Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
PLoS One ; 17(11): e0276751, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36355808

RESUMEN

Despite new antivirals are being approved against SARS-CoV-2 they suffer from significant constraints and are not indicated for hospitalized patients, who are left with few antiviral options. Repurposed drugs have previously shown controversial clinical results and it remains difficult to understand why certain trials delivered positive results and other trials failed. Our manuscript contributes to explaining the puzzle: this might have been caused by a suboptimal drug exposure and, consequently, an incomplete virus suppression, also because the drugs have mostly been used as add-on monotherapies. As with other viruses (e.g., HIV and HCV) identifying synergistic combinations among such drugs could overcome monotherapy-related limitations. In a cell culture model for SARS-CoV-2 infection the following stringent criteria were adopted to assess drug combinations: 1) identify robust, synergistic antiviral activity with no increase in cytotoxicity, 2) identify the lowest drug concentration inhibiting the virus by 100% (LIC100) and 3) understand whether the LIC100 could be reached in the lung at clinically indicated drug doses. Among several combinations tested, remdesivir with either azithromycin or ivermectin synergistically increased the antiviral activity with no increase in cytotoxicity, improving the therapeutic index and lowering the LIC100 of every one of the drugs to levels that are expected to be achievable and maintained in the lung for a therapeutically relevant period of time. These results are consistent with recent clinical observations showing that intensive care unit admission was significantly delayed by the combination of AZI and RDV, but not by RDV alone, and could have immediate implications for the treatment of hospitalized patients with COVID-19 as the proposed "drug cocktails" should have antiviral activity against present and future SARS-CoV-2 variants without significant overlapping toxicity, while minimizing the onset of drug resistance. Our results also provide a validated methodology to help sort out which combination of drugs are most likely to be efficacious in vivo, based on their in vitro activity, potential synergy and PK profiles.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , SARS-CoV-2 , Humanos , Antivirales/farmacología , Antivirales/uso terapéutico , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Alanina/farmacología , Alanina/uso terapéutico , Pulmón , Combinación de Medicamentos
2.
PLoS One ; 9(11): e112568, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25392925

RESUMEN

The mosquito-borne West Nile virus (WNV) causes human and animal disease with outbreaks in several parts of the world including North America, the Mediterranean countries, Central and East Europe, the Middle East, and Africa. Particularly in elderly people and individuals with an impaired immune system, infection with WNV can progress into a serious neuroinvasive disease. Currently, no treatment or vaccine is available to protect humans against infection or disease. The goal of this study was to develop a WNV-vaccine that is safe to use in these high-risk human target populations. We performed a vaccine efficacy study in non-human primates using the contemporary, pathogenic European WNV genotype 1a challenge strain, WNV-Ita09. Two vaccine strategies were evaluated in rhesus macaques (Macaca mulatta) using recombinant soluble WNV envelope (E) ectodomain adjuvanted with Matrix-M, either with or without DNA priming. The DNA priming immunization was performed with WNV-DermaVir nanoparticles. Both vaccination strategies successfully induced humoral and cellular immune responses that completely protected the macaques against the development of viremia. In addition, the vaccine was well tolerated by all animals. Overall, The WNV E protein adjuvanted with Matrix-M is a promising vaccine candidate for a non-infectious WNV vaccine for use in humans, including at-risk populations.


Asunto(s)
Fiebre del Nilo Occidental/prevención & control , Vacunas contra el Virus del Nilo Occidental/uso terapéutico , Virus del Nilo Occidental/clasificación , Animales , Anticuerpos Antivirales/sangre , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Culicidae , Europa (Continente) , Inmunidad Celular , Inmunidad Humoral , Inyecciones Intradérmicas , Interferón gamma/inmunología , Macaca mulatta , Estructura Terciaria de Proteína , Proteínas del Envoltorio Viral/inmunología , Carga Viral , Viremia/inmunología
3.
Soft Matter ; 10(38): 7653-60, 2014 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-25137385

