. P13, P14, P15, P16, P17), and a reverse primer that includes the sequence for the restriction enzyme PstI (Sec. P2, P4, P6, P8) , CHEEP) . Subsequently, a third PCR reaction is carried out in which the DNA template is represented by the amplified products of the tat gene and the gene of interest. The forward primer is represented by the primer used to amplify tat (Sec. Pll) and the reverse primer, by one used to amplify the gene of interest (Sections P2, P4, P6, P8, PIO). The amplified tat / gene of interest is purified with agarose gel, digested with PstI and cloned into pCVO. After cloning, the sequence of the inserted genes is controlled by DNA sequencing, while protein expression is determined by transfection, as described above (Ref. 41). The sequences of the primers mentioned above are: Sec. Pl. Direct primer Rev: 5 'ATGGCAGGAAGAAGC3' Sec . P2 Reverse primer Rev: 5 'CTATTCTTTAGTTCC3' Sec. P3. Nef direct primer: 5 'ATGGGTGGCAAGTGG3' Sec. P4. First reverse Nef: 5 'TCAGCAGTCCTTGTA3' Sec. P5. Direct primer Gag: 5 'ATGGGTGCGAGAGCG3' Sec. P6 Reverse primer Gag: 5 'TTATTGTGACGAGGG3' Sec. P7. Direct primer IL-12: 5 'TGTGGCCCCCTGGG3' Sec. P8. Reverse primer IL-12: 5 'TTAGGAAGCATTCAG3' Sec. P9 Direct primer IL-15: 5 'ATGAGAATTTCGAAA3 • Sec. PÍO. Reverse primer 1L-15: 5 • TCAAGAAGTGTTGAT3 'Sec. Pll. Tat direct primer: 5 'ATGGAGCCAGTAGAT3' Sec. P12. Reverse primer Tat: 5 • CTATTCCTTCGGGCC3 'Sec. P13. Tat / Rev direct primer: 5 * GGCCCGAAGGAAATGGCA GGAAGAAGC3 'Sec. P 14. Tat / Nef direct primer: 5' GGCCCGAA6GAAATGGGT GGCAAGTGG3 'Sec. P15. Tat / Gag direct primer: 5 'GGCCCGAAGGAAATGGGTGCG AGAGCG3' Sec. P16. Tat / IL-12 direct primer: 5 'GGCCCGAAGGAAATGTGGC CCCCTGGG3' Sec. P17. Tat / IL-15 direct primer: 5 '-GGCCCGAAGGAAATGAGAAT TTCGAAA3' Example 4. Inoculation of healthy Macaca fascicularis of a Tat protein vaccine: Safety evaluation, tolerability, specific immune response protective efficacy against exposure to the virus Tolerability, safety and the ability to induce a specific immune response (humoral and cellular) and protection against exposure to recombinant Tat protein vaccine virus, produced by The method described and purified through heparin affinity columns was determined in the experimental model of cynomorph monkey (Macaca fascicularis). In order to induce a broad immune response, we use aluminum phosphate (Alum) that has been tested in many models and is the only one approved for human use. Among the particulate adjuvants we use RIBI (which belongs to the group of emulsifiers or which is composed of monophosphoryl lipid A, dimhalic trehasola and a main structure of the bacillus Calmette-Guerin bacterial wall) (Ref. 7, 109). In the first pilot experiment, we evaluated tolerability, safety and the ability to induce a specific immune response (humoral and cellular). Therefore, 3 monkeys were inoculated according to the following protocol: monkey 1 (Ml) was inoculated with 100 μg of the recombinant Tat protein, resuspended in 250 μl of autologous serum and 250 μl of RIBI, subcutaneously in a place; Monkey 2 (M2) was inoculated with 10 μg of recombinant Tat protein, resuspended in 250 ml of autologous serum and 250 μl of RIBI, subcutaneously at one site; and monkey 3 (M3) is the control monkey and was not inoculated. 10 ml of blood were extracted from all the monkeys on days -42 and -35 before the first vaccine inoculation, in order to determine the basal parameters. The serum and plasma samples were frozen at -20 ° C or -80 ° C and subsequently used to resuspend the protein inoculum. Monkeys 1 and 2 were inoculated at time 0 and after 2, 5, 10, 15, 22, 27, 32 and 37 weeks. The immunization protocol was interrupted at week 37 for monkey Ml and at week 41 for monkey M2. The animals were sacrificed to study the immunological parameters in various organs and tissues (vessel and lymph nodes), such as the evaluation of the presence of a proliferative response to Tat and the activities of CAF and CTL against Tat. CAF activity is the antiviral activity mediated by CD8 + lymphocytes, and is not restricted by ßl CPH and is cytolytic. On the same days as the inoculation of the immunogen, 10 ml of blood was taken from each animal to perform laboratory tests (guimicofísico analysis, electrolytes, leukocytes, platelet counts and quantification of hemoglobin), the evaluation of immunological parameters such as the presence of specific immunoglobulins (IgM, IgG, IgA), Thl (IL-2, IFN?) and Th2 type (IL-4, IL-10) cytokine levels, the production of chemokines (RANTES, MlP-la and MlP -lß), the lymphocytic phenotype (CD4, CD8, CD3, CD14, CD20, CD25, CD56, HLA-DR, CD45RA), the presence of specific cytotoxic activity (CTL), the presence of antiviral activity (CAF) and the presence of total antiviral activity (TAA) mediated by PBMC and by autologous serum. In addition, to evaluate the in vivo presence of a cell-mediated immune response, all vaccinated and control monkeys were subjected to the Tat skin test. The results of this experiment are as follows. No alterations of the chemophysical, hematological and behavioral parameters were observed. In monkeys vaccinated without control, no signs of inflammation and neovascularization were detected at the inoculation sites. These results indicate that the Tat protein is well tolerated by animals and that it is non-toxic at the doses administered and by the given inoculation route. In minoo Ml and M2, the presence of Tat-specific IgG antibodies was detected at week 5 after the first inoculation. At week 37, IgG was detected against Tat up to a plasmatic dilution of 1: 6400 in both monkeys and, at week 41, up to a plasma dilution of 1: 12800 in the M2 monkey. The results are shown in Figures 2 and 3. In the control monkey M3, antibodies against Tat were detected with low titer, probably induced by repeated inoculations of a small amount of Tat that was injected into this monkey to control the specificity of the skin test reactions. In the Ml and M2 monkeys, the antibodies against Tat were directed mainly against the amino terminal region (amino acids 1-20) of Tat, with a titer of 1: 3200 (Figure 4). In the M2 monkey, vaccinated with 10 μg of Tat, antibodies directed against the amino acids 36-50 or 46-60 of Tat were also detected, with titers of 1:50 and 1,100, respectively (figure 4). The ability of monkey serum to neutralize Tat was determined by in vitro assays that measure the inhibition of rescue of HIV-1 replication in HLM-1 cells after the addition of exogenous Tat protein, as previously described (Ref. 41). These assays demonstrated that the plasma of the Ml and M2 monkeys, at week 27 after the first inoculation, block the replication of the virus induced by exogenous Tat, determined by quantification of the p24 antigen in the culture supernatants. Conversely, the preimmune plasma of the same monkeys does not block Tat activity (Table 4) TABLE 4 Neutral activity of monkey plasma in the replication of virus induced by extracellular Tat3 a Plasma-neutralizing activity was determined in HLM-1 cells (HeLa-CD4 * cells containing an integrated copy of an HIV-1 provirus defective in the tat gene). HLM-l cells were seeded at 6 x 10 5 cells / well in 24-well plates and incubated at 37 ° C for 16 hours. The cells were washed twice with PBS, containing 0.1% bovine serum albumin (BSA) and cultured for 48 hours with 0.3 ml of fresh medium in the presence of recombinant Tat protein and an equal volume of animal plasma, extracted in the time O (preimmune plasma) or week 27 (immune plasma). Negative controls are represented by cells treated only with accumulated pre-immune plasma, with accumulated immune plasma or with PBS containing 0. 1% BSA (PBS + 0. 1% BSA), without Ta t. In all the control samples, no effects are observed on the rescue of virus replication. Each plasma is tested in duplicate. The presence of virus released by the cells is assayed by quantification of the p24 Gag antigen, using a commercial p24 antigen capture ELISA (NEN-Dupont). The results are expressed as the percentage of inhibition of virus rescue [measured for each plasma as the average value of p24 (pg / ml) in two wells] by the immune plasma, compared to the preimmune plasma (0% inhibition). ). The Ml and M2 monkeys were vaccinated with the recombinant Tat protein (100 μg and 10 μg, respectively), resuspended in 250 μl of autologous serum and 250 μl of RIBI, and injected subcutaneously in a serum. The results indicate the presence of a proliferative response to Tat at week 22 (table 5) in monkeys vaccinated with the recombinant Tat protein, which is higher in the M2 monkey that received 10 μg of recombinant Tat protein in each reinforcement.
TABLE 5 Proliferative response to Tata a Ficoll-density gradient-isolated PBMC were plated at a concentration of 2 x 10 5 cells / well in triplicate in a 96-well flat-bottom plate, cultured in RPMI-1640 supplemented with fetal bovine serum (FCS9 10% and stimulated with Tat (1 or 5 μg / ml), 4 μg / ml of PHA or tetanus toxoid (TT) to which the monkeys were vaccinated.The non-stimulated controls were incubated in RPMI, medium 10% FCS. of cell proliferation was measured on day 5 by incorporation of 3 [H] -thymidine as previously described (Ref 39, 22) The results are expressed in the stimulation index and calculated as follows: average of the test sample (cpm) / average of the control (cpm) The values higher than 2.5 were considered positive.The monos Ml and M2 were immunized subcutaneously with 100 μg or 10 μg of recombinant Ta t resuspended in 250 μl of autologous serum and 250 μl of RIBI, M3 represents a control mono. shown in Table 6, no cytotoxic activity was detected for Tat in Ml and M2 monkeys immunized with recombinant Tat.
TABLE 6 Analysis of cytotoxic activity to Tat (CTL) a a Ficoll density centrifugation-isolated PBMC were resuspended at a concentration of 1 x 10 7 cells / ml in RPMO 1640 supplemented with 10% heat-inactivated FCS and seeded in a 24-well plate (500 μl per well) for 12 hours. hours at 37 ° C in the presence of 1 μg of. Tat. One day later, cells incubated without Tat were centrifuged at 1500 rpm and resuspended in 50 μl of RPMI 1640 supplemented with 10% FCS, incubated for 3 hours at 37 ° C with 1 μg of Tat, washed, resuspended in 500 μl of fresh medium and are added to the well containing the previously stimulated PBMC. On day 2, the cells are diluted with 1 ml of medium containing IL-2 (2 IU / ml) and cultured for 14 days. The autologous B lymphocytes isolated from each monkey before the vaccine protocol were used as target cells (BLCL). For this end, isolated PBMC were seeded by Ficoll density centrifugation on day 35, at a concentration of 3 x 10 5 cells / well in a 96 well plate and cultured for 2 or 3 weeks in the presence of 50% of a medium collected from a cell line that produces Papiovirus as previously described (Ref. 28). 10 B cell lines obtained for each animal expand and freeze. To test the toxicity, the Del fia cytotoxic test (Wallac, Turku, Finland) was used based on the separated fluorescence time (Ref. 12, 13, 14). For this purpose, BLCL is cultured at a concentration of 1 x 106 cells / 200 μl of RPMI 1640 supplemented with 10% FCS containing 4 μg of Tat for 12 hours at 37 ° C. As the control, another aliquot of BLCL antologa is incubated with the same medium, without Tat. BLCL are washed and resuspended in 1 ml of RPMI 1640 supplemented with 10% FCS containing 5 μl of fluorescent fluorescence ligand and incubated for 15 min at 37 ° C, according to the manufacturer's instructions. After 5 washes, the BLCL are resuspended at a concentration of 5 x 10"cells / ml, and centrifuged immediately in order to harvest the supernatant used to measure the background level. duplicate at a concentration of 2.5 x 10 4 cells / 100 μl in medium containing IL-2 and properly diluted in a 96-well plate, 5 × 10 3 target cells / 100 μl (cultured with or without Tat) is added to each well. The objective: voter relations were 1:50, 1:25, 1: 12.5, 1: 6.25, 1: 3.125.
The PBMC and target cells (pulsed or non-pulsed with Tat) are incubated for 2 hours at 37 ° C with: i) 20 μl of Tri tón 5% to measure the maximum release, ii) 100 μl of growth medium to detect spontaneous release, iii) 200 μl of supernatant of target cells to detect the background level. At the end of the incubation period, the plates are centrifuged, 20 μl of each supernatant is transferred to a new plate and incubated in the presence of 200 μl of a Europium solution included in the kit. Fluorescence is measured after 20 min of incubation with a time-resolved fluorescence reader (Victor, Wallac, Turku, Finland). The specific activity of CTL is measured as follows:% specific release = [(average sample detection - background) - (spontaneous release - background)] (maximum release - background) - (spontaneous release - background)] x 100. The test is considered positive when the specific release of Ta t is greater than 4% at most of the elector: target ratios tested. The value of 4% is arbitrary and established based on previous control experiments. ND: Not determined. The M2 monkey is immunized subcutaneously with 10 μg of recombinant Tat resuspended in 250 μl of autologous serum and 250 μl of RIBI. M3 represents a monkey with troll. ND: not done. * PBMC are isolated from peripheral lymph nodes when they have been sacrificed to M2. In addition, the results show that at weeks 22, 27 and 37, the presence of soluble antiviral activity mediated by CD8 + T lymphocytes (CAF), measured as the capacity of cell supernatants of monkeys with CD8 + T lymphocytes to inhibit acute infection. of SHIV 89. 6P chimeric virus in CEMxl74 cells, or to control the reactivation of chronic HIV-1 infection in OM-10-1 cells (table 7). CAF activity is generally observed in vaccinated monkeys, as compared to control animals.
TABLE 7 Analysis of the presence of soluble antiviral antiviral activity mediated by CD8 + T lymphocytes (CAF) a To PBMC of monkeys vaccinated with 100 μg (Ml) and 10 μg (M2) of recombinant Tat protein and from a control monkey (M3), which was not vaccinated but presented repeated skin tests with Tat that are isolated by gradient of Ficoll density. Cultures enriched with T CD8 + T lymphocytes are isolated from PBMC by magnetic beads against CD8 (Dynabeads, Dynal, Norway) according to the manufacturer's instructions. The purity of the crops is monitored by FACS analysis using a series of antibodies directed against specific cell markers (CDS, CD4, CD8).
Cultures rich in CD8 + are planted (in duplicate) to x 105 cells / 500 μl per well in 48-well plates, previously coated with a monoclonal antibody against CD3 (2.5 μg / ml, BioSurce International, Camarillo, CA) for 12 hours at 4 ° C and grown in RPMI 1640, containing 10% fetal bovine serum and IL-2 (20 U / ml). 250 μl of medium is collected every three days, for 2 weeks, and replaced with an equal volume of fresh medium. Cell supernatants are centrifuged, filtered (0.45 μm) and stored at -80 ° C.
The cellular supernatants derived from all points in time, with the exception of the first, accumulate and the presence of antiviral activity is tested as well as their capacity to inhibit viral replication in the two systems, represented by acute and chronic infection, respectively. . For the acute infection system, the cell line CEM x 174 is used, which is derived from the line of human B cells 721,174 fused with the human T cell line CEM (Ref. 143). 2 × 10 5 cells are incubated in polypropylene tubes with or without 200 μl of CD8 + supernatants, prepared as described above, for 2 hours at 37 ° C. Cells are washed three times with fresh medium, seeded at 2 x 104 cells per well, in 96-well plates, and incubated in 200 μl with (treated cells) or without (untreated cells) different volumes (50 μl, 5 μl and 0.5 μl) of culture supernatants derived from T CD8 + lymphocytes from monkeys injected with the vaccine or the Monkey control. After infection, aliquots of the culture supernatants are harvested every three days, and replaced with an equal volume of complete medium to which previously added CD8 + supernatant of the vaccinated and control monkeys has been added. The results shown in the table correspond to day 7 after infusion and are expressed as percentages (%) of inhibition of viral replication of cells treated with CD8 + supernatants derived from vaccinated monkeys compared to non-infected cells. treated. Viral replication is determined by measuring RT values, as described (Ref. 54) or p27 Gag values by ELISA, in the cellular supernatants collected at each point in time. The OM-10-1 cell line (Ref. 20, 21) is used for the chronic infection system, which represents a line of human T lymphocytes chronically infected with HIV-1. Cells are seeded (in duplicate) at 5 x 10 4 cells / 200 μl per well, in 96-well plates, in the presence of antibodies against TNFβ (40 μg / ml) with or are different volumes (50 μl, 5 μl, 0.5 μl) of cell supernatant of CD8 + T lymphocytes derived from monkeys vaccinated or control. The cells are activated to proliferate by PMA (10 ~ 7 M). After 24 hours, aliquots of the culture medium are collected to determine viral replication by measuring RT or p24 Gag levels by ELISA. The results are represented as% inhibition of reactivation of infection in treated cells, compared to untreated cells. The results of the acute and chronic infection shown in the table refer to cells treated with 5 μl of supernatant derived from CD8 + cell cultures. ND: not done. The analysis of delayed hypersensitivity (DTH) by means of a skin test shows that the vaccinated monkeys (Ml and M2) as the control (M3) are negative (table 8).
TABLE 8 Tata skin test Tat 1 and 5 μg in 150 μ and PBS-BSA 0.1 or buffer alone, are inoculated intradermally in a previously affected dorsal area of the vaccinated monkeys and control (control of specificity of the response) at 10 weeks, 15, 22, 27, 32 and 37 after the first immunization. The Ml and M2 monkeys were vaccinated with recombinant Tat protein (100 μg and 10 μg, respectively) in 250 μl of autologous serum and 250 μl of RIBI, injected subcutaneously in a serum. The M3 monkey is a control monkey that has not been vaccinated. The appearance of a nodular erythema after 48-72 hours is suggestive of a delayed hypersensitivity reaction (DTH): ++, 0 > 5m; +, 0 > 1 -4 mm, +/-, eri subject without hardening; - 0 < 1 mm The results of this pilot experiment indicate that the Tat recombinant protein, produced and purified according to a protocol described by us, is not toxic at doses of 10 and 100 μg administered subcutaneously. In addition, the Tat protein induces a specific and extensive immune response with antiviral activities, both humoral and cell-mediated. An immune response against stronger and specific Tat is observed in the monkey M2, vaccinated with 10 μg of recombinant protein. In addition, the RIBI adjuvant does not show any apparent signs of toxicity in vaccinated monkeys. Based on these results, a second pilot experiment was designed in order to determine the effects of immunization with 10 μg of Tat combined with RIBI or alumina adjuvants. The monkeys were injected subcutaneously at one site, according to the following protocol. The monkeys Ml-3; 10 μg of recombinant Tat protein in 250 μl of autologous serum and 250 μl of RIBI. Monkeys M4-6: 10 μg of recombinant Tat protein in 250 μl of autologous serum and 250 μl of alumina.
Mono M7: 250 μl of RIBI and 250 μl of autologous serum (control mono). Mono M8: 250 μl of alum and 250 μl of autologous serum (control monkey). 10 ml of blood is extracted from each monkey on day -9 preceding the first immunization in order to carry out the examinations described in the previous pilot experiment and to determine the basal parameters of each animal. The monkeys are inoculated at time 0 and after 2, 6, 11, 15, 21, 28 and 32 weeks. At week 36 monkeys Ml-6 received the last boost with 16 μg of recombinant Tat protein in 200 μl of ISCOM (immunostimulant complex) and 300 μl of PBS. ISCOM is an adjuvant consisting of saponin A from bird feathers, cholesterol and phospholipids which increase the humoral and cell-mediated immune response (Ref 109, 90). Monos M7 and M8 were injected at the same time points with adjuvant only. At each vaccination point and at weeks 40, 44 and 50, 10 ml of blood is drawn from the animals to analyze the clinical and immunological parameters described in the previous pilot experiment. In addition, urine samples and vaginal swabs are collected to analyze the presence of secretory IgA specific for Tat. In order to evaluate the protective effect of Tat immunization against infection, vaccinated and control monkeys were challenged with the chimeric "simian / human immunodeficiency virus" (SHIV), strain 89.6P, which contains the HIV tat gene. -l which has previously grown and been titled in Macaca fascicularis (Ref. 128, 129, 69). After exposure, the animals are modified (every two weeks during the first month, every four weeks for the next three months and every 8 weeks until 6-12 months) for virological parameters such as plasma p27 antigenemia and plasma and cellular viral load . To confirm that the infection has occurred, antibodies against SIV are also analyzed by means of commercial equipment used for the detection of antibodies against HIV-2, which also recognizes antibodies against SIV (Elavia Ac-Ab-Ak II equipment, Diagnostic Pasteur, Paris, France). Till the date, the results of the second pilot experiments are as follows. No alterations of the chemophysical, hematological and behavioral parameters are observed. The monkeys show no sign of inflammation or neurovascularization at the site of "inoculation," a specific antibody response (IgM, IgG) is observed, and at week 15, the antibody titers against Tat (IgG) reached high levels, and vary from 1: 6400 to 1: 25600 (Figures 5-7) Antibodies fractionated essentially with the amino terminal region (amino acids 1-20) of Tat, with titers ranging from 1: 1600 to 1: 3200 (Figure 8) is shown at week 22.) In addition, antibodies directed against Tat amino acids 46-60, with titers ranging from 1: 100 to 1: 200, were also detected (Figure 8). to neutralize the activity of Tat is measured by determining the inhibition of viral rescue in HLM-1 cells incubated with serial amounts of exogenous Tat, as previously described in the first pilot experiment.The results of these experiments have shown that the immune plasma (diluted 1: 2) of monkeys Ml-6 at week 15 blogs viral replication induced by 30 ng / ml of exogenous Tat, determined by measuring the p24 antigen released within the culture medium. Conversely, the preimmune plasma of monkeys Ml-6 or the plasma of control monkeys (M7, M8) does not blog Tat activity (table 9). In addition, the immune plasma (diluted 1: 2) of monkeys Ml-6, extracted in week 21, blogged the replication of the virus induced by 60 ng / ml, 120 ng / ml, 240 ng / ml and 500 ng / ml of Exogenous Tat. In particular, this plasma determines a 10-fold reduction in virus replication induced by very high doses of extracellular Tat (240 ng / ml and 500 ng / ml) (table 9) TABLE 9 Neutralizing activity of immune plasma in the replication of virus induced by extracellular Tat3 Tat (30 ng / ml I + Preinmune M5 0 Tat (30 ng / ml) + Preinmune M6 0 Tat (30 ng / ml) + Immune Ml (week 15) 89.8 Tat 30 ng / ml + Immune M2 (week 15) 78.7 Tat 30 ng / ml + Immune M3 (week 15) 100 Tat 30 ng / ml + Immune M4 (week 15) 100 Tat 30 ng / ml + Immune M5 (week 15) 70.8 Tat 30 ng / ml + Immune M6 (week 15) 94 2 Tat 60 ng / ml + Preinmune Ml 0 Tat (60 ng / ml, + Preinmune M2 0 Tat (60 ng / ml) + Preinmune M3 0 Tat (60 ng / ml) + Preinmune M4 0 Tat (60 ng / ml) + Preinmune M5 0 Tat (60 ng / ml) + Preinmune M6 0 Tat (60 ng / ml) + Immune Ml (week 21) 96.3 Tat (60 ng / ml) + Immune M2 (week 21) 100 Tat (60 ng / ml) + Immune M3 (week 21) 100 Tat (60 ng / ml) + Immune M4 (week 21) 98.7 Tat (60 ng / ml) + Immune M5 (week 21) 99 Tat (60 ag / ml) + Immune M6 (week 21) 98.8 Tat (120 ng / ml i + Preimmune accumulation Ml-6 0 Tat 120 ng / ml) + Immune Ml (week 21) 59.2 Tat 120 ng / ml + Immune M2 (week 21) 90.4 Tat 120 ng / ml + Immune M3 (week 21) 96.8 Tat 120 ng / ml + Immune M4 (week 21) 98.3 Tat 120 ng / ml + Immune M5 (week 21) 100.
Tat 120 ng / ml + Immune M6 (week 21) 97.8 Tat 240 ng / ml + Preimmune accumulation Ml-6 0 Tat (240 ng / ml + Immune Ml (week 21) 26.1 Tat (240 ng / ml + Immune M2 (week 21) 49.4 Tat (240 ng / ml + Immune M3 (week 21) 70.3 Tat (240 ng / ml) + Immune M4 (week 21) 91.2 Tat (240 ng / ml) + Immune M5 (week 21) 94.5 Tat (240 ng / ml) + Immune M6 (week 21) 86 Tat (500 ng / ml) + Preimmune accumulation Ml-6 0 Tat (500 ng / ml) + Immune Ml (week 21) 32.7 The capacitance of p ast with Tat to neutralize Tat activity was determined in HLM-l cells, as described in the legend to table 4. The recombinant Ta t protein (30 ng / ml, 60 ng / ml, 120 ng / ml, 240 ng / ml and 500 ng / ml) is added alone or together with an equal volume of mono preimmune plasma or at week 15 or 21 (immune plasma). Ml-3 monkeys were vaccinated with 10 μg of Tat in 250 μl of autologous serum and 250 μl of RIBI; . M4 -6 monkeys were vaccinated with 10 μg of Tat in 250 μl of autologous serum and 250 μl of alumina; two control monkeys were injected with RIBI (250 μl and 250 μl of autologous serum) (M7) or with alumina (250 μl and 250 μl of autologous serum) (M8). The results are represented as described in the legend of Table 4. The ability of the monkeys plasma to neutralize extracellular Tat activity released by cells during acute infection is tested in CEM x 174 cells infected with the guimeric virus SHIV 89.6P. On day 7 after infection, replication of the virus is observed in 50% of control cells infected with SHIV and cultured with the preimmune plasma of Ml-6 monkeys. Conversely, no replication of virus was detected in infected cells that grew in the presence of the immune plasma of monkeys Ml-6 extracted at week 44 (Table 10).
TABLE 10 Immune plasma neutralizing activity in the transmission of virus infection3 SHIV + Immune M4 Neg SHIV + Immune M5 Neg SHIV + Immune M6 Neg CEM x 174 cells (3 x 10 * cells / 150 μl) were infected in 96-well plates infected with the SHIV 89 chimeric virus. 6P (5 x 10'5 TCIDS0 / cell) for 2 hours at 37 ° C in RPMI 1640 containing 10% FCS. The cells are washed twice with RPMI 1640 and resuspended in 150 μl of complete medium added with 5% preimmune plasma of mono or immune plasma (week 44) for animals vaccinated with 10 μg of recombinant Tat and RIBI (Ml -3) or alumina (M4 -6). The animal plasma is preheated at 56 ° C for 30 min and analyzed by ELISA to monitor antibody titers against Tat. Each serum is tested in duplicate. On days 3, 5 and 7 after infection, 120 μl of culture medium is collected and replaced with an equal volume of fresh medium containing 5% preimmune or immune plasma of monkeys Ml -6. The ability of plasma to neutralize extracellular Ta t, released during acute infection, and to control transmission of infection in vi tro was determined by detecting viral p27 Gag in the culture medium by ELISA (Coul ter International, Miami, FL ). The results, represented as p27 values (pg / ml), correspond to the mean value of two wells for each serum on day 7 after infection.
In addition, a proliferative response to Tat was observed from week 11 (table 11). TABLE 11 Proliferative response to Tata 4 μg / ml of PHA, 10 μg / ml of tetanus toxoid (TT) and 5 μg / ml of Tat, and are tested as described in the legend for table 5. Monkeys Ml -3 were inoculated with 10 μg of recombinant Tat protein in 250 μl of autologous serum and 250 μl of RIBI; monkeys M4-6 were inoculated with 10 μg of recombinant Tat in 250 μl of autologous serum and 250 μl of alumina; Two control monkeys were inoculated with RIBI (250 μl and 250 μl of autologous serum) (M7) and with alumina (250 μl and 250 μl of autologous serum) (M8). ND, not done. A strong response of cytotoxic T cells (CTL) is detected in a monkey vaccinated with the Tat and RIBI (Ml) protein and in two monkeys vaccinated with the Tat protein and alumina (M4 and M5), while a CTL response is observed more weak in the M6 monkey immunized with Tat and alumina (figure 9 and table 12) TABLE 12 Analysis of the CTL response The assay is performed as described in Table 6. Ml-3 monkeys are immunized with 10 μg of recombinant Tat in 250 μl of autologous serum and 250 μl of RIBI; monkeys M4 -6 are inoculated with 10 μg of re-combining Tat in 250 μl of autologous serum and 250 μl of alumina; two control monkeys were inoculated with RIBI (250 μl and 250 μl of autologous serum) (M7) and alumina (250 μl and 250 μl of autologous serum) (M8). ND, not done. At week 44, the presence of total antiviral activity (TAA) is determined. TAA is measured as the PBMC capacity of monkeys vaccinated with recombinant Tat proteins, cultured in the presence of autologous serum, to be resistant to infection by SHIV 89.6P (Table 13).
TABLE 13 Analysis of the presence of total antiviral activity (TAA) ' PBMC was co-administered at week 44 of vaccine monkeys with 10 μg of recombinant Tat protein and RIBI (Ml -3) or alumina (M4 -6) and of control monkeys incubated with RIBI (M7) or alumina (M8). PBMC are grown, purified by Ficoll gradient and seeded in triplicate at 5 x 10e / 200 μl per well in 48 well plates, in RPMI 1640 - containing 10% FCS and 5% autologous plasma previously heated at 56 ° C for 30 min, in the presence of 5 ng / ml of monoclonal antibody against CDS and 2 U / ml of IL-2, for 48-72 hours at 37 ° C. The cells were infected with serial dilutions of SHIV 89 chimeric virus. 6P (10-2, 10.3, 1Q-4, 10.5 TCIDS0 / cell) for 2 hours at 37 ° C, washed 3 times with PBSA-A and resuspended in 50% conditioned medium and 50% fresh medium at 5 x 10 5 cells / ml / well. On days 3, 7, 10, 14 and 17 after infection, aliquots of culture medium are collected and replaced with equal volumes of fresh medium. Viral replication in cell supernatants is determined by the ELISA test for p27 Gag (Coul ter International, Miami, FL). The results are shown as the minimum infectious dose of SHIV (TCIDS0 / cell) on days 7 and 17 after infection capable of infecting monkey lymphocytes. The results demonstrate the presence of soluble antiviral activity mediated by CD8 + T lymphocytes (CAF) (table 14). A total increase in CAF activity is observed in vaccinated monkeys, compared to the control animals.