RESUMEN

Mannobiose-modified polyethylenimines (PEI) are used in gene therapy to generate nanoparticles of DNA that can be targeted to the antigen-presenting cells of the immune system. We report that the sugar modification alters the DNA organization within the nanoparticles from homogenous to shell-like packing. The depth-dependent packing of DNA within the nanoparticles was probed using AFM nano-indentation. Unmodified PEI-DNA nanoparticles display linear elastic properties and depth-independent mechanics, characteristic of homogenous materials. Mannobiose-modified nanoparticles, however, showed distinct force regimes that were dependent on indentation depth, with 'buckling'-like response that is reproducible and not due to particle failure. By comparison with theoretical studies of spherical shell mechanics, the structure of mannobiosylated particles was deduced to be a thin shell with wall thickness in the order of few nanometers, and a fluid-filled core. The shell-core structure is also consistent with observations of nanoparticle denting in altered solution conditions, with measurements of nanoparticle water content from AFM images, and with images of DNA distribution in Transmission Electron Microscopy.


Asunto(s)
ADN/química , Mananos/química , Nanopartículas/química , Nanopartículas/ultraestructura , Polietileneimina/química , Microscopía de Fuerza Atómica , Microscopía Electrónica de Transmisión
4.
PLoS One ; 9(2): e87837, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24503579

RESUMEN

West Nile virus (WNV) is a mosquito-borne flavivirus that is endemic in Africa, the Middle East, Europe and the United States. There is currently no antiviral treatment or human vaccine available to treat or prevent WNV infection. DNA plasmid-based vaccines represent a new approach for controlling infectious diseases. In rodents, DNA vaccines have been shown to induce B cell and cytotoxic T cell responses and protect against a wide range of infections. In this study, we formulated a plasmid DNA vector expressing the ectodomain of the E-protein of WNV into nanoparticles by using linear polyethyleneimine (lPEI) covalently bound to mannose and examined the potential of this vaccine to protect against lethal WNV infection in mice. Mice were immunized twice (prime--boost regime) with the WNV DNA vaccine formulated with lPEI-mannose using different administration routes (intramuscular, intradermal and topical). In parallel a heterologous boost with purified recombinant WNV envelope (E) protein was evaluated. While no significant E-protein specific humoral response was generated after DNA immunization, protein boosting of DNA-primed mice resulted in a marked increase in total neutralizing antibody titer. In addition, E-specific IL-4 T-cell immune responses were detected by ELISPOT after protein boost and CD8(+) specific IFN-γ expression was observed by flow cytometry. Challenge experiments using the heterologous immunization regime revealed protective immunity to homologous and virulent WNV infection.


Asunto(s)
Inmunidad Celular , Vacunas de ADN , Proteínas del Envoltorio Viral/inmunología , Fiebre del Nilo Occidental/inmunología , Fiebre del Nilo Occidental/prevención & control , Vacunas contra el Virus del Nilo Occidental/inmunología , Virus del Nilo Occidental/inmunología , Animales , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Especificidad de Anticuerpos/inmunología , Femenino , Inmunización , Inmunización Secundaria , Ratones , Nanopartículas/química , Células Th2/inmunología , Proteínas del Envoltorio Viral/genética , Fiebre del Nilo Occidental/mortalidad , Vacunas contra el Virus del Nilo Occidental/efectos adversos , Virus del Nilo Occidental/genética
5.
J Acquir Immune Defic Syndr ; 64(4): 351-9, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-24169120