TABLE 14 Analysis of the presence of soluble antiviral activity mediated by CD8 + T lymphocytes (CAF) a mediated by CD8 + T lymphocytes (CAF) of monkeys vaccinated with 10 μg of recombinant Tat protein and RIBI (Ml -3) or alumina (M4 -6), and of control monkeys incubated with RIBI (M7) or alumina (M8). Acute infection is tested in CEM x 174 cells infected with SHIV 89. 6P. The test is carried out as described in table 7 and the results refer to day 7 after infection. The presence of CAF is tested in the system of chronic infection in the Ul cell line (Ref. 47), which is a promonocitic human cell line chronically infected by HIV-1. Ul cells, seeded at 1 x 10 4 cells / 250 μl per well in 96-well plates, are incubated with PMA (10'BM) to induce the reactivation of HIV-1 infection, with or without different volumes (50 μl, 5 μl, 0.5 μl) of culture supernatants from CD8 + T lymphocytes derived from vaccinated monkeys and control. Three days after treatment with PMA, the presence of HIV-1 in the culture medium is determined by the RT assay or by ELISA for p24 Gag. The results are presented as% inhibition of HIV-1 replication in cells treated with CD8 + T cell supernatants compared to untreated cells. The results of inhibition of acute and chronic infection refer to cells treated with 5 μl of CD8 + supernatants. The production of cytokines (? LFN, IL-4, TNFa) and of the RANTES chemokine from PBMC of monkeys vaccinated with Tat and RIBI (Ml-3) or Tat and alumina (M4-6) and control monkeys M7 and M8 it was also determined (table 15). to H Ul or Ln s Ul TABLE 15 Analysis of cytokine and chemokine production Or co L? s H a Analysis of the production of cytokines and chemokines after 48 and 96 hours of culture (48/96) of PBMC of monkeys vaccinated with 10 μg of Tat and RIBI (Ml-3) or alumina (M4-6). Monkeys control (M7 and M8) were inoculated with RIBI or alumina adjuvants. PBMC, extracted at week 44 and purified by Ficoll gradient were seeded at 1 x 106 cells / ml in 24-well plates and grown in RPMI 1640 containing 10% FCS. The PBMC were not stimulated (control), to evaluate the spontaneous release of cytokines and chemokines, or stimulated with PHA (2 μg / ml), tetanus toxoid (TT, 5 μg / ml) or Tat (1 or 5 μg / ml). The aliquots of the culture supernatants were collected at 48 and 96 hours after the stimulation to determine the presence of cytokines and chemokines, by means of commercial ELISA equipment from BioSource International (Camarillo, CA, United States) par determined production of cytokines, and R &D Systems (Abdigdon, Oxon, United Kingdom) to assess the production of RANTES. The results are shown as pg / ml at 48 and 96 hours of culture (48/96), respectively. The cut-off values were (in pg / ml): IFN? , 31.2; IL-4, 3.12; TNFa, 15.6; RANTES, 6.25. (-) the values were lower than the corresponding cut-off values. Nd: not done. In addition, at week 5, five monkeys vaccinated with the recombinant protein (M2-6), showed a positive reaction in the skin test for Tat, with a strong reaction of delayed hypersensitivity (Table 16 and Figure 19). In monkeys 4 and 5, the skin test reaction was even higher in the following weeks (Table 16). TABLE 16 Tata skin test shock absorber alone, in tradérmica in a shaved area of the back of vaccinated monkeys. The control animals were not inoculated (ND, it was not done) at weeks 11, 15, 21, 28, 32, 36 and 44 after the first immunization. Ml-3 monkeys were vaccinated with 10 μg of recombinant Ta t protein in 250 μl of autologous serum and 259 μl of RIBI; the M4 -6 monkeys were vaccinated with 10 μg of recombinant Tat protein in 250 μl of autologous serum or 250 μl of alumina; two control monkeys were inoculated with RIBI (250 μl and 250 μl of autologous serum) (M7) or alumina (250 μl and 250 μl of autologous serum) (M8). The presence of an erythematous nodule after 48-72 hours is suggestive of a delayed hypersensitivity reaction (DTH): ++,? = 5 mm; +,? = 1 -4 mm; +/- eri theme without hardening; ? (1 mm) Post-exposure results indicate that 4/6 of the vaccinated monkeys were protected against ition with 10 MID50 of SHIV 89.6P, as shown by the results of the virological tests (Table 17). Gag p27 antigen was detected in plasma of monkeys Ml, M2, M4 and M6, no proviral DNA was found by PCR in lymphocytes of these monkeys and cytoviremia was negative, monkeys M3 and M5 were ited as shown by the presence of Gag p27 antigen in the plasma by detection of proviral DNA in the cells and by positive cytoviremia (Table 17) Both controls (M7 and M8) were ited, on the basis of the same virological assays.To control additionally the itive capacity of the Viral dose used for the exposure, another monkey not previously exposed (M13) was added to the control animals and was ited with 2.85 MID50 of SHIV 89.6P (corresponding to a dose of virus 3.5 times less than the dose used for the exposure of animals in the protocol). The M13 monkey was ited on the basis of all virological tests. To confirm that the animals were exposed to the virus, the presence of antibodies against SIV antigens, encoded by the chimeric SHIV 89.6P virus (Gag, Pol, RT, Nef), was analyzed as already described above in this Example. The presence of antibodies against SIV in monkeys that were negative for virological parameters (Ml, M2, M4 and M6) confirms that these animals were exposed to the virus and indicate that an abortive SHIV infection occurred in these monkeys. Monkeys that showed low titers of antibody against SIV were studied for in vitro production of specific antiviral IgG (IVAP) (Ref. 177, 178) according to the following method. PBMC (2 x 106 / well) was seeded in 24-well plates, and stimulated with P M (2 μg / ml, Sigma, St. Louis, United States). After 7 days of incubation (at 37 ° C in the presence of 5% C02 and 95% humidity), the culture supernatants were harvested to perform an assay for antibody production against HIV by a commercial ELISA kit for the detection of HIV-1 and HIV-2 antibodies (Abbott, HIV-1 / HIV-2 EIA Third Generation Plus). All the exposed monkeys were positive for the production of Env antibodies against HIV, since Env HIV-l is present in SHIV 89.6P. to t l H o l H O l TABLE 17 Analysis d? virological parameters Analysis of virological parameters after exposure of monkeys vaccinated with 10 μg of recombinant Tat protein and RIBI or Alumina (M4 -6). The control monkeys (M7 and M8) were inoculated with RIBI or alumina, respectively. The M13 monkey was an unexposed animal previously infected with 2.85 MID50 of SHIV 89. 6P. a Plasma antigenemia was assessed by the p27 Gag ELISA (Innogenetics, Belgium) and expressed as p27 values (pg / ml). Neg, the value was lower than the corresponding cut-off value (18 pg / ml). DNA was purified by whole blood using the QIAamp blood equipment (Angiogen Gmbh and Qiagen Inc., Hilden, Germany). The DNA quality was monitored by PCR amplification of the ß-globin gene, as previously described (Ref. 141). The presence of proviral DNA was analyzed by semi-stationary PCR amplification of SIV ga. PCR was performed on 1 μg of cellular DNA using primers SG1096Ngag (corresponding to nucleotides 1096-1119 in the genome SIVmac251: 5 • TTAGGCTACGACCCGGCGGAAAGA3) and SG1592CgagD (mapped in numbers 1569-1592 of the SIVma genome c251: 'ATAGGGGGTGCAGCCTTCTGACAG3') which amplify a 496 base pair of the cracr SHIV gene, as described (Ref. 153). To quantify the number of proviral DNA copies, in each experiment a standard curve was prepared using the plasmid pCMRII-? Gag (containing a deletion of 100 base pairs in the acr gene of SIVmac251) as a DNA template and the primers described before they amplify a DNA fragment of 396 base pairs. The PCR products were analyzed by electrophoresis and quantified by densitometrical analysis (Ul tr sean LX Enhancer Laser, LKB, Bromma, Sweden). The relationship between DO values and the number of molecules of the? Gag plasmid was correlated by means of linear regression analysis (Statgraphics, Manugistics, Inc. Cambridge, MA). OD values were linear up to 1000 molecules (correlation coefficient = 0.954 + 0. 026). The number of proviral DNA copies SHIV / μg of cellular DNA was determined by interpolating the OD values of each sample with the standard curve. The sensitivity of the assay was 1 copy of provirus / μg of DNA. c Toviramia was determined in cocultivation trials. For this purpose, 1 x 104 CEM x 174 cells were cultured in the presence of serial dilutions of PBMC lacking CD-8 under study. (a total of 12 dilutions, from 1 x 106 to 3. 9 x 103 cells per well) in 96-well plates. On days 3, 7 and 10 after infection, 150 μl was removed to test the presence of p27 Gag by ELISA (Innogenetics, Belgium) and replaced with an equal volume of fresh medium. The results are analyzed using the formula of Reed and Muench to determine the number of PBMCs productively infected per million total cells. d The presence of antibodies against SHIV was determined in serial dilutions of animal plasma tested in duplicate using the Elavia Ac-Ab-Ak II equipment (Diagnostic Pasteur, Paris, France), according to the manufacturer's instructions. The highest dilution at which the plasma values were higher than the cutoff value is shown. The isolation of the virus, instead of the toviremia, was performed for the monkey M13. To this end, PBMC (3 x 10e) of monkeys infected with different doses of SHIV 89 were cultured. 6P, purified by Ficoll, cultured with CEM x 174 cells. (1 x 106) in 1 ml of medium containing 10% FCS. After 24 hours, the cells were diluted to 1 x 106 / ml and cultured for 3 days. Then 2 ml of medium was collected and the cells were re-seeded at 3 x 10 5 / ml in 7 ml. He disposed of excess cell. This procedure was repeated twice a week, for 4 weeks. The presence of virus was determined by p27 Gag ELISA (Innogenetics, Belgium) and then by RT assay. Virus isolation was considered positive (+) when both trials (p27 and RT) were positive in three sequential samples. Conversely, the isolation of the virus was considered negative (-) • A qualitative DNA-PCR test was performed for the M13 monkey. The virological data are superimposed on the absolute numbers of CD4 lymphocytes that are markedly reduced in infected monkeys (M3, M5, M7, M8) and high and stable in animals negative to the virus (Ml, M2, M4, M6) as shown in Table 18 TABLE 18 FACS Analysis of CD4 + and CD8 + S Lymphocytes H H a FACS analysis of CD4 + and CD8 + lymphocytes from monkeys vaccinated with 10 μg of recombinant Tat protein and RIBI (Ml -3) or Alumina (M4 - 6). The control monkeys (M7 and M8) were inoculated only with RIBI adjuvant or alumina, respectively. Monkey M13 is a previously unexposed animal infected with 2.85 MID50 of SHIV 89. 6P. The analysis was performed by a fluorescence activated cell sorter (FACS) as described (Ref. 137), using labeled monoclonal antibodies (anti-CD4-FITC, BioSource, anti-CD8 -PerCp, Becton-Dickinson). ND, not done. The results before exposure indicate that Tat as the immunogen, as well as RIBI and alumina as the adjuvants (or ISCOM that is used as an adjuvant in the last booster), are well tolerated by animals and are not toxic, confirming the results of safety and tolerability of immunization with Tat obtained in the first pilot experiment. However, these data confirm the observations of the first pilot experiment, which supports additional evidence of the fact that the recombinant Tat protein induces a strong humoral and cellular response, specific for Tat with antiviral effects in vitro and in vivo. Post-exposure results (4/6 protected monkeys) confirm the hope of in vitro results and indicate that the Tat vaccine induces protection against infection and therefore against the disease. The monitoring of the two vaccinated and infected monkeys will clarify the effects of vaccination on the progress of the disease.
Example 5. Inoculation in Macaca fascicularis of a DNA vaccine against Tat; safety analysis, tolerability, specific immune response and protection effectiveness against viral exposure It is proposed that the direct inoculation of plasmid pCV-Tat DNA containing the tat gene cDNA, and the pCVO plasmid as control DNA. The plasmid DNAs to be administered to the animals are amplified in E. coli (strain DH5) according to standard procedures (Ref. 110) and with protocols established by the "European Agency for the evaluation of medical products"; Unit " ("Technical Report Series No. 17, January 1997), purified by two gradients of CsCl and dialyzed for approximately 48-72 hours against 100 volumes of PB.S.
The DNA is then verified by digestion with restriction enzymes. The functionality of the plasmid DNA is controlled by transfection of 5-10 μg of DNA using calcium phosphate techniques (Ref. 110) in H3T1 cells (1 x 106), which contain an integrated copy of the HIV-1 LTR-indicator plasmid. CAT, and 48 hours later, by the CAT activity analysis (Ref. 55). Tolerability, safety, ability to induce a specific immune response (both humoral and cellular) and the efficacy of protection against post-immunization virus exposure with plasmid DNA pCV-Tat were evaluated in cynomorph monkeys (Macaca fascicularis) . In a first pilot experiment, three monkeys are immunized according to the following protocol: monkey Ml is inoculated with 200 μg of pCV-Tat in 300 μl of PBS per day i.d. in two loin sites, near the axillary lymph nodes (150 μl / site); the M2 monkey is inoculated with 500 μg of pCV-Tat in 500 μl of PBS via i.m. in 2 loin sites (250 μl / site). On days 1 or 5 before inoculation i.m., 250 μl of physiological solution, which contains 0.5% bupivacaine and 0.1% methylparaben and are injected into the two previously marked sites, where the plasmid DNA will be inoculated. This is done in order to increase the uptake and expression of DNA in the muscle (Ref. 37, 45). The M3 monkey was not inoculated and was used as a control animal. However, starting at week 10, this monkey was inoculated with 6 μg (5 + 1 μg) i.d. of Tat as a control for skin tests. 10 ml of blood was extracted from all the monkeys at 42 and 35 days preceding the first inoculation for analysis of the basal parameters. The monkeys were inoculated at time 0 and after 5, 10, 15, 22, 27, 32 and 37 weeks. Finally, at week 42, the animals received the last boost with recombinant Tat protein (16 μg) in 200 μl of ISCOM and 300 μl of PBS. The animals were observed daily to determine clinical parameters, as described in Example 4. In addition, 10 ml of blood was drawn on the same day of inoculation, as described in Example 4. The protective effect of the vaccine was determined after of the exposure of the monkeys with 10 MID50 of SHIV89.6P, which was injected intravenously at week 65. The post-exposure monitoring, which is still carried out, is performed as described in Example 4. The results of this experiment are as follows. No alterations of clinical, haematological and behavioral parameters were observed in two vaccinated monkeys and in the control monkey. No inflammatory signs or neovascularization were observed at the injection site. These results indicate that the pCV-Tat DNA is well tolerated by animals and is not toxic at the doses and days of inoculation used in the experiment. Monkey Ml, vaccinated with 200 μg of DNA via i.d., developed IgG antibodies specific for Tat from week 32 (figure 11). Antibody titers (from week 32 to week 58) varied between 1: 100 and 1: 800 (Figure 12). At week 37, the epitope mapping analysis (performed as described in the legend for Figure 4) shows that these antibodies were directed against specific regions of Tat, mapping at amino acids 1-20, amino acids 46-60 and amino acids 65-80 with titers of 1: 200, 1 : 100 and 1:50, respectively (data not shown). In monkey M2, vaccinated with 500 μg of DNA by the i.m. route, antibodies against Tat were seldom detected (with a titer of 1:50, not shown) for the entire study period. The results are shown in Figure 11. The plasma capacity of monkey Ml, vaccinated with 200 μg of DNA by i.d. to update the Tat activity, by assaying the inhibition of the rescue of viral replication in HLMl cells incubated with exogenous Tat proteins, as described in Example 4. This assay shows that the monkey Ml plasma, diluted 1: 2 and obtained at week 37, reduces viral replication induced by 30 mg / ml of endogenous Tat. Conversely, plasma from the same monkey obtained at time 0 (pre-immune) does not block extracellular Tat (Table 19).
TABLE 19 Plasma neutralizing activity in the rescue of viral infection induced by extracellular Tata Samples Inhibition Tat + preimmune Ml 0 Tat + Immune Ml 51 a The ability of antibodies against Tat to neutralize the activity of Tat in HLMl cells was determined when adding ng / ml of recombinant Tat protein, previously incubated with an equal volume of plasma obtained at time 0 (pre-immune) or at week 37 (immune) for monkey Ml, vaccinated with 200 μg of plasmidic DNA pCV-Tat via i. d. The assay was performed and the results are expressed as described in Table 4. The results shown in Table 20 demonstrate the presence of a proliferative response to Tat at week 42 in the monkey Ml immunized with 200 μg of DNA by id, while in the M2 monkey, this type of cellular response was not detected.
TABLE 20 Proliferative response to Tata a PBMC are isolated, stimulated with PHA (4 μg / ml), tetanus toxoid (TT) and Tat (1 or 5 μg / ml) and tested as described in Table 5. Monkeys were vaccinated with 200 μg. (Ml) of pCV-Ta t by way i. d. or with 500 μg (M2) of pCV-Ta t via i .m. The monkey (M3) was not vaccinated but was inoculated from week 10 with 6 μg (5 + 1 μg) i. d. of Tat as a contine for skin tests. ND: not done.
Cytotoxic activity against Tat (CTL) was detected in monkey Ml at week 42 and 48, and monkey M2 at week 48. In addition, a positive CTL response was observed at week 48 in monkey M3, which was inoculated from week 10 with 6 μg of Tat as a control for skin tests (Table 21).
TABLE 21 Specific cytotoxic activity analysis for Tat (CTL) 1 a The assay is carried out as described in Table 6. The monkeys were vaccinated with 200 μg (Ml) of pCV-Tat via i.d. or with 500 μg (M2) of pCV-Tat via i.m. The monkey (M3) was not vaccinated but was inoculated from week 10 with 6 μg (5 + 1 μg) of i.d. of Tat as a control for skin tests. ND: not done.
The results shown in Table 22 indicate that at week 52 the presence of total antiviral activity (TAA) in the two monkeys vaccinated with 200 and 500 μg of DNA.
TABLE 22 Analysis of total antiviral activity (TAA) ' aThe assay is performed as described in Table 13. The monkeys were inoculated with 200 μg (Ml) of pCV-Ta t via i. d. , or with 500 μg of pCV-Tat via i .m. Monkey (M3) was not inoculated, but from week 10 it received 6 μg (5 + 1 μg) i. d. of Tt as a control for skin tests. PBMC were harvested at week 52 from the primary immunization and infected with SHIV 89. 6P (10 ' 2, 10'4, 10'6, 10'8 TCID50 / cell). The results are represented as the minimum infective dose of SHIV (TCID50 / cell) that is still capable of infecting cells.
The results shown in Table 23 indicate the presence of soluble antiviral activity (CAF) mediated by CD8 + T lymphocytes, at week 22 and 27, in both vaccinated monkeys. This activity is lower in the control monkey.