RESUMEN

BACKGROUND: HIV-specific cellular immune responses are associated with control of viremia and delayed disease progression. An effective therapeutic vaccine could mimic these effects and reduce the need for continued antiretroviral therapy. DermaVir, a topically administered plasmid DNA-nanomedicine expressing HIV (CladeB) virus-like particles consisting of 15 antigens, induces predominantly central memory T-cell responses. METHODS: Treated HIV-infected adults (HIV RNA <50 and CD4 >350) were randomized to placebo or escalating DermaVir doses (0.1 or 0.4 mg of plasmid DNA at weeks 1, 7, and 13 in the low- and intermediate-dose groups and 0.8 mg at weeks 0, 1, 6, 7, 12, and 13 in the high-dose group), n = 5-6 evaluable subjects per group. Immunogenicity was assessed by a 12-day cultured interferon-γ enzyme-linked immunosorbent spot assay at baseline and at weeks 9, 17, and 37 using 1 Tat/Rev and 3 overlapping Gag peptide pools (p17, p24, and p15). RESULTS: Groups were comparable at baseline. The study intervention was well tolerated, without dose-limiting toxicities. Most responses were highest at week 17 (4 weeks after last vaccination) when Gag p24 responses were significantly greater among intermediate-dose group compared with control subjects [median (IQR): 67,600 (5633-74,368) versus 1194 (9-1667)] net spot-forming units per million cells, P = 0.032. In the intermediate-dose group, there was also a marginal Gag p15 response increase from baseline to week 17 [2859 (1867-56,933), P = 0.06], and this change was significantly greater than in the placebo group [0 (-713 to 297), P = 0.016]. CONCLUSIONS: DermaVir administration was associated with a trend toward greater HIV-specific, predominantly central memory T-cell responses. The intermediate DermaVir dose tended to show the greatest immunogenicity, consistent with previous studies in different HIV-infected patient populations.


Asunto(s)
Vacunas contra el SIDA/inmunología , Fármacos Anti-VIH/administración & dosificación , Fármacos Anti-VIH/uso terapéutico , Vacunas contra el SIDA/administración & dosificación , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Quimioterapia Combinada , Humanos , Esquemas de Inmunización , ARN Viral , Carga Viral , Viremia
6.
Immunotherapy ; 5(8): 825-8, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23902551

RESUMEN

Evaluation of: Pleguezuelos O, Stoloff GA, Caparros-Wanderley W. Synthetic immunotherapy induces HIV virus specific Th1 cytotoxic response and death of an HIV-1 infected human cell line through classic complement activation. Virol. J. 10(1), 107 (2013). AIDS vaccine development represents an unprecedented challenge in both immunogen design and delivery to induce potent and long-lasting HIV-specific immune responses, including neutralizing antibodies and cytotoxic T lymphocytes (CTL). Pleguezuelos and coworkers recognized that immunogen design must address both HIV and HLA diversity to make a global vaccine. The HIV-v synthetic polypeptide vaccine described here sets a new standard in antigen design by selecting conserved regions of global HIV-1 and HIV-2 isolates and epitopes from most frequent HLA types of the human population. The new vaccine induced both antibody and CTL responses. Importantly, the authors demonstrated vaccine-specific HLA restricted CD8(+) CTL responses for one HLA allele that was involved in the antigen design. HIV-v vaccine is a new candidate to be tested in human subjects carrying frequent HLA types.


Asunto(s)
Muerte Celular , Activación de Complemento , Epítopos de Linfocito T/inmunología , VIH-1/inmunología , Inmunoterapia/métodos , Células TH1/inmunología , Proteínas Virales/inmunología , Animales , Femenino , Humanos , Masculino
7.
Nanomedicine ; 9(8): 1245-54, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23747740

RESUMEN

Allergen-specific immunotherapy (ASIT) the only disease-modifying treatment for IgE-mediated allergies is characterized with long treatment duration and high risk of side effects. We investigated the safety, immunogenicity and efficacy of a novel ASIT, called DermAll, in an experimental allergic rhinitis model. We designed and characterized DermAll-OVA, a synthetic plasmid pDNA/PEIm nanomedicine expressing ovalbumin (OVA) as model allergen. DermAll-OVA was administered topically with DermaPrep device to target Langerhans cells. To detect the clinical efficacy of DermAll ASIT we quantified the nasal symptoms and characterized the immunomodulatory activity of DermAll ASIT by measuring cytokine secretion after OVA-stimulation of splenocytes and antibodies from the sera. In allergic mice DermAll ASIT was as safe as Placebo, balanced the allergen-induced pathogenic TH2-polarized immune responses, and decreased the clinical symptoms by 52% [32%, 70%] compared to Placebo. These studies suggest that DermAll ASIT is safe and should significantly improve the immunopathology and symptoms of allergic diseases. FROM THE CLINICAL EDITOR: A novel allergen-specific immunotherapy for IgE-mediated allergies is presented in this paper, using an experimental allergic rhinitis model and a synthetic plasmid pDNA/PEIm nanomedicine expressing ovalbumin as model allergen. Over 50% reduction of symptoms was found as the immune system's balance was favorably altered toward more TH2-polarized immune responses.


Asunto(s)
Alérgenos/inmunología , Ovalbúmina/inmunología , Rinitis Alérgica Perenne/inmunología , Rinitis Alérgica Perenne/prevención & control , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico , Administración Tópica , Alérgenos/genética , Animales , Citocinas/inmunología , Femenino , Inmunización , Inmunoglobulina E/sangre , Inmunoglobulina E/inmunología , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Ratones , Ratones Endogámicos BALB C , Nanomedicina , Ovalbúmina/genética , Plásmidos/administración & dosificación , Plásmidos/genética , Plásmidos/inmunología , Plásmidos/uso terapéutico , Rinitis Alérgica , Rinitis Alérgica Perenne/sangre , Células Th2/inmunología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
8.
Nanomedicine ; 9(1): 28-38, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22659241

RESUMEN

Combination antiretroviral therapy (cART) successfully suppresses HIV replication. However, daily and lifelong treatment is necessary to manage patient illness because cART neither eradicates infected cells from reservoirs nor reconstitutes HIV-specific immunity that could kill infected cells. Toward the cure of HIV, different nanomedicine classes have been developed with the following disease-modifying properties: to eradicate the virus by activation of latently infected CD4+ T-cells and reservoirs flushing; to kill the infected cells in the reservoirs by boosting of HIV-specific T cells; and to prevent infection by the use of microbicides with improved epithelial penetration and drug half-life. Preclinical and clinical trials consistently demonstrated that DermaVir, the most advanced nanomedicine, induces long-lasting memory T-cell responses and reduces viral load in comparison with placebo. DermaVir and the nanomedicine pipelines have the potential to improve the health of HIV-infected people at lower costs, to decrease antiretroviral drug exposure, and to contribute to the cure of HIV/AIDS. FROM THE CLINICAL EDITOR: Despite the leaps and bounds in the development of antiretroviral therapy, HIV remains a significant public health challenge. In this review, applications of nanomedicine- based technologies are discussed in the context of HIV treatment, including virus elimination by activation of latently infected CD4+ T-cells; infected cell elimination in the reservoirs by boosting HIV-specific T cells, and by preventing infection by the use of microbicides with improved epithelial penetration and drug half-life.


Asunto(s)
Fármacos Anti-VIH/uso terapéutico , Infecciones por VIH/tratamiento farmacológico , Nanomedicina , Fármacos Anti-VIH/administración & dosificación , Linfocitos T CD4-Positivos/virología , Química Farmacéutica , VIH/inmunología , VIH/aislamiento & purificación , Humanos
9.
Actual. SIDA ; 20(76): 38-47, jun. 2012. ilus
Artículo en Español | LILACS | ID: lil-654861