TABLE 23 Analysis of the soluble antiviral activity mediated by CD8 + cells (CAF) a a Analysis of the presence of soluble antiviral activity produced by CD8 + T lymphocytes (CAF) derived from monkeys inoculated with 200 μg (Ml) and 500 μg (M2) of pCV-Tat and monkey M3. Antiviral activity was tested in acute and chronic infection in CEM x 174 cells infested with SHIV 89. 6P and in OM-10-1 cells chronically infected with HIV-1, as described in Table 7. The results were represent as percentage (%) of inhibition of viral replication in cells treated with supernatant from CD8 + T lymphocytes compared to untreated cells. The results of the acute and chronic infection shown in the Table refer to samples treated with 5 μl of CD8 + culture supernatant. ND, not done. The results shown in Table 24 demonstrate that the monkey Ml, inoculated with 200 μg of DNA by the i.d. has a positive skin test for Tat at week 22.
TABLE 24 Skin test for Tata "Tat id (1 and 5 μg) is inoculated in 150 μl of PBS-0.1% BSA or buffer alone (control) in a previously trichotomized area of the upper loin of the vaccinated animals and in the control monkey (control for the specificity of the response) at weeks 10, 15, 22, 27, 32, 37, 42, 48, 52 and 58 from the primary immunization.The monkey Ml was inoculated id with 200 μg of plasmid pCV-Tat DNA, while that the macaque M2 received 500 μg of the same plasmid, im The monkey M3 (control) was not vaccinated but from week 10 it received 6 μg (5 + 1 μg) id of Tat as a control for the skin tests. an erythematous nodule, 48 to 72 hours later, indicates the presence of delayed-type hypersensitivity (DT). ++, e = 5 mm, +? 1-4 mm; ±, non-hardening issue; -, < 1 mm.
These results indicate that the plasmid pCVTat (pCVTat-DNA) is well tolerated and safe intradermally and intramuscularly at the doses given. In addition, these results demonstrate that immunization with pCVTat-DNA induces a humoral immune response (although less than that induced by immunization with the recombinant Tat protein) as cellular against Tat, with antiviral effects. Regarding the protective efficacy after exposure (performed at week 65 from the initial immunization), the virological data, which include measurements of antigenemia and cytoviremia and the determination of the number of proviral DNA copies (DNA-PCR) in the PBMC, indicate that the monkey M2, immunized im with Tat-DNA, it is protected by exposing it with 10 MID50 of SHIV-89.6P, while the macaque Ml, immunized i.d. with a lower dose of Tat-DNA (200 μg) becomes infected, which suggests that, with respect to immunization with DNA, the i.m. It is more effective than inoculation i.d. The M3 control monkey is also resistant to infection. However, as previously described, this monkey, unlike the controls of the other experimental protocols, received repeated skin tests for Tat in order to control the test specificity (Table 24), and antibodies against Tat were detected, although in low titers (1: 100) from week 32 of the start of immunization (data not shown). In addition, the proliferative response to Tat in this monkey shows a weak and sporadic reactivity to the antigen (Table 20). Finally, monkey M3 showed the presence of specific CTL against Tat (Table 21). Although preliminary, these data indicate that the repeated injection i.d. 6 μg of Tat can result in animal immunization and protection from exposure. Therefore, monkey M3 will be considered vaccinated i.d. with the Tat protein and it will be studied as such.
H Ul or Ul O l TABLE 25 Analysis of virological parameters ? or Monkey Ml has been immunized i. d. with 200 μg of pCVTat, the monkey M2 with 500 μg of pCVTat, i .m. The maque M3 was injected several times with 6 μg of Tat protein, intradermally, in order to control the specificity of the cutaneous test. Therefore, from the time of exposure the monkey M3 is considered as a vaccinated monkey. The virological parameters were evaluated as described in the legend for Table 17. The FACS evaluation of the percentage and absolute number of CD4 and CD8 lymphocytes confirmed the virological data, with a clear (approximately 4-fold) reduction of CD4 lymphocytes in the infected monkey, in advance in the first post-exposure analysis (day 30) and was subsequently confirmed (day 60) (Table 26). t Ul t o H H Ul O Ul TABLE 26 Analysis by FACS of the CD4 and CDS sub-juncts tO t to Ul or H Ul? i The analysis was performed as indicated in the legend to Table 18. Monkey Ml has been immunized i.d. with 200 μg of plasmid DNA pCVTat, the monkey M2 with 500 μg of pCVTat-DNA, i.m. The macaque M3 was vaccinated with 6 μg of Tat protein intradermally.
Based on these results, a second experiment was designed in which the effects of immunization with pCVTat-DNA were evaluated in 3 monkeys (M9-M11) compared to the control monkey (M12) that received pCVO-DNA. All the animals were inoculated i.m. in two sites on the spine with a total of 1 mg of pCVTat (M9-M11) or pCVO (M12). Either 1 or 5 days before the vaccination, 250 μl of saline containing 0.5% bupivacaine and 0.1% methylparaben were inoculated into the two marked sites, in which successively the plasmid was injected. The macaques were vaccinated at time 0 and at weeks 6, 11, 15, 21, 28, and 32. A final booster was performed at week 36 with the recombinant Tat protein (16 μg) resuspended in 200 μl of ISCOM and 300 μl of PBS. The animals were monitored every day to determine clinical parameters, as described in Example 4. In addition, 10 ml of blood was removed 9 days before the primary immunization and in each immunization, as described in Example 4. With the In order to evaluate the protective effects of vaccination, the monkeys were exposed at week 50 from the start of immunization by intravenous (iv) injection of 10 MID50 of SHIV-89.6P. The post-exposure follow-up is still being carried out and is carried out as described in Example 4. The results of this experiment are as follows. No changes were observed in terms of behavior, clinical parameters and blood chemistry in both vaccinated and control animals. No signs of inflammation or vascular neoformations were detected at the injection sites. These results confirm that 1 mg of the i.m. injected pCVTat DNA plasmid is well tolerated and non-toxic. IgG against Tat is detected from week 15 (Figure 13), with titers ranging from 1:50 to 1: 100 (data not shown). In addition, a proliferative response to Tat is detected as early as two weeks in a monkey (Mil) (Table 27).
TABLE 27 Proliferative response to Tata a PBMC is isolated, it is stimulated with 4 μg / ml of PHA, tetanus toxoid (TT, 10 μg / ml) or Tat (1 and 5 μg / ml) and tests are carried out as described in Table 5. It is injected to the monkeys i .m. with 1 mg of either pCVTat (M9-M11) or pCVO (M12, control). ND, not determined.
CTL was detected against Tat in week 32 after immunization (Table 28).
TABLE 28 Analysis of cytotoxic activity against Tat (CTL) ' aThe assay is performed as described in Table 6. The macaques i .m were injected. with 1 mg of either pCVTat (M9-M11) or pCVO (M12, control).
The PBMC obtained from the Mil monkey at week 44 are resistant to infection in vitro with serial dilutions of the chimeric SHIV-89.6P virus by a previously described assay that detects the presence of total antiviral activity (TAA). In fact, TAA is evaluated as the capacity of PBMCs of monkeys vaccinated with pCVTat DNA, which grow in the presence of autologous serum, to resist infection with serial dilutions of virus (Table 29).
TABLE 29 Analysis of total antiviral activity (TAA) aThe test is performed as described in Table 13. The i.m. macaques are injected. with 1 mg of either pCVTat (M9-Mll) or pCVO (M12, control). PBMC are extracted at week 44 from the first immunization and infected in vitro with 10'2, 10-3, 10-4, 10-5 TCID50 of SHIV-89.6P. The results are expressed as the minimum infectious dose of SHIV (TCID5Q / cell) yet capable of infecting the cells. * There were no cultures that were infected at the highest SHIV concentration used in the trial (10 '2 TCIDS0 / cell). ** Crops become negative on day 17 after infection.
The results shown in Table 30 demonstrate the presence of soluble antiviral activity (CAF) mediated by CD8 + T lymphocytes in vaccinated monkeys and in the control monkey (M12) injected with empty vector (pCVO).
TABLE 30 Analysis of soluble antiviral activity (CAF) mediated by CD8 + T lymphocytes (CAF) a a Analysis of the presence of soluble antiviral activity mediated by CD8 + T lymphocytes (CAF). PBMC were obtained from three monkeys (M9-M11) injected with 1 mg of pCVTat and of the control monkey (M12) inoculated with 1 mg of pCVO. The acute infection test is carried out in CEMxl 74 cells infected with SHIV-89. 6P, as described in Table 14. The chronic infection assay is carried out on Ul cells chronically infected with HIV-1, as described in Table 14. The results are expressed as the percentage (%) of inhibition of viral replication in cultured cells in the presence or absence (control) of 5 μl of CD8 + T cell supernatants.
The production of cytokines (? IFN, IL-4, TNFa) and RANTES chemokine at week 44 in PMBC is evaluated from monkeys both vaccinated and control (Table 31).
H t t l s i o ui TABLE 31 Analysis of Cytokine and RANTES3 Production aThe test is performed as described in Table 15. The i.m. macaques are injected. with 1 mg of either pCVTat (M9-M11) or pCVO (M12, control). PBMCs are harvested at week 44 after the first immunization. The results are shown as pg / ml of cytokines and RANTES detected at 48 and 96 hours (48/96) respectively. (-), the values were below the cutoff value. The cut-off values (pg / ml) were:? LFN: 31.2; IL-4: 3.12; TNF-a: 15.6; RANTES: 62.5. ND: not done.
The results show the presence of a weak reactivity to the skin tests with Tat in a monkey (M9) in week 11 (Table 32). TABLE 32 Tata skin test aSe inoculate i. d. Tat (1 and 5 μg) in 150 μl of PBS-A, BSA 0. 1% or the buffer only (control) in a previously trichotomized area of the upper loin of vaccinated animals, but not in control monkeys, in weeks 11 , 15, 21, 28, 32, 36 and 44 from the initial immunization. The macaques were injected i .m. with 1 mg of either pCVTat (M9-11) or pCVO (M12, control). The appearance, at 48 to 72 hours later, of an eritro-nodule nodule indicates the presence of delayed-type hypersensitivity (DTH): + +, f > _ 5m; + f 1 -4 mm; ±, eri subject without hardening; -, f < 1 mm Post-exposure results indicate that all vaccinated animals were protected from infection with 10 MID50 of SHIV-89.6P, as indicated by virological tests (plasma antigenemia, determination of proviral DNA copy number, cytoviremia) and all they were negative (table 33). In addition, the presence of antibodies against SIV in the Mil monkey indicates the exposure of the virus or an abortive infection. On the contrary, it was not detected in the remaining monkeys, therefore it was decided to carry out the in vitro antibody production assay (IVAP) as well as the lymphoproliferative response to SIV antigens. These trials are underway and preliminary data indicate that there is presence of Env antibodies against HIV in all monkeys inoculated with DNA. The macaques will be inoculated with a higher dose of the virus, since even the M12 control animal was resistant to infection. This monkey has been vaccinated with the empty vector pCVO. Recent data from the literature have shown the adjuvant role played by certain DNA sequences that are much more frequent in bacteria than in eukaryotic cells, and this, similarly to LPS and mañosa, represents a strong stimulus for natural immunity (Ref 179). Therefore, it is conceivable that the protection observed in the M12 monkey may be due to the induction of antiviral immunity not specified by these bacterial sequences, such is the production of INFa, INFß, IL-12 and IL-18, which are known to exert immunomodulatory and antiviral functions. This is strongly suggested by the presence in this macaque of antiviral activities TAA (table 29) and CAF (table 30) in the absence of specific humoral and cellular immunity against Tat. In fact, these assays also measure specific antiviral activities without antigen. The M13 monkey that has not been previously exposed, inoculated with a virus dose 3.5 times lower compared to that injected in the M12 macaque, was infected. These results confirm that an exposure dose of 10 MID50 with which the M12 monkey was inoculated is infectious (Table 33). Based on this result, the inventor plans to use the pCVO vector or part thereof as an adjuvant.
TABLE 33 Analysis of virological parameters a, b.c, dThe assays were performed as described in table 17. The macacoa were injected i.m. with 1 mg of either pCVTat (M ^ Mll) or pCVO (M12, control). The M13 monkey is a previously unexposed animal, infected with 2.85 MIDS0 of SHIV89.6P. eEl Viral isolation was performed instead of cytoviremia and was positive. fDNA PCR is not quantitative and was positive.
In FACS analysis of the CD4 and CD8 subsets (table 34) confirms the virological data. In fact, a significant decrease in the percentage and absolute number of CD4 lymphocytes was observed at 15 and 60 days after exposure only for the monkey previously not exposed M13, which was infected as indicated by the positive result of plasma antigenemia, Proviral DNA and virus isolation (table 33).
TABLE 4 FACS Analysis of CD4 and CD8 Lymphocytes 00 H t t H l O Ul Ul or U aThe test was performed as described in table 18. The i.m. macaques were injected. with 1 mg of either pCVTat (M9-M11) or pCVO (M12, control). The M13 monkey is a previously unexposed animal, infected with 2.85 MIDS0 of SHIV89.6P. "3 These results demonstrate that vaccination with the pCVTat plasmid is well tolerated and non-toxic, and confirms the safety and tolerability of DNA vaccination, obtained in this first pilot study. In addition, these data provide evidence that the pCVTat-DNA plasmid induces a specific humoral immune response (albeit weaker than that induced by the Tat protein) and cellular, with antiviral effects, part of which may be due to DNA sequences individuals present in the pCVO vector that can function as adjuvants. Immunization protocols that will include combinations of DNA encoding other HIV-1 and cytokine genes described in Example 3 will be evaluated. In these experiments, HIV containing the genes for HIV tat, rev and nef will be used (Ref. 146, 85, 142, 65, 94, 129). The pCVO and pCVTat plasmids will be inoculated into the animals using other delivery systems that can enhance the immunization effectiveness, such as liposomes, nanoparticles, erythrocytes, gene cannon supply or Tat DNA that will be delivered through the use of vectors of herpes, as described in prophetic examples 9 and 10.
Example 6. Therapeutic vaccine A vaccination protocol was carried out, based on Tat protein and Tat DNA to evaluate the safety and toxicity of the Tat vaccine in already infected individuals. The experiment was performed in monkeys infected with increasingly smaller doses of SHIV89.6P, and with immunodeficiency disease (AIDS). The viral concentrate used for the infection was obtained from the spleen and lymph nodes of an infected baboon monkey 14 days before. The lymphocytes, purified by mechanical separation, were divided into two aliquots (1.5 x 106 cells / ml each). To an aliquot, CD8 + T cells were suppressed by the use of immunomagnetic spheres (Dynal, Norway). Both cultures were stimulated with PHA (1 μg / ml) for 3 days and seeded at a concentration of 1 × 10 6 cells / ml in the presence of 50 U / ml of IL-2. Viral replication was detected by the presence of reverse transcriptase (RT) in the culture medium harvested after 3 days. Prior to the test, the supernatant was rinsed and ultracentrifuged at 100,000 rpm for 11 minutes at + 4 ° C (Beckman TL-100 ultracentrifuge) and the pellet was smooth. 30 μl of the suspension (TRIS 1M HCl, pH 8, MgCl 2, 0.5M, 1M KCl, 1 mg / ml Poly A, 100 μg / ml oligo-dT 12-18, DTT 0.02 were added to the reaction mixture). M, 1, 23 [Methyl 1-methyl-1-methylthymidine HCl-1) and incubated at 37 ° C for 60 minutes. The reaction is stopped by adding 500 μl of 0.1 M Na pyrophosphate, pH 5, and 600 μl of 20% trichloroacetic acid (TCA), and the sample is placed by spot on a 0.45 m filter (Millipore) and then read with a ß counter after the addition of 5 ml of scintillation mixture (Filter Count, Packard). The culture medium contains more than 20,000 cpm, centrifuged and supplemented with human serum 10% AB. The virus is concentrated by ultracentrifugation at 30,000 rpm (90 minutes at 4 ° C), resuspended in RPMI 1640 containing 10% human serum (group AB) and then stored in small aliquots in liquid nitrogen. The viral concentrate is titrated in vi tro in human CEMxl74 and C8166 cell lines (3 x 103 TCID50 / cell) and also in vivo in baboon monkeys (3.17 x 105-69 MID50 / ml). A first pilot experiment was performed in 7 infected monkeys i.v. with plasmid SHIV89.6P prepared as described above. Each monkey received 1 ml of SHIV diluted in buffered saline solution supplemented with 2% human serum (AB, Rh-), according to the following protocol. A monkey (IM1) was inoculated with 1: 500 of a viral dilution; 2 monkeys (IM2, IM3) received the 1: 5,000 dilution; two monkeys (IM4, IM5) were inoculated with 1: 50,000; the IM6 monkey received the 1: 500,000 dilution; the last monkey (IM7) received a 1: 5,000,000 dilution. Each monkey had blood drawn on the next day before infection with SHIV for the determination of baseline parameters. The serum and plasma samples were frozen at -20 ° C or -80 ° C and then used to resuspend the protein inoculum. At time 0, all monkeys were inoculated with SHIV89.6P. The monkeys were checked daily. In addition, on day 0 and 2 and 4 weeks after blood was drawn and 10 ml of blood were used for hematochemical determinations (chemical-clinical analysis, electrolytes, white blood cells and platelet count, hemoglobin) and virological analysis and immunological (ie, determination of plasma p27 Ag and viral load in plasma and cells). In week 4 after infection, 6 monkeys were infected (IM1-6). The IM7 monkey, which received the lowest viral dilution (1: 5,000,000), was negative SHIV (Table 35).