RESUMEN

Hallazgos recientes en el área de la intensificación de los tratamientos antirretrovirales demuestran que la forma de llegar a una curación para el VIH exige la inclusión de nuevas estrategias de tratamiento como las vacunas terapéuticas. La vacuna terapéutica DermaVir incluye elementos tecnológicos clave en el diseño de una vacuna racional: un único inmunógeno plásmido de ADN (pADN) que expresa a 15 antígenos del VIH, formulación de nanomedicina y una única administración tópica de la vacuna enfocada a las células dendríticas. Luego de su administración tópica sobre la piel preparada, las células epidérmicas de Langerhans activadas transportan la nanomedicina DermaVir a los nódulos linfáticos para expresar a los antígenos del VIH codificados como pADN e inducir a las células T precursoras/de memoria con una alta capacidad de proliferación. Se han demostrdo la seguridad, la inmunogenicidad y la eficacia preliminar de la DermaVir en varios modelos animales y en humanos con infección por VIH. Esta novedosa tecnología de vacunación terapéutica podría constituir un nuevo paradigma en el trtamiento contra el VIH.


Recent findings on the field of antiretroviral treatment intesnsification demonstrate thar the way towards a cure for HIV requires the involvement of novel treatment strategies like therapeutic vaccines. DermaVir therapeutic vaccine includes key technological elements of rational vaccine design: a single plasmid DNA (pDNA) immunogen that expresses 15 HIV antigens, nanomedicine formulation and a unique dendritic cell-targeting topical vaccine administration. Following topical administration on the prepared skin, DermaVir nanomedicine is transported by activated epidermal Langerhans cells to the lymph nodes to express the pDNA-encoded HIV antigens and induce precursor/memory T cells with high proliferation capacity. Safety, immunogenicity and preliminary efficacy of DermaVir have been demonstrated in several animal models and HIV-infected human subjects. This novel therapeutic vaccination technology might offer a new treatment paradigm against HIV.


Asunto(s)
Humanos , Vacunas contra el SIDA , Antígenos VIH/inmunología , VIH , Nanomedicina , Resultado del Tratamiento
10.
PLoS One ; 7(5): e35416, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22590502

RESUMEN

BACKGROUND: The GIHU004 study was designed to evaluate the safety and immunogenicity of three doses of DermaVir immunization in HIV-infected subjects on fully suppressive combination antiretroviral therapy (cART). METHODOLOGY/PRINCIPAL FINDINGS: This first-in-human dose escalation study was conducted with three topical DermaVir doses targeted to epidermal Langerhans cells to express fifteen HIV antigens in draining lymph nodes: 0.1 mg DNA targeted to two, 0.4 mg and 0.8 mg DNA targeted to four lymph nodes. Particularly, in the medium dose cohort 0.1 mg DNA was targeted per draining lymph node via ∼8 million Langerhans cells located in 80 cm(2) epidermis area. The 28-days study with 48-week safety follow-up evaluated HIV-specific T cell responses against Gag p17, Gag p24 and Gag p15, Tat and Rev antigens. DermaVir-associated side effects were mild, transient and not dose-dependent. Boosting of HIV-specific effector CD4(+) and CD8(+) T cells expressing IFN-gamma and IL-2 was detected against several antigens in every subject of the medium dose cohort. The striking result was the dose-dependent expansion of HIV-specific precursor/memory T cells with high proliferation capacity. In low, medium and high dose cohorts this HIV-specific T cell population increased by 325-, 136,202 and 50,759 counts after 4 weeks, and by 3,899, 9,878 and 18,382 counts after one year, respectively, compared to baseline. CONCLUSIONS/SIGNIFICANCE: Single immunization with the DermaVir candidate therapeutic vaccine was safe and immunogenic in HIV-infected individuals. Based on the potent induction of Gag, Tat and Rev-specific memory T cells, especially in the medium dose cohort, we speculate that DermaVir boost T cell responses specific to all the 15 HIV antigens expressed from the single DNA. For durable immune reactivity repeated DermaVir immunization might be required since the frequency of DermaVir-boosted HIV-specific memory T cells decreased during the 48-week follow up. TRIAL REGISTRATION: ClinicalTrial.gov NCT00712530.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Infecciones por VIH/inmunología , VIH-1/inmunología , Memoria Inmunológica , Vacunas contra el SIDA/inmunología , Adolescente , Adulto , Antígenos Virales/inmunología , Estudios de Cohortes , Relación Dosis-Respuesta Inmunológica , Epidermis/inmunología , Femenino , Estudios de Seguimiento , Infecciones por VIH/terapia , Humanos , Interferón gamma/inmunología , Células de Langerhans/inmunología , Ganglios Linfáticos/inmunología , Masculino , Persona de Mediana Edad
11.
Nanomedicine ; 8(4): 497-506, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21839051