TABLE 35 Detection of the presence of SHIV89.6P in monkeys infected with serial viral dilutions a Isolation of the virns and bplasma p27 Ag (pg / ml) were carried out as described in the legend for table 17. The monkeys were inoculated i. v. with serial dilutions of virus concentrate, as described in the text.
After 7 weeks from infection, all animals showed severe immunodeficiency symptoms and were vaccinated with both the Tat protein and the pCVTat plasmid DNA according to the following protocol. Monkeys IM1, IM3, IM5 and IM6 received the Tat protein (20 μg) dissolved in 250 μl of PBS-A supplemented with 0.1% BSA and 20% autologous plasma, and then added to 250 μl of alumina adjuvant. The protein inoculum was carried out subcutaneously at a single site on the upper loin of the monkey, while the pCVTat plasmid (1 mg) was resuspended in 1 ml of PBS-A, and i.m. in a different place on the spine. The IM2 and IM4 monkeys (controls) were injected with 250 μl of alumina and 250 μl of PBS-A, 0.1% BSA and 20% autologous plasma, s.c. at a site on the upper spine and with pCV-0 (1 mg) resuspended in 1 ml of PBS-A, i.m., at a site on the upper loin different from the previous one. The IM7 uninfected monkey was not vaccinated. The vaccination protocol consisted of a time 0, corresponding to 7 weeks after infection with SHIV and at 1, 4, 5, 10, 11, 13, 14, 17, and 18 weeks later. To evaluate the effects of this vaccination on the progress of the disease, each macaque was checked daily to determine the presence or signs of disease and at time 0 and after 3, 8, 12, 16 and 21 weeks, were extracted 10 ml of blood for laboratory tests (chemical-clinical analysis, electrolytes, white blood cells and platelet count, hemoglobin) for the evaluation of immunological status (presence of specific immunoglobulins, measurement of Thl and Th2, production of cytokines and chemokines), for characterization of lymphocytes by FACS analysis (CD4, CD8, CD28, CD40, CD86, CD20, CD2, CD26, and CD20), and finally for evaluation of virological parameters (detection of proviral DNA by quantitative semicrystallized PCR, plasma viral load by RT) Competitive PCR, plasmatic p27 Gag antigen by ELISA and presence of antibodies against SHIV Ab, as previously described). Other reinforcements were made based on the immunological, virological and clinical results. After the last inoculum, the monitoring was performed every month and at the time of appearance of clinical changes. At each point in time, samples of PBMC, serum, plasma and urine were frozen for future tests, as previously described. The results already available from this experiment, obtained from week 8 after immunization are those described. In both vaccinated and control asymptomatic monkeys, there are no signs of inflammation or neo-angiogenesis at the inoculation sites nor are there general symptoms of disease. There is no evidence of changes in clinical status in asymptomatic monkeys. In addition, viral replication activation is not detected. Taken together, these results indicate the absence of toxicity or increased viral replication in monkeys vaccinated with biologically active Tat protein or DNA (table 36).
TABLE 36 Analysis of virological parameters The tests were carried out as described in Table 17. Monkeys IM1, IM3, IM5 and IM6 were injected with 20 μg of Tat protein and adjuvant of alumina s.c. and with 1 mg of pCVTat i.m. The IM2 and IM4 monkeys (infected controls) were injected with s.c. alumina adjuvant. and with 1 mg pCVO i.m. IM7 is a monkey not previously exposed, not infected. Analysis by FACS indicates that no changes were observed in CD4 + and CD8 + T lymphocytes after vaccination (Table 37).
H H Ul O Ul O Ul TABLE 37 FACS analysis of CD4 + and CD8 + lymphocytes Ul 00 . In analysis by FACS, it was performed as described in the legend of Table 18. Monkeys IM1, IM3, IM5 and IM6 were injected with 20 μg of Tat protein and adjuvant of Ul alumina s.c., and with 1 mg of pCVTat i.m. The monkeys IM2 and IM4 (infected controls) were? injected with alumina adjuvant s.c. and 1 mg pCVO i.m. IM7 is a monkey not previously exposed, not infected. 10 These data confirm that both the Tat protein and the plasmid pCVTat, at the doses and days of inoculation used, were well tolerated and without any toxic effect in the vaccinated monkeys and, in addition, did not increase the viral replication or the decline of CD4 T cells in infected animals.
Example 7. Co-stimulation of purified CD4 + lymphocytes from monkeys infected with SIV, with spheres coated with anti-CD3 / 28 which results in a logarithmic expansion of the number of cells without significant viral replication or transmission Peripheral blood mononuclear cells were deleted from the CD8 + cell population by the use of immunomagnetic spheres against CD8 (Dynal, Oslo; Dynabeads M-450 CD8). The degree of purification was valued by FACS analysis and it was considered acceptable if it was above 95%. The cells lacking CD8 (called CDd'PBMC) were grown in the presence of 2 μg / ml of PHA and 40 U / ml of IL-2 or in immunomagnetic spheres previously coated with the two monoclonal antibodies against CD3 (Clone FN18, BioSource ) and CD28 antigens (Clone 9.3) (anti-CD3 / 28 spheres). To improve the binding of the anti-CD3 / 28 spheres with the target cells, the incubation was performed on a rotating positioning wheel. Then, the bound cells (designated CD28-CD3 + CD28 +) were selected with a magnet and plated in culture. Three times a week, the cell concentrations were adjusted to the initial level and IL-2 was added as indicated; In addition, regarding cells stimulated with anti-CD3 / 28 spheres, preliminary results suggest that the continuous stimulation regime coupled with a constant control in the sphere: cell ratio, adjusted at each time point, is highly effective in the induction of the proliferative response. Our previous studies have shown that, in the absence of exogenous IL-2, the population of CD28-CD3 + CD28 + cells proliferates better than CD8-PBMC simulated with anti-CD3 / 28 spheres. In addition, the addition of exogenous IL-2 (40 U / ml, three times per week) significantly increases the kinetics of proliferation both in terms of cell numbers and duration of effect (Figure 14). To evaluate the antiviral activity of this stimulation, the purified CD28-CD3 + CD28 + cells from four uninfected monkeys were infected on day 0 with 0.1 M.O.I. of SIV and then cultured under continuous stimulation. CD8-PBMC stimulated with PHA and IL-2 was the control of the experiment. Viral infection was followed by detection of the p27 Gag antigen in the culture supernatant by a commercial ELISA test (Coulter, Hialeah, FL). The p27 Gag antigen levels (ng / ml) are measured on day 6 and 12 after infection. As shown in Figure 15, there is a significant difference in infection in the two stimulation regimens. In fact, on day 6 after infection, the p27 antigen in cultures stimulated with CD3 / 28 spheres is 40% to 87% lower than cultures stimulated with PAH plus IL-2, and on day 12, this difference increases in two out of four monkeys. This suggests a reduction in susceptibility in the viral infection. In only one case (MK 9401) we observed a viral spread in both stimulation regimes. The results described here show that Macaca fascicularis is a good model for the ex vivo expansion of lymphocyte subpopulation by co-stimulation of anti-CD3 / 28 spheres, without viral replication. This represents the rationale for the therapeutic vaccine we propose, based on the expansion and reinfusion of specific autologous antiviral lymphocytes, in individuals infected with HIV.
Example 8. Use of dendritic cells for vaccination Dendritic cells (DC) and macrophages, to a lesser degree, are capable of effectively presenting antigens to T lymphocytes and inducing, in this subset of cells, the proliferation or acquisition of specific cytotoxic activities. These cells are called "antigen-presenting cells" (APC) and can initiate an immune response. Therefore, DC can be used in ex vivo immunization protocols. For this reason, DC precursors were isolated from Macaca fascicularis peripheral blood by culture in adherent cells after 7 days of stimulation with GM-CSF and IL-2. Alternatively, CD34 + cells were purified with immunomagnetic spheres and then cultured in vitro with GM-CSF and TNF-α for 14 days. To confirm that DC was isolated, morphological analysis and phenotypic characterization (FACS analysis and immunohistochemistry) were performed. Functional analysis is based on the unique ability of DC to induce proliferation of allogeneic lymphocytes. The results obtained completely confirm the effectiveness of the purification and the functional characterization of DC. In detail, to isolate the DC precursors, the PBMC obtained by centrifugation of a Ficoll density gradient, were stratified again in a discontinuous Percoll gradient (50% and 42.5%). The cell fraction which, after centrifugation at 500 g for 30 minutes, was between the two gradients consisted mainly of monocytes (as confirmed by FACS analysis, data not shown). These cells were kept at 4 ° C to avoid cell adhesion to the plastic tubes and were then harvested, washed, counted and plated in culture at 37 ° C. The day after, adherent cells were not removed by washing with 4 mild washes. To induce differentiation in DC, a complete medium supplemented with GM-CSF (200 ng / ml, Leucomax, Sandoz, Milan, Italy) and IL-4 (200 U / ml, Pepro tech, London, England) was added to the adherent cells. ). As a control, complete medium without cytokines was added to induce normal monocyte differentiation of the macrophage line. Twice a week, half of the supernatant was replaced with fresh medium identical to that used on day 0. The maturation of DC in wells treated with cytokines was detected by typical morphological changes, such as group formation, loss of adherence and development. of cellular ramifications. The monocyte / macrophage adherent cells grew without cytokine and were detached by EDTA treatment (0.5 mM in PBS-A), washed twice, counted and resuspended in fresh medium at different concentrations depending on the experiment performed. For the reactions of mixed allogeneic leukocytes (AMLR), the obtained APCs (DC or macrophages) were tested with a fixed amount of allogeneic T lymphocytes, purified by Ficoll and Percoll gradients, and by adhesion, and then frozen. The AMLR was performed in 48-well plates with 0.5 x 106 T lymphocytes and serial dilutions of APC. On day 4 of culture, a fixed amount of the cell suspension was seeded in a 96-well plate, in triplicate. 1 μCi of 3 H-thymidine was added to each well and the plate was then incubated at 37 ° C for 16 hours. At the end of the incubation, the amount of 3H-thymidine incorporated by the cells is measured with a β-counter and expressed as counts per minute (cpm). The results indicate that the DC that are obtained are potent APC as demonstrated by the greater induction of proliferation in human allogeneic lymphocytes, in comparison with the stimulation of macrophages, and by the ability to induce proliferation of T lymphocytes in monkeys at all concentrations used (figure 16B). For use in vaccination, DC will be resuspended at a concentration of 1 x 10 cells / 100 μl in RPMI 1640 supplemented with 5% autologous serum, 10 mM Hepes buffer, 100 U / ml of penicillin-streptomycin, 0.5 mg / ml of amphotericin B and 0.03% of glutamine, and then incubated for 32 hours at 37 ° C in the presence of Tat protein or Tat peptides, or combination of Tat, Rev, Nef, Gag and / or cytokines. Then, these treated DCs will be inoculated twice or more in the next 2-4 weeks from the first injection, intravenously. Alternatively, DCs will be transduced with vectors containing the Tat gene alone or associated with other vectors mentioned before and then injected intravenously.
Prophetic example 9 The described immunogens will be used in order to induce and / or enhance a specific immune response at the mucosal level. One of the approaches is based on the use of "genetically engineered" bacteria (S. gordonii and Lactobacillus) to express the aforementioned viral antigens. These bacteria colonize the oral and vaginal mucosa of mice and induce a specific response of both local and systemic antibodies against heterologous antigens expressed on the surface of recombinant bacteria (Ref. 116, 104, 106, 121, 117, 139, 105, 107) . These bacteria can work as live vectors of vaccines and can take advantage of the fact of causing a prolonged stimulation of the immune system. In addition, we will evaluate the possibility of coexpressing, on the bacterial surface, viral antigens and molecules involved in the immune response such as the B subunit of the temperature sensitive toxin of E. coli or cytokines. The preparation of the recombinant strains of S. gordonii will be carried out as previously described (Ref. 116), briefly, (i) the chromosomal integration of the recombinant DNA molecules; (ii) transcriptional fusions with strong chromosomal promoters; (iii) transcriptional fusions with the gene that codes for the M6 protein, a surface protein of Streptococcus. Recombinant strains of S. gordonii will be used to colonize the vaginal mucosa of monkeys. It has been shown that recombinant strains of S. gordonii, which express the V3 region of HIV-1 gpl20 and the E7 protein and the HPV-16 protein, permanently colonize the vaginal mucosa of the mouse after a single inoculum, which induces a specific antibody response to the antigen both locally and systemically. The systemic response is in a prevalence composed of IgG2a antibodies, which suggests a Thl type response (Ref. 105, 106). They selected human Lactobacillus vaginal strains, which are capable of colonizing the vaginal mucosa of monkeys. Subsequently, an already developed genetic system will be used, which allows the expression of heterologous antigens on the surface of Lactobacillus (Rush, 1997). This strategy is based on: (i) cloning of genetic fusions (emmi / heterologous gene) into insertion vectors which carry homologies with the conjugative transposon Tn916; (ii) transformation of the vectors into bacterial strains which work as an intermediate host (Bacillus subtilis); (iii) conjugative mobilization of the recombinant transposons of B. Subtilis to Lactobacillus. Recombinant strains of Lactobacillus will be used to colonize the vaginal mucosa of monkeys. Vaginal samples will be obtained using special absorbent filters (Ref. 38, 105, 106). Colonization will be evaluated by plating the vaginal samples on selective plaques and the expression of HIV antigens in vivo will be monitored by immunofluorescence of vaginal swabs (Ref: 105). By using already standardized methods (Ref. 38), vaginal swabs will be used for: i) Pap test, in the case of vaginal vaccination; ii) presence of vaccine antigens in the cells; iii) phenotypic characterization of the cells by cytofluorometric analysis (CD1, CD2, CD4, CD5, CD8, CD14, CD14, CD28, CD40, CD25, HLA-DR); iv) evaluation of cytokine production (IL-2, INF ?, TNFa, IL-4, IL-10, IL-15, semiquantitative RT-PCR), determination of the presence of cytokines and β-chemokines in mucosal fluids ELISA assays; v) dosing of total and specific immunoglobulins (IgA and IgG) in the mucosal fluid by ELISA [Di Fabio et al. , Vaccine 15: 1 (1997)]. One month after the last inoculum of the immunogen, the monkeys will be inserted intravenously or through the mucosal route with SHIV89.6P. The monitoring of the monkeys is carried out as described in example 4. Blood samples will be obtained in order to perform routine or systematic laboratory tests, the evaluation of both humoral and cellular immunological parameters, as described in the example 4. The inventor considers that this method can be used successfully to induce specific immunization in monkeys, using the vaginal route.
Alternatively, mucosal immunity can be induced by administration of the protein inmonogens, described above, directly through the mucosal route in the presence of adjuvants, such as heat-sensitive E. coli toxin and cholera toxin, or using other bacterial delivery systems. and non-bacterial such as cytofectins and liposomes' or through inoculation by other routes which are capable of inducing a more efficient and protective immune response (Ref. 83, 81, 62). In addition, the inventor considers that the recombinant herpes vectors, which express the viral proteins described above, can be excellent systems for inducing an effective mucosal immune response. The recombinant viral vectors of herpes simplex virus type 1 (HSV-1) will be used to express viral proteins for the induction of systemic (through skin immunization, id) and mucosal (through the oral, vaginal or nasal) responses ). Non-pathogenic and non-replicative herpes vectors (Ref. 99) will be used for their ability to induce large exogenous sequences without interfering with the efficacy of the insertion (Ref. 52, 64). Therefore, vectors capable of containing more than one HIV gene (accessory, regulatory and structural) will be constructed. Mucosal immunity can be induced by an oral, vaginal or nasal vaccine. Herpes vectors can be used in these vaccine approaches, since HSV-1 can be administered directly by the mucosal route (Ref. 176, 75). The recombinant viruses will be constructed using a two-step method which facilitates the insertion of exogenous sequences into the viral genome. The first stage requires the insertion of an expression cassette with a reporter gene (ß-galactosidase, LacZ) cloned into the PacI restriction site, which is not present in the HSV-1 genome, flanked by the target sequence that is desired of HSV-1, using the standard procedure for homologous recombination, to interrupt the HSV-1 gene. The recombinant virus is selected by plaque formation with a blue phenotype, using "x-gal staining". The digestion of viral DNA with Pací releases the marker gene and generates two large fragments of viral DNA, unable to produce infectious viral particles. The second step consists of a cotransfection of viral DNA, digested with the same plasmid used to create the deletion, wherein the reporter gene is replaced by the desired gene. Recombinant viruses will be identified through selection of plaques with a white phenotype after "x-gal staining". This recombination will lead to the elimination of the Pací sites, which allows the use of this method to insert many genes in the different HSV-1 genome (Ref. 74). By crossing the different vectors that contain the genes alone, we are probably able to create all the different genetic combinations. The vector containing all the desired genes will be isolated by analysis with different markers, phenotypes and selective growth in competent cells. All combinations will be created by alternating DNA transfections and viral recombinations. The vectors expressing the tat, rev, nef or gag single genes will be constructed using, as a base vector, one that contains the mutations in the 4- / 22- / 27- / 41 genes, which is better for the low toxicity and a strong expression of the exogenous gene, in comparison with other non-replicative HSV-1 vectors. Constitutive promoters, such as those of HCMV (human cytomegalovirus immediate early promoter) or IPCO lep (immediate early promoter of infected cell protein) and LTR of Moloney murine leukemia virus, will be used to induce the expression of the genes mentioned before. Non-replicative HSV-1 vectors expressing HIV proteins in different combinations will be constructed. The production of these viruses containing more different genes will be obtained by a genetic crossing over the vectors containing single genes described in the previous point. Double vectors will be created, triples and quadruples. The vectors will be inoculated on the i.d monkeys. or through the mucosal route (oral, vaginal or nasal) with particular attention to this last type of administration (Ref. 176, 101, 102). The vaccination protocol consists of multiple inocula at different time points, which must be determined in relation to the immunogen or the combination of immunogens. During the immunization, the animals are monitored for the evaluation of hematochemical and immunological parameters in example 4. With already standardized methods, vaginal samples will be obtained that will be studied as previously described in this example.
Example 10 prophetic Supply systems Tat (protein and / or DNA) will be inoculated alone or in combination (as described above) using new delivery systems such as erythrocytes or nanoparticles. The supply system that involves the use of erythrocytes is based on the possibility of supplying the antigen bound to autologous erythrocytes. In fact, the erythrocytes, at the end of their life period (approximately 120 days in humans), are removed from the circulation by the macrophages, which are known to function as professional antigen presenting cells. This property can be used for vaccine strategies. Therefore, the antigens will bind to the erythrocytes with a particular technique (Ref. 95, 96), which allows the preservation of the immunogenic properties of the antigen (Ref: 29, 30). By means of this procedure, the biotination of erythrocytes can be carried out in the absence of a significant modification of their properties and lifespan (Ref. 95). Phagocytosis of old erythrocytes by macrophage cells will begin an immune response. The opsonization antibodies of the erythrocytes that present the antigen will help the removal of the antigen from the circulation. The main advantages of this methodology are: 1) a small amount of antigen necessary to induce a humoral and cellular immune response, 2) long-lasting immunization due to the long-lasting presence of antigens carried by the erythrocytes in the periphery, 3) adjuvant functions provided by the system itself. In fact, it has been demonstrated in animal studies that the administration of antigens bound in the membrane of autologous erythrocytes induces a similar or superior immune response in comparison with the immune response obtained with the same antigen administration with Freund's adjuvant.
(Ref. 29). These properties are very useful for developing an HIV vaccine, particularly when it is necessary to increase the immunogenicity of the antigen and the availability of the antigen, and when a low number of immunizations are required. In addition, this strategy can be used when adjuvants are not included in the vaccination protocol. In fact, it has been shown in the mouse model that antigens administered through autologous erythrocytes induce similar or superior immune responses compared to those obtained with the same antigen administration with Freund's adjuvant which is known as the most powerful adjuvant available. commercially (Ref. 29), although it has not been approved for human studies due to the importance of the side effects. Therefore, the adjuvant effect of erythrocytes presenting Tat protein, alone or in combination with other immunogens previously described, will be analyzed in non-human primates. A comparison will be made between these data and those obtained with the administration of Tat protein in the presence of alumina, RIBI or ISCOM. The use of nanoparticles can represent an additional supply strategy. The functional nanoparticles represent an important system for the transport and release of proteins and DNA (Ref. 27, 172). Nanospheres are colloidal polymer particles of different chemical composition, with a large diameter range from 10 to 1000 nm. It is possible to absorb different kinds of substances on the surface or within the nanospheres (oligonucleotides, drugs, proteins, peptides, DNA) and then they are taken to the cytoplasm or cell nucleus where they are released slowly. In addition, a small amount of immunogen needs to be delivered due to the characteristics of the nanospheres. Nanoparticles are a suitable delivery system, especially for molecules with low stability in the extracellular environment or when the supply is directed to a specific target cell. The inventor considers that nanospheres can be used for the delivery of viral antigens described above. It is possible to prepare and characterize three types of nanospheres designed for the delivery and controlled release of DNA (nanospheres type 1 and 2) and proteins (nanospheres type 3). For the supply of DNA, two types of nanospheres are available (nanospheres type 1 and 2). The first type of nanospheres (type 1 nanospheres) has a triple layer structure with an outer layer of polyoxyethylene glycol (PEG). Recent reports based on studies of surreptitious systems (Ref. 180, 78) show that PEG produces invisible nanospheres for Kupfer cells. In contrast, the innermost layer is made of monomers with surfactant characteristics containing quaternary ammonium groups that reversibly absorb the DNA through an ion exchange mechanism and an internal core made of methyl methacrylate as a monomer. These nanospheres are obtained by microemulsion polymerization involving the polymerization of a vinyl or vinylidene monomer in the presence of a mixture of surfactant reagents. These reagents are therefore capable of polymerizing the monomer. Of these, one has a quaternary ammonium group that interacts with the oligonucleotides and the other has a long chain of PEG. The second type of DNA delivery system is made of nanospheres and functional microspheres (type 2 nanospheres) with hydrogel characteristics. These nanospheres can be made in the presence of DNA to trap it within the delivery system. Core-shell nanospheres are needed to supply proteins (type 3 nanospheres). They are manufactured by an internal core of polymethyl methacrylate and an outer shell of water-soluble static copolymer of acrylic acid and methyl methacrylate known to have a high degree of affinity for proteins (Ref. 79, 80). This copolymer is commercially available (EUDRAGIT) and is obtained with different percentages of the two comonomers. The preparation process leading to the elaboration of this second type of nanospheres involves dispersion polymerization. The synthesis involves the radical polymerization of a vinyl or vinylidene monomer in the presence of EUDRAGIT which has steric stabilizing functions. After the nucleation of the nanospheres, EUDRAGIT is arranged outside the particles. Therefore, the concentration of the radical initiator, the ratio between the monomer and EUDRAGIT and the reaction time are modified, numerous samples of nanospheres with different morphological and chemical characteristics are obtained.
Therefore, it can be evaluated whether the delivery of Tat protein or Tat DNA by nanoparticles, alone or in combination with the aforementioned immunogens (either protein or DNA) will induce an immune response against HIV. In particular, humoral or cellular mediated immune responses will be evaluated and compared with those obtained with immunogens not provided in the mono model. The inventor considers that the information derived from these studies can be useful for the development of an HIV vaccine. In addition, the information derived from this experimental protocol will also be transferred to other vaccine studies, particularly those studies that work with recombinant proteins or peptides of low immunogenicity. The possibility of developing a vaccine with a single administration leads to enormous advantages in terms of vaccine efficacy and reduction of vaccine program management costs.
References 1. Agostini et al., Blood 90: 1115 (1997) 2. Albini et al., Proc. Nati Acad. Sci. USA 92: 4838 (1995) 3. Allan et al., Science 230: 813 (1985) 4. Antibodies - A laboratory manual, Eds. Hadow E., Lane D., Cold Spring Harbor Laboratory (1988) 5 Arya et al., Science 229: 69 (1985) 6 Aryoshi et al., AIDS 9: 555 (1995) 7 Audibert et al., Immunol. Today 14: 28_ (193) 8 Badolato et al., Blood 90: 2804 (1997) 9 Barillari et al., J. Immunol. 149: 3727 (1992) Barillari et al., Proc. Nati Acad. Sci. USA 90: 7941 (1993) 11 Barillari et al., Proc. Nati Acad. Sci USA 149: 3727 (1993) 12 Blomberg et al., J. Immunol. Methods 160: 27-34 (1993) 13, Blomberg et al., J. Immunol. Methods 168: 267-273 (1994) 14, Blomberg et al., J. Immunol. Methods 193: 199-206 (1996) 1.5, Bohan et al., Gene Expr. 2: 391 (1992) 16 Bourgault et al., J. Virol. 66:75 (1992) 17, Boyer et al., Nature Med. 3: 526 (1997) 18, Bruisten et al., J. Infect. Dis. 166: 620 (1992) 19, Buseyne et al., J. Virol. 67: 694 (1993) 20. Butera et al., J. Viro¡. 65: 4645 (1991) 21. Butera et al., J. Virol. 68: 2726 (1994) 22. Cafaro et al., AIDS Res. Hum. Retrov. 7: 204 (1991) 23. Carrol et al, Science. 276: 273-276, (1997) 24. Carson et al., J. Clin. Invest. 99: 937 (1997) 25. Chang et al., J. Biomed. Sci. 2: 189 (1995) 26. Chang et al., AIDS 11: 1421 (1997) 27. Chavany et al., Phar. Res. 9: 441 (1994) 28. Chen et al., J. Immunol. 149: 4060 (1992) 29. Chiarantini et al., Vaccine 15: 276 (1997) 30. Chiarantini et al., Clin. Diag. Lab. Immunol. 5: 235 (1998) 31. Chirmule et al., J. Virol. 69: 492 (1995) 32 Choppin et al., J. Immunol. 147: 569 (1991) 33. Corallini et al., Cancer Res. 53: 1 (1993) 34. Corallini et al., Cancer Res, 53: 5569 (1993) 35. Culman et al., J. Immunol. 146: 1560 (1991) 36. Couillin et al., J. Exp. Med. 180: 1129 (1994). 37. Danko et al., Vaccine 12: 1499 (1994) 38. Di Fabio et al., Vaccine 15: 1 (1997) 39, Ensoli et al., IV International Conference on AIDS, Stockholm, 1: 241 (1988) 40, Ensoli et al., Nature 345: 84 (1990) 41, Ensoli et al., J. Virol. 67: 277 (1993) 42. Ensoli et al., Nature 371: 674 (1994) 43. Ensoli et al., AIDS Updates, Eds. V. DeVita, Jr., Hellman S., Rosenberg S.A., Lippincott J.B., Philadelphia; 7: 1 (1994) 44 Felber et al., Proc. Nati Acad. Sci. 86: 1495 (1989) 45 Fine et al., Ann. Plast. Surg. 20: 6 (1988) 46, Fiorelli et al., J. Clin. Invest. 95: 1723 (1995) 47 Folks et al., Science 238: 800 (1987) 48 Franchini et al., Virology 155: 593 (1986) 49, Frankel et al., Cell 55: 1189 (1988) 50. Fugier-Vivier et al, J. Exp. Med. 186: 813 (1997) 51. Gait et al., Trends Biochem. Sci. 18: 255 (1993) 52. Glorioso et al., Ann. Rev. Microbiol. 49: 675 (1995) 53. Gobert et al., Virology 176: 458 (1990) 54, Goletti et al., J. Virol. 69: 2540 (1995) 55, Gorman et al., Mol. Cell. Biol. 2: 1044 (1982) 56. Grabstein et al., Science 264: 965 (1994) 57. Grosjean et al, J. Exp. Med. 186: 801 (1997) 58. Guy et al., Nature 330: 266 (1987) 59. Harrer et al., AIDS Res. Hum. Retrov. 12: 585 (1996) 60 Harrich et al., EMBO J. 16: 6 (1997) 61 Hinkula et al., J. Virol. 71: 5528 (1997) 62 Honenbang et al., Ln ect. Immun. 62:15 (1994) 63 Huang et al., EMBO J. 13: 2886 (1994) 64 Huard et al. Gene Ther. 2: 385 (1995) 65 Igarashi et al., AIDS Res. Hum. Retrov. 10: 1021 (1994) 66. Jonuleit et al., J. Immunol. 158: 2610 (1997) 67. Jullien et al., J. Immunol. 158: 800 (1997) 68. Kanai et al., J. Immunol. 157: 3681 (1996) 69. Karlosson et al., J. Virol. 71: 4218 (1997) 70. Kashanchi et al., J. Virol. 70: 5503 (1996) 71. Kestler et al., Science 248: 1109 (1991) 72. Kim et al., Oncogene 7: 1525 (1992) 73. Koup et al., J. Virol. 68: 4650 (1994) 74. Krisky et al., Gene Ther. 4: 1120 (1997) 75. Kuklin et al., J. Virol. 240: 245 (1998) 76. Landes, Austin, p. 107 (1997) 77. Lanzavecchia, Science 260: 937 (1993) 78. Lasic et al., Chemical Reviews 95: 2601 (1995) 79. Laus et al., Polymer 37: 343 (1996) 80. Laus et al. , Polymers for Adv. Techn. 