RESUMEN

Here we characterize the structure, stability and intracellular mode of action of DermaVir nanomedicine that is under clinical development for the treatment of HIV/AIDS. This nanomedicine comprises pathogen-like pDNA/PEIm nanoparticles (NPs) having the structure and function resembling spherical viruses that naturally evolved to deliver nucleic acids to the cells. Atomic force microscopy demonstrated spherical 100 - 200 nm NPs with a smooth polymer surface protecting the pDNA in the core. Optical absorption determined both the NP structural stability and biological activity relevant to their ability to escape from the endosome and release the pDNA at the nucleus. Salt, pH and temperature influence nanomedicine shelf-life and intracellular stability. This approach facilitates the development of diverse polyplex nanomedicines where the delivered pDNA-expressed antigens induce immune responses to kill infected cells. FROM THE CLINICAL EDITOR: The authors investigated DermaVir nanomedicine comprised of pathogen-like pDNA/PEIm nanoparticles with structure and function resembling spherical viruses. DermaVir delivery of pDNA expresses antigens that induce immune responses to kill HIV infected cells.


Asunto(s)
Vacunas contra el SIDA , Síndrome de Inmunodeficiencia Adquirida/tratamiento farmacológico , Núcleo Celular , ADN , VIH-1 , Nanopartículas , Transfección/métodos , Vacunas contra el SIDA/química , Vacunas contra el SIDA/farmacología , Línea Celular , ADN/química , ADN/farmacología , Humanos , Nanopartículas/química , Nanopartículas/ultraestructura , Tamaño de la Partícula
12.
Vaccine ; 29(4): 744-53, 2011 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-21109034

RESUMEN

We describe here a single plasmid DNA immunogen representing the broadest antigen repertoire among HIV vaccine candidates. This pDNA was "ANTIGENeered" for the regulated expression of thirteen complete and two non-functional HIV protein antigens. These proteins self assemble into complex virus-like particles (VLP(+)). Multiple irreversible safety features were introduced by genetic modifications including the complete impairment of integration, reverse transcription, the function of Nef and the 3'LTR. Epitope analysis predicted that the pDNA-derived protein repertoire can potentially present over 3000 T cell epitopes. However, the expressed antigens have different potential to induce HIV-specific CD4(+) and CD8(+) T cells supporting our hypothesis that HIV vaccines should contain all possible regulatory and structural proteins. This immunogen is the active pharmaceutical ingredient of DermaVir, a therapeutic vaccine product candidate that recently successfully completed Phase II clinical trials and meets the safety, immunogenicity and cost requirements of an HIV vaccine.


Asunto(s)
Vacunas contra el SIDA/inmunología , Antígenos VIH/inmunología , Vacunas de ADN/inmunología , Vacunas contra el SIDA/genética , Ensayos Clínicos como Asunto , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Antígenos VIH/genética , Humanos , Datos de Secuencia Molecular , Plásmidos , Análisis de Secuencia de ADN , Vacunas de ADN/genética , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Vacunas de Virosoma/genética , Vacunas de Virosoma/inmunología
13.
Int J Pharm ; 392(1-2): 261-7, 2010 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-20347027