7: 548 (1996) 81. Lehnen et al., Vaccine Research 1: 319 (1992) 82. Levine et al., Science. 272: 1939-1943 (1996) 83. Lewis et al., Vaccine Press, Ed. Robinson, Farrar, Wibiin; Human Press, Totowa, New Jersey (1996) 84 Li et al., Proc. Nati Acad. Sci. USA 94: 8116 (1997) 85. Li et al., J. AIDS 5: 639 (1992) 86. Li et al., Science 268: 229 (1995) 87. Li et al., Proc. Nati Acad. Sci. USA 94: 8116 (1997) 88. Lippincott J.B., Stockholm, Sweden, 31-May-3 June (1997) 89. Littaua et al., J. Virol. 65:40 (1991) 90. Lóvgren et al. ", Vaccine 14: 753 (1996) ~ 91. Lu et al., J. Virol. 70: 3978 (1996) 92. Lubaki et al., J. lnfect. Dis. 175: 1360 (1997) 93. Lucey et al., Clin. Lab Lab. Immunol. (1997) 94. Luciw et al., Proc. Nati Acad. Sci. 92: 7490 (1995) 95. Magnani et al., Biotech. Appl. Biochem. 16: 188 (1992) 96. Magnani et al., Biotech. Appl. Biochem. 20: 335 (1994) 97 Malim et al., Nature 338: 254 (1989) 98 Mann et al., EMBO J. 10: 1733 (1991) 99 Marconi et al., Proc. Nati Acad. Sci 93: 11319 (1996) 100 Marcuzzi et al., J. Virol. 66: 4228 (1992) 101 McLean et al., J. lnfect. Dis. 66: 341 (1994) 102 McLean et al., Vaccine 14: 987 (1996) 103 Mcfarland et al., J. Inf. Dis. 170: 766 (1994) 104 Medaglini et al., Proc. Nati Acad. Sci. USA 92: 6868 (1995) 105. Medaglini et al., Biotech. Annu. Rev. 3: 297 (1997) 106, Medaglini et al., Vaccine 15: 1330 (1997) 107, Medaglini et al., Am. J. Reprod. Immunol. 39: 199 (1998) 108 Meyerhans et al., Cell 58: 901 (1989) 109 Morein et al., AIDS Res. Hum. Retrov. S10: S109 (1994) 110 Molecular cloning - A laboratory manual; Eds. Maniatis T., Fritsch E.F., Sambrook J., Cold Spring Harbor Laboratory, Cold Spring Harbor, New York (1992) 111 Myers et al. , Human Retroviruses and AIDS: A compilation and analysis of nucleic acid and amino acid sequences, Los Alamos Laboratory, Los Alamos, NM p.l (1993) 112 Myers et al. , Human Retroviruses and AIDS. Theoretical Biology and Biophysics Group. Los Alamos, NH (1995) 113 Neuvet et al., J. Virol. 70: 5572 (1996) 114 Nietfield et al., J. Immunol. 154: 2189 (1995) 115, Nixon et al., Nature 336: 484 (1988) 116 Oggioni et al., Vaccine 13: 775 (1995) 117 Oggioni, et al., Gene 169: 85 (1996) 118 O ' Hagan et al., Novel Delivery Systems for Oral Vaccines, Eds. O 'Hagan, D.T. CRC Press Boca Raton, FL, p. 176 (1994) 119 Parslow, Human Retroviruses, Ed. B.R. Cullen, IRL press, Oxford, England, p. 101 (1993) 120 Pilkington et al., Mol. Immunol. 33: 439 (1996) 121. Pozzi et al., In "Gram-positive bacteria as vaccine vehicles for mucosal immunization", eds. Pozzi G. & Weiis, J.M. - Landes, Austin, p. 35 (1997) 122 Puri et al., Cancer Res., 52: 3787 (1992) 123 Puri et al., AIDS Res. 11:31 (1995) 124 Quesada-Rolander et al., ABS 6-SI 2nd European Conference on Experimental AIDS Research, Stockholm, Sweeden, May 31-June 3 (1997) 125 Quinn et al., Biochem. Biophys. Res. Commun. 239: 6 (1997) 126, Ratner et al., Nature 313: 277 (1985) 127, Re et al. , J. Acquir. Immun. Defic. Syndr. 10: 408 (1995) 128 Reimann et al., J. Virol. 70: 3189 (1996) 129 Reimann et al., J. Virol. 70: 6922 (1996) 130 Reiss et al., J. Med Virol. 30: 163 (1990) 131. Reiss et al., AIDS Res. Hum. Retrov. 5: 621 (1989) 132, Riley et al, J. Immunol. 158: 5545-5553, (1997) 133 Rinaldo et al., AIDS Res. Hum. Retrov. 11: 481 (1995) 134 Rinaldo et al., J. Virol., 69: 5838 (1995) 135, Rodman et al., Proc. Nati Acad. Sci. USA 90: 7719 (1993) 136. Rodman et al., J. Exp. Med. 175: 1247 (1992) 137, Rosenberg et al., Int. Immunol. 9 (5): 703 (1997) 138. Rosenthal et al., Seminars in Immupology 9: 303 (1997) 139 Rush, et al., In "Gram-positive bacteria as vaccine vehicles for mucosal immunization", eds. Pozzi G. & Wells, J.M. - Landes, Austin, p. 107 (1997) 140 Sadaie et al., New Biol. 2: 479 (1990) 141, Saiki et al., Science 230: 1350 (1985) 142, Sakuragi et al., J. Gen. Virol. 73: 2983 (1992) 143 Salter et al., Immunogenetics 21: 235 (1985) 144. Schnorr et al, Proc. Nati Acad. Sci. USA. 94: 5326 (1997) 145 Sharma et al., Biochem. Biophys. Res. Co. 208: 704 (1995) 146, Shibata et al., J. Virol. 65: 314 (1991) 147 Sipsas et al., J. Clin. Invest. 99: 752 (1997) 148 Sodroski et al., Science 227: 171, (1985) 149 Steinaa et al., Arch. Virol. 139: 263 (1994) 150 Steinman R.M. , Exp. Hematol. 24: 859 (1996) 151 Tahtinen et al., Virology 187: 156 (1992) 152 Theoretical Biology and Biophysics, Los Alamos, NH, (1995) 153 Titti et al., Cell. Pharmacol. AIDS 3: 123 (1996) 154 Trinchieri, Curr. Opin. Hematol. 4:59 (1997) 155, van Baalen et al., J. Gen. Virol., 77: 1659 (1996) 156. van Baalen et al., J. Gen. Virol. 78: 1913 (1997) 157 Vellutini et al., AIDS Res. Hum. Retrov. 11:21 (1995) 158. Venet et al., J. Immunol. 148: 2899 (1992) 159. Viscidi et al., Science 246: 1606 (1989) 160. Vogel et al., Nature 335: 601 (1988) 161. Voss et al., Virology 208: 770 (1995) 162. Wain-Hobson, Curr. Opin. Genet Dev. 3: 878 (1993) 163 Westendorp et al., J. Virol. 68: 4177 (1994) 164 Westendorp et al., Nature 375: 497 (1995) 165 Wolf et al., J. Immunol. 146: 3074 (1991) 166 Yang et al., J. Virol. 70: 4576 (1996) 167 Yang et al., J. Virol. 70: 5799 (1996) 168 Yasutomi et al., J. Virol. 70: 678 (1996) 169 Zauli et al., Blood 86: 3823 (1995) 170 Zauli et al., Blood 80: 3036 (1996) 171. Zauli et al., J. Immunol. 157: 2216 (1996) 172. Zobel et al., Antisense Nucieic Acid Drug Dev. 7: 483 (1997) 173. Gibellini et al., Blood 89: 1654 (1997) 174. Bauer et al., J. Infect. Dis. 165: 419 (1992) 175. Klein et al., J. Exp. Med. 181: 1365 (1995) 176. Bowen et al., Res. Virol. 143: 269 (1992) 177. Zamarchi et al., AIDS Res. Human Retrov. 9: 1139 (1993) 178 Fiore et al., AIDS 5: 1034 (19912) 179 Román et al., Nature Med. 3: 849 (1997) 180, Alien et al., Biochim. Biophis. Acta 1237: 99 (1995) LIST OF SEQUENCES < 1 10 > ISTITUTO SUPERIORE DI SANITA * < 120 > Tat of HIV-l or derivatives thereof, alone or in combination for prophylactic and therapeutic vaccination against AIDS, tumors and related syndromes < I 30 > 1354PTWO < 140 > RM97A000743 < 141 > 1997-12-01 < 160 > 34 < 170 > Patentln Ver. 2.0 < 210 > 1 < 21 1 > 261 < 212 > A DN < 213 > AIDS-associated retrovirus < 400 > 1 atggagccag tagatcctag actagagccc tggaagcatc caggaagtca gcctaaaact 60 gcttgtacca attgctattg taaaaagtgt tgctttcatt gccaagmg tttcataaca 120 aaagccttag gcatctccta tggcaggaag aagcggagac agcgacgaag acctcctcaa January 80 ctcatcaagt ggcagtcaga ttctctatca aagcagccca cctcccaatc ccgaggggac 240 g 261 ccgacaggcc cgaaggaata < 210 > 2 < 21 1 > 83 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 2 Glu Pro VaL Asp Pro Arg Leu G iu Pro Trp Lys His Pro Gly Ser Gln 10 15 Pro Lys Thr Ala Cys Thr Asn Cyl Tyr Cys Lys Lys Cys Cys Phe His 20 25 30 Cys Gln Val Cys Phe He Thr Lys Wing He Ser Tyr Gly Arg Lys Lys 35 40 45 Arg Arg Gin Arg Arg Pro Pro Gln Gly Ser Gln M u His Gln Val 50 55 60 Ser Leu Ser Lys GIn Pro Thr Ser Gln Ser Arg Gly Asp Pro Gly 65 70 75 80 Pro Lys Glu < 210 > 3 < 21 1 > 260 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 3 atggagccag tagatcctag actagagccc tggaagcatc caggaagtca gcctaaaact 60 gcggtaccaa ttgctattgt aaaaagtgtt gctttcattg ccaagtttgt ttcataacaa 120 aagccttagg catctcctat ggcaggaaga agcggagaca gcgacgaaga cctcctcaagl 80 gcagtcagac tcatcaagtt tctctatcaa agcagcccac ctcccaatcc cgaggggacc 240 260 cgacaggccc gaaggaatag < 210 > 4 < 21 1 > 261 < 212 > DNA < 213 > retrovirus associated with AIDS < 400 > 4 atggagccag tagatcctag actagagccc tggaagcatc caggaagtca gcctaaaact 60 gcttgtacca attgctattg taaaaagtgt tgctttcatt gccaagtttg tttcataaca 120 aacgccttag gcatctccta tggcaggaag aagcggagac agcgacgaag acctcctcaa 180 ctcatcaagt ggcagtcaga aagcagccca ttctctatca cctcccaatc ccgaggggac ccgacaggcc cgaaggaata 240 g 261 < 210 > 5 < 21 1 > 252 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 5 atggagccag tagatcctag actagagccc tggaagcatc caggaagtca gcctaaaact 60 gcttgtacca attgctattg taaaaagtgt tgctttcatt gccaagtttg tttcataaca 120 aaagccttag gcatctccta tggcaggaag aagcggagac agcgacgaag acctcctcaa 180 ggcagtcaga ctcatcaagt ttctctatca aagcagccca cctcccaatc cccgacaggc ccgaaggaatag 240 252 < 210 > 6 < 21 1 > 252 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 6 atggagccag tagatcctag actagagccc tggaagcatc caggaagtca gcctaaaact 60 gcttgtacca attgctattg taaaaagtgt tgctttcatt gccaagtttg tttcataaca 120 aacgccttag gcatctccta tggcaggaag aagcggagac agcgacgaag acctcctcaa 180 ctcatcaagt ggcagtcaga aagcagccca ttctctatca cctcccaatc cccgacaggc ccgaaggaat 240 ag 252 < 210 > 7 < 211 > 86 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 7 Met Glu Pro Val Asp Pro Arg Leu Glu Pro Trp Lys His Pro Gly Ser 10 15 Gln Pro Lys Thr Wing Gly Thr Asn Cys Tyr Cys Lys Lys Cys Cys Phe 20 25 30 His Cys Gln Val Cys Phe He Thr Lys Wing Leu Gly He Ser Tyr Gly 35 40 45 Arg Lys Lys Arg Arg Gln Arg Arg Pro Pro Gln Gly Ser Gln Thr 50 55 60 His Gin Val Ser Leu Ser Lys Gln Pro Thr Ser Gln Ser Arg Gly Asp 65 70 75 80 Pro Thr Gly Pro Lys G lu 85 < 210 > 8 < 2 I 1 > 86 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 8 Met Glu Pro Val Asp Pro Arg Leu G lu Pro Trp Lys His Pro G l and Ser 1 5 10 15 Gln Pro Lys Thr A la Cys Thr Asn Cys Tyr Cys Lys Lys Cys Cys Phe 20 25 30 His Cys Gln Val Cys Phe lie Thr Thr A la Leu Gly He Ser Tyr Gly 35 40 45 Arg Lys Lys Arg Arg Gln Arg Arg Pro Pro Gin Gly Ser Gin Thr 50 55 60 His G ln Val Ser Leu Ser Lys Gln Pro Thr Ser G In Ser Arg Gly Asp 65 70 75 '80 Pro Thr Gly Pro Lys G lu 85 < 210 > 9 < 21 1 > 83 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 9 Met Glu Pro Val Asp Pro Arg Leu Glu Pro Trp Lys His Pro Gly Ser 1 5 10 15 Gln Pro Lys Thr Ala Cys Thr Asn Cys Tyr Cys Lys Lys Cys Cys Phe 20 25 30 His Cys Gln Val Cys Phe lie Thr Lys Wing Leu Gly He Ser Tyr Gly 35 40 45 Arg Lys Lys Arg Arg Gin Arg Arg Pro Pro Gln Gly Ser Gln Thr 50 55 60 H is G ln Val Ser Leu Ser Lys Gln Pro Thr Ser GIn Ser Pro Thr G ly 65 70 75 80 Pro Lys G lu < 210 > 10 < 21 1 > 83 < 212 > PRT < 2 13 > retrovirus associated with AIDS < 400 > 10 Met Glu Pro Val Asp Pro Arg Leu Glu Pro Trp Lys His Pro Gly Ser 1 5 10 15 Gln Pro Lys Thr Ala Cys Thr Asn Cys Tyr Cys Lys Lys Cys Cys Phe 20 25 30 His Cys Gin Val Cys Phe He Thr Thr Ala Leu Gly He Ser Tyr Gly 35 40 45 Arg Lys Lys Arg Arg Gln Arg Arg Pro Pro Gln Gly Ser Gln Thr 50 55 60 His Gln Val Ser Leu Ser Lys Gln Pro Thr Ser Gln Ser Pro Thr Gly 65 70 75 80 Pro Lys Glu < 210 > 11 < 21 1 > 20 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 1 1 Met Glu Pro Val Asp Pro Arg Leu Glu Pro Trp Lys His Pro Gly Ser 1 5 10 15 Gln Pro Lys 20 < 210 > 12 < 21 1 > 20 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 12 Ala Cys Thr Asn Cys Tyr Cys Lys Lys Cys Cys Phe His Cys Gln Val I 5 10 15 Cys Phe lie Thr 20 < 210 > 13 < 211 > 16 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 13 Gin Val Cys Phe He Thr Lys Wing Leu Gly lie Ser Tyr Gly Arg Lys II 5 10 15 < 210 > 14 < 2II > 15 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 14 Ser Tyr Gly Arg Lys Lys Arg Arg Gln Arg Arg Arg Pro Pro Gln 1 5 10 15 < 210 > 15 < 21 1 > 16 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 15 Arg Pro Pro Gln Gly Ser Gln Thr His Gln Val Ser Leu Ser Lys Gln 1 10 15 < 210 > 16 < 21 1 > 16 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 16 His GIn Val Ser Leu Ser Lys Gln Pro Thr Ser Gln Ser Arg Gly Asp 1 5 10 15 < 210 > 17 < 211 > 14 < 212 > PRT < 213 > AIDS-associated retrovirus < 400 > 17 Pro Thr Ser Gln Ser Arg Gly Asp Pro Tm Gly Pro Lys Glu 1 5 10 < 210 > 18 < 211 > 15 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 18 atggcaggaa gaagc 15 < 210 > 19 < 211 > 15 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 19 ctattcttta gttcc 15 < 210 > 20 < 211 > 15 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 20 atgggtggca agtgg 15 < 210 > 21 < 21 I > 15 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 21 tcagcagtcc ttgta 15 < 210 > 22 < 21 1 > 15 < 2 I 2 > A DN < 213 > AIDS-associated retrovirus < 400 > 22 atgggtgcga gagcg 15 < 210 > 23 < 211 > 15 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 23 ttattgtgac gaggg 15 < 210 > 24 < 211 > 15 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 24 atgtggcccc ctggg 15 < 210 > 25 < 211 > 15 < 212 > DNA < 213 > retrovirus associated with AIDS < 400 > 25 ttaggaagca ttcag 15 < 210 > 26 < 21 1 > 15 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 26 atgagaam cgaaa 15 < 210 > 27 < 211 > 15 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 27 tcaagaagtg ttgat 15 < 210 > 28 < 211 > 15 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 28 atggagccag tagat 15 < 210 > 29 < 21 I > 15 < 212 > A DN < 213 > AIDS-associated retrovirus < 400 > 29 ctattccttc gggcc 15 < 210 > 30 < 21 1 > 27 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 30 ggcccgaagg aaatggcagg aagaagc 27 < 210 > 31 < 21 1 > 27 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 3 1 ggcccgaagg aaatgggtgg caagtgg 27 < 210 > 32 < 21 1 > 27 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 32 ggcccgaagg aaatgggtgc gagagcg 27 < 210 > 33 < 21 1 > 27 < 212 > DNA < 213 > retrovirus associated with AIDS < 400 > 33 ggcccgaagg aaatgtggcc ccctggg 27 < 210 > 34 < 21 1 > 27 < 212 > DNA < 213 > AIDS-associated retrovirus < 400 > 34 ggcccgaagg aaatgagaat ttcgaaa 27 It is noted that in relation to this date, the best method known to the applicant to carry out the aforementioned invention, is the conventional one for the manufacture of the objects or products to which it refers.

Claims (56)

MS Having described the invention as above, the content of the following claims is claimed as property:
1. Biologically active isolated Tat protein, fragments thereof and / or mutants and / or tat DNA, for use as a vaccine, Tat at picomolar to nanomolar concentrations is able to: (i) enter and localize in the activated endothelial cell nuclei or dendritic cells; and / or ~ (ii) activate the proliferation, migration and invasion of Kaposi's sarcoma cells (KS) and cytokine-activated endothelial cell proteins.
2. Biologically active isolated Tat protein, fragments thereof and / or mutants and / or Tat DNA, according to claim 1, further characterized because it is capable of: (iii) activating virus replication when added to infected cells, as measured by: a) the rescue of defective proviruses in Tat in HLM-l cells after the addition of exogenous protein; and / or b) by transactivating the expression of the HIV-1 gene in cells transfected with an HIV-1 promoter-indicator plasmid.
3. Biologically active isolated Tat protein, fragments thereof and / or Tat mutants and / or DNA, according to claim 2, further characterized in that it is capable of: (iv) inducing in mice the development of KS-like lesions in the presence of angiogenic factors or inflammatory cytokines.
4. Biologically active isolated Tat protein, fragments thereof and / or mutants and / or Tat DNA, according to claims 1 to 3, characterized is supplied in amounts ranging from 10 ng / ml to 1 μg / ml.
5. Biologically active isolated Tat protein, fragments thereof and / or mutants and / or Tat DNA, according to claims 1 to 4, characterized is used in the prophylactic and / or therapeutic treatment of AIDS, tumors, syndromes and symptoms associated with HIV infection.
6. A protein or peptide or DNA vaccine, prophylactic and / or therapeutic, against AIDS, tumors associated with AIDS, syndromes and symptoms associated with HIV infection, comprising biologically active Tat and / or its mutants and / or the portion of the DNA protein or peptides, according to claims 1 to 4.
7. The vaccine according to claim 6, characterized in that Tat has the following nucleotide sequence (Sec. 1): 5 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTC AGCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCTTTCATTG CC-AGTTTGTTTC? TAAC-AAAAGCCTTAGGCATCTCCTATGGCAGGAAGAA GCGGAGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTT CTCTATCAAAGCAGCCCACCTCCCAATCCCGAGGGGACCCGACAGGCCC GAAGGAATAG_3_'and any other Tat variant of any type and subtype of HIV.
8. The vaccine according to claim 6, characterized in that Tat has the following amino acid sequence: NH2-MEPVDPRLEPWKHPGSQPKTACTNCYCKKCCFHCQVCFITKALG ISYGRKKRRQRRRPPQGSQTHQVSLSKQPTSQSRGDPTGPKE-COOH and any other Tat variant of any type and subtype of HIV.
9. The vaccine in accordance with the claim 6, wherein the mutants are selected from those having the following nucleotide sequences, or parts thereof: Nucleotide cys22 mutant (Seq. 2) 5 'TGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCGGTACCAATTGCTATTGTAAAAAGTGTTGCTTTCATTGCC CAAGTTTGTTTCATAACAAAAGCCTTAGGCATCTCCTATGGCAGGAAGAA GCGGAGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGT TTCTCTATCAAAGCAGCCCACCTCCCAATCCCGAGGGGACCCGACAGGCC CGAAGGAATAG_3_' Nucleotide sequence lys41 (Sec. 3) 5 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCMCATTGCCA AGTTTGTTTCATAACAAACGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTTCTCTAT CAAAGCAGCCCACCTCCCAATCCCGAGGGGACCCGACAGGCCCGAAGGAAT AG 3 ' Nucleotide sequence of the RGD mutant? (Sec. 4) 5 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCTCATTGCCA AGTTTGTTTCATAACAAAAGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCT (- ^ AGGCAGTCAGACTCATCAAGTTTCTCTAT CAAAGCAGCCCACCTCCCAATCCCCGACAGGCCCGAAGGAATAG_3_' Nucleotide sequence of mutant Iys41-RGD (Sec 5)?.AGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCMCATTGCCA AGTTTGTTTCATAACAAACGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTTCTCTAT CAAAGCAGCCCACCTCCCAATCCCCGACAGGCCCGAAGGAATAG_3_'
10. The vaccine according to claim 6, characterized in that the mutants are selected from those having the following amino acid sequence or part thereof: Amino acid sequence of the cys22 mutant NH2 -MEPVDPRLEPWSQPKTACTNCYCKKCCFHCQVCFITKA LGISYGRKKRRORRRPPQGSQTHQVSLSKQPTSOSPTGPKE-COOH Amino acid sequence of lys41 NH2-MEPVDPRLEPWSQPKTACTNCYCKKCCFHCQVCFITTAL GISYGRKKRRORRRPPQGSQTHQVSLSKQPTSQSRGDPTGPKE-COOH Nucleotide sequence of the RGD mutant? NH2 -MEPVDPRLEPWSQPKTACTNCYCKKCCFHCQVCFITKALG ISYGRKKRRORRRPPQGSQTHQVSLSKQPTSQSPTGPKE-COOH Nucleotide sequence of the mutant Iys41-RGD? NH2 -MEPVDPRLEPWSQPKTACTNCYCKKCCFHCQVCFITTALG ISYGRKKRRORRRPPQGSQTHQVSLSKQPTSOSPTGPKE-COOH
11. Vaccine according to claim 6, characterized in that the Tat portions are selected from the Pep peptide sequences. 1. MEPVDPRLEPWSQPKT Pep. 2. ACTNCYCKKCCFHCQVCFIT Pep. 3. QVCFITKALGISYGRK Pep. 4. SYGRKKRRQRRRPPQ Pep. 5. RPPQGSQTHQVSLSKQ Pep. 6. HQVSLSKQPTSQSRGD Pep. 7. PTSQSRGDPTGPKE
12. The vaccine according to claims 6 to 11, characterized in that it comprises proteins or peptides conjugated with the universal T epitope auxiliary of tetanus toxoid or any auxiliary T peptide.
13. The vaccine according to claims 6 to 12, characterized in that it is in combination with recombinant proteins or HIV peptides Nef, Rev or Gag, or part thereof.
14. The vaccine according to claim 6, characterized in that it comprises the Tat fusion proteins. (wild type) or its mutants) / Nef, Tat (wild type or its mutants) / Rev, Tat (wild type or its mutants) / Gag or parts thereof.
15. The vaccine in accordance with the claim 6-14, in combination with recombinant inmurTomodulatory cytokines or other molecules, or parts thereof, that increase the antiviral immune response.
16. The vaccine in accordance with the claim 15, characterized in that the cytokines are IL-12 and / or IL-15 or IFNa or IFNβ.
17. The vaccine according to claim 6, characterized in that it comprises Tat fusion proteins (wild type or mutants thereof) / immunomodulating cytokines, Tat (wild type or mutants thereof) / IL-12, Tat (wild type or its mutants) / IL -15, Tat (wild type or its mutants) / other molecules or part thereof, which increase the antiviral immune response.
18. The DNA vaccine, according to claims 6, 7 and 9, characterized in that it comprises DNA encoding wild type Tat or its mutants, or parts thereof, inserted into expression vectors.
19. The DNA vaccine, according to claims 6, 7 and 9, in combination with an expression vector that includes the HIV rev, nef and gag genes, or part thereof.
20. The DNA vaccine, according to claims 18 or 19, characterized in that the vector is a Tat co-expressing plasmid (wild type or mutants thereof) / Rev, Tat (wild type or mutants thereof) / Nef, Tat (wild type or its mutants) / Gag or part of them.
21. The DNA vaccine according to claims 6, 7 and 9, in combination with DNA molecules inserted into expression vectors encoding immunomodulatory cytokines or other immunomodulatory molecules, or parts thereof, which increase the antiviral immune response.
22. The DNA vaccine, according to claim 21, characterized in that the cytokine is IL-12 and / or IL-15.
23. The DNA vaccine, according to claims 21 or 22, characterized in that the vector is a plasmid coexpressing Tat (wild type or its mutants) / IL-12, Tat (wild type or mutants thereof) / IL-15, Tat (wild type or its mutants) / other molecules or parts thereof, capable of increasing the antiviral immune response.conformance vaccine * with claims 18 to 23, characterized in that the vector is pCVO.
25. The vaccine according to claims 6-24, characterized in that it includes autologous dendritic cells treated and / or untreated, according to the preceding claims.
26. The vaccine according to claims 6 to 25, characterized in that it includes adjuvants capable of increasing the antiviral immune response.
27. The vaccine according to claim 26, characterized in that the adjuvant is selected from alumina, ISCOM, RIBI and corresponding mixtures.
28. The vaccine according to claims 6 to 27, characterized in that it comprises systems for delivery.
29. The vaccine according to claim 28, characterized in that the systems for delivery are selected from among nanoparticles, herpes vectors, red cells, bacteria and combinations thereof.
30. The vaccine according to claim 29, characterized in that the bacteria are selected from Streptococcus gordonii and Lactobacillus.
31. The vaccine according to claims 29 and 30, characterized in that the bacterium is modified to express viral antigens.
32. The vaccine according to claims 6 to 31, characterized in that it is used for the immunization of peripheral blood cells of infected individuals, expanded by co-stimulation with magnetic spheres coated with antibodies against CD3 and against CD28.
33. The therapeutic vaccine, according to claims 6 to 32, characterized in that it is combined with inhibitors of viral replication.
34. The vaccine in accordance with the claim 6 to 33, characterized in that the active principle is delivered to the mucosa.
35. The vaccine according to claim 34, characterized in that the active principle is administered nasally, orally, vaginally and / or rectally.
36. The vaccine according to claims 6 to 33, characterized in that the active principle is administered through a systemic or local route.
37. The vaccine according to claim 36, characterized in that the active principle is administered through an intramuscular, subcutaneous or intradermal route.
38. The vaccine according to claim 37, characterized in that the active ingredient is administered intradermally in amounts of 1-6 μg, without adjuvants.
39. The vaccine according to claims 34 to 38, characterized in that the active principle is transported in a biologically acceptable fluid.
40. The vaccine according to claims 34 to 39, characterized in that it also comprises pharmaceutically acceptable carriers and excipients to maximize the activity of the principle.
41. The vaccine according to claims 34-40, characterized in that it comprises as an active ingredient Tat, according to claim 1, in preventive and / or therapeutic amounts.
42. Tat mutant protein, biologically active, characterized because it has the nucleotide sequence that is selected from: 5 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCGGTACCAATTGCTATTGTAAAAAGTGTTGCTTTCATTGCC CAAGTTTGTTTCATAACAAAAGCCTTAGGCATCTCCTATGGCAGGAAGAA GCGGAGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGT TTCTCTATCAAAGCAGCCCACCTCCCAATCCCGAGGGGACCCGACAGGCC CGAAGGAATAG_3_' Nucleotide sequence 'lys41 (3 Sec.) - 2Í9 -5' ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCMCATTGCCA AGTTTGTTTCATAACAAACGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTTCTCTAT CAAAGCAGCCCACCTCCCAATCCCGAGGGGACCCGACAGGCCCGAAGGAAT AG 3' Nucleotide sequence of the RGD mutant? (Sec. 4) 5 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCTCATTGCCA AGTTTGTTTCATAACAAAAGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTTCTCTAT CAAAGCAGCCCACCTCCCAATCCCCGACAGGCCCGAAGGAATAG_3_'sequence of the mutant Iys41-RGD? (Sec. 5) 5 'ATGGAGCCAGTAGATCCTAGACTAGAGCCCTGGAAGCATCCAGGAAGTCA GCCTAAAACTGCTTGTACCAATTGCTATTGTAAAAAGTGTTGCMCATTGCCA AGTTTGTTTCATAACAAACGCCTTAGGCATCTCCTATGGCAGGAAGAAGCGG AGACAGCGACGAAGACCTCCTCAAGGCAGTCAGACTCATCAAGTTTCTCTAT CAAAGCAGCCCACCTCCCAATCCCCGACAGGCCCGAAGGAATAG_3_'
43. Amino acid sequence of biologically active Tat mutants, characterized in that they are selected from: Amino acid sequence of the cys22 mutant NH2-MEPVDPRLEPWKHPGSQPKTACTNCYCKKCCFHCQVCFITKA LGISYGRKKRRORRRPPQGSQTHQVSLSKQPTSOSPTGPKE-COOH Amino acid sequence of the lys41 mutant NH2-MEPVDPRLEPWKHPGSQPKTACTNCYCKKCCFHCQVCFITTAL GISYGRKKRRORRRPPQGSQTHQVSLSKQPTSQSRGDPTGPKE-COOH Amino acid sequence of the RGD mutant? NH2 -MEPVDPRLEPWKHPGSQPKTACTNCYCKKCCFHCQVCFITKALG ISYGRKKRRORRRPPQGSQTHQVSLSKQPTSQSPTGPKE-COOH Amino acid sequence of the mutant Iys41-RGD? NH2-MEPVDPRLEPWKHPGSQPKTACTNCYCKKCCFHCQVCFITTALG. ISYGRKKRRORRRPPQGSQTHQVSLSKQPTSOSPTGPKE-COOH
44. Biologically active Tat mutants, characterized by having the peptide sequence selected from: Pep. 1. MEPVDPRLEPWKHPGSQPKT Pep. 2. ACTNCYCKKCCFHCQVCFIT Pep. 3. QVCFITKALGISYGRK Pep. 4. SYGRKKRRQRRRPPQ Pep. 5. RPPQGSQTHQVSLSKQ Pep. 6. HQVSLSKQPTSQSRGD Pep. 7. PTSQSRGDPTGPKE
45. Expression vector, characterized in that it comprises a DNA sequence that is selected from those included in accordance with claim 2, or part thereof.
46. PCVO expression vector, characterized in that it includes a DNA sequence that is selected from among those included in accordance with claim 42, or part thereof.
47. Expression vector pCVO, according to claim 46, characterized in that it comprises a DNA sequence that codes for a gene that is selected from tat, rev, nef, gag, IL-12, IL-15 and combinations thereof. •
48. Transformed cells, characterized in that they include the vector according to claims 45 to 47.
49. The use of the vector pCVO, characterized in that it is modified according to claims 46 to 47, to prepare a preventive and / or therapeutic vaccine against AIDS, tumors associated with AIDS, syndromes and symptoms associated with HIV infection.
50. Dendritic cells characterized in that they are treated with Tat protein or its peptides or mutants, according to claims 43 and 44, or combinations with Rev, Nef and Gag proteins, and / or cytokines.
51. Dendritic cells, according to claim 49, characterized in that they are transduced with an expression vector comprising the tat gene.
52. The use of Tat in its non-oxidized and non-aggregated form, to prepare a vaccine, according to claims 6-41.
53. The use of lyophilized Tat, to prepare a vaccine according to claims 6-41, lyophilized Tat is resuspended in a biologically acceptable fluid for administration.
54. The use of Tat protein, according to claims 1 to 5, to prepare a protein or peptide or DNA vaccine, preventive and / or therapeutic against AIDS, tumors associated with AIDS, syndromes and symptoms associated with HIV infection.
55. The use of Alum, ISCOM, RIBI and other adjuvants, alone or in combination, to prepare a vaccine according to claim 6.
56. The use of paramagnetic spheres coated with monoclonal antibodies against CD3 and against CD8, to make a vaccine, according to claim 6.
MXPA/A/2000/005325A 1997-12-01 2000-05-30 Hiv-1 tat, or derivatives thereof for prophylactic and therapeutic vaccination MXPA00005325A (en)

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
RMRM97A000743 1997-12-01

Publications (1)

Publication Number Publication Date
MXPA00005325A true MXPA00005325A (en) 2001-12-13

Family

ID=

Similar Documents

Publication Publication Date Title
EP1035865B1 (en) Hiv-1 tat, or derivatives thereof for prophylactic and therapeutic vaccination
Paliard et al. Priming of strong, broad, and long-lived HIV type 1 p55gag-specific CD8+ cytotoxic T cells after administration of a virus-like particle vaccine in rhesus macaques
EP1425035B1 (en) Use of biologically active hiv-1 tat, fragments or derivatives thereof, to target and/or to activate antigen-presenting cells, and/or to deliver cargo molecules for preventive or therapeutic vaccination and/or to treat other diseases
Johnson et al. Induction of vigorous cytotoxic T-lymphocyte responses by live attenuated simian immunodeficiency virus
EP0882134B1 (en) Methods and compositions for protective and therapeutic genetic immunization
Cui et al. Strong T cell type-1 immune responses to HIV-1 Tat (1–72) protein-coated nanoparticles
KR20140122735A (en) Immunogens for hiv vaccination
Caputo et al. Recent advances in the development of HIV-1 Tat-based vaccines
Notka et al. Accelerated clearance of SHIV in rhesus monkeys by virus-like particle vaccines is dependent on induction of neutralizing antibodies
Hulskotte et al. Vaccine-induced virus-neutralizing antibodies and cytotoxic T cells do not protect macaques from experimental infection with simian immunodeficiency virus SIVmac32H (J5)
KR101814857B1 (en) Pharmaceutical compositions for preventing and/or treating an hiv disease in humans
Heeney et al. HIV-1 vaccine-induced immune responses which correlate with protection from SHIV infection: compiled preclinical efficacy data from trials with ten different HIV-1 vaccine candidates
Leung et al. The kinetics of specific immune responses in rhesus monkeys inoculated with live recombinant BCG expressing SIV Gag, Pol, Env, and Nef proteins
Tohidi et al. Induction of a robust humoral response using HIV-1 VLPMPER-V3 as a novel candidate vaccine in BALB/C mice
Mahdavi et al. HIV-1 Gag p24-Nef fusion peptide induces cellular and humoral immune response in a mouse model
CA2722240C (en) Tat protein for preventing or treating aids
US20010004531A1 (en) AIDS DNA vaccine that prevents SIVmac239 virus infection in monkeys
MXPA00005325A (en) Hiv-1 tat, or derivatives thereof for prophylactic and therapeutic vaccination
Poteet et al. SHIV Virus-like particles (VLPs) Vaccination Induces Partial Protection from SHIV Challenge in a Rhesus Macaque Model
Protect Highly Attenuated Vaccine Strains of
WO2003082908A2 (en) Use of the p17 protein of hiv in isolated form for the preparation of a medicament for administration to hiv seropositive patients, as well as pharmaceutical compositions comprising said protein
Reduce Multispecific Vaccine-Induced Mucosal
Mumper et al. Compositions Comprising Human Immunodeficiency Virus Tat Adsorbed to the Surface of Anionic Nanoparticles
SIVmac239 Tat-Vaccinated Macaques Do Not Control
AU1971201A (en) Methods and compositions for protective and therapeutic genetic immunization