RESUMEN

DermaVir vaccine is a novel "pathogen-like" nanomedicine containing a plasmid DNA complexed with a polyethylenimine that is mannobiosylated to target antigen-presenting cells and to induce immune responses (pDNA/PEIm). To develop a commercially viable vaccine product we have systematically investigated the variability of raw materials and their relationship with the product's biological activity. We demonstrated that the cGMP quality requirements are not sufficient to reproducible formulate the nanomedicine with optimal biological activity. Unexpectedly, we found that the high cationic concentration of the pDNA favored the biological activity, but did not support the stability of the nanomedicine. Similarly, the presence of EDTA in the pDNA increased the size of the nanoparticle to microparticles causing the drop of its biological activity. A new parameter, the Cl/N ratio of the PEIm, also influenced the biological activity together with the chemical properties of the solvent. Based on these findings we have developed a pDNA/PEIm formulation capable to maintain the physical stability and the biological activity of the nanomedicine. This work illustrates some of the key steps that must be taken for the implementation of "Quality by Design" (QbD) approach for a biotech product.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Portadores de Fármacos/química , Nanoestructuras/química , Tecnología Farmacéutica/métodos , Vacunas contra el SIDA/química , Química Farmacéutica/métodos , ADN/administración & dosificación , ADN/química , Estabilidad de Medicamentos , Almacenaje de Medicamentos , Electroforesis en Gel de Agar , Glucosa/administración & dosificación , Glucosa/química , Antígenos VIH/genética , Mananos/química , Tamaño de la Partícula , Plásmidos/administración & dosificación , Plásmidos/química , Polietileneimina/química , Soluciones , Espectrofotometría Ultravioleta
15.
Vaccine ; 26(40): 5188-95, 2008 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-18462844

RESUMEN

IL-7 and IL-15 are key cytokines involved in the generation and maintenance of memory CD8+ T-cells. We evaluated these cytokines as molecular adjuvants for topical HIV-1 DermaVir vaccine. We found that mice receiving DermaVir formulated with HIV-1 Gag plasmid in the presence of IL-7- or IL-15-encoding plasmid significantly enhanced Gag-specific central memory T-cells, as measured by a peptide-based cultured IFN-gamma ELISPOT. Additionally, IL-15 significantly improved DermaVir-induced Gag-specific effector memory CD8+ T-cell responses, measured by standard IFN-gamma ELISPOT. In a DermaVir prime/vaccinia vector boost regimen, the inclusion of IL-15 together with DermaVir significantly improved Gag-specific effector memory T-cell responses. Our study demonstrates that IL-15 is more potent than IL-7 in enhancing HIV-1-specific central memory T-cells induced by topical DermaVir. IL-15 adjuvanted DermaVir might be an alternative prime in a prophylactic vaccine regimen.


Asunto(s)
Vacunas contra el SIDA/inmunología , Adyuvantes Inmunológicos , Memoria Inmunológica , Linfocitos T/inmunología , Vacunas contra el SIDA/administración & dosificación , Administración Tópica , Animales , Linfocitos T CD8-positivos/inmunología , Femenino , Productos del Gen gag/inmunología , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , VIH-1/inmunología , Interleucina-15/administración & dosificación , Interleucina-15/inmunología , Interleucina-7/administración & dosificación , Interleucina-7/inmunología , Ratones , Ratones Endogámicos BALB C , Vacunación
16.
J Immunol ; 180(9): 5907-15, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18424710

RESUMEN

Evidences have recently suggested that the preservation of vaccine-induced memory rather than effector T cells is essential for better outcome and survival following pathogenic SIV challenge in macaques. However, an equivalent demonstration in humans is missing, and the immune correlates of HIV-1 control have been only partially characterized. We focused on the quantification of Ag-specific T cell precursors with high proliferative capacity (PHPC) using a peptide-based cultured IFN-gamma ELISPOT assay (PHPC assay), which has been shown to identify expandable memory T cells. To determine which responses correlate with viral suppression and positive immunologic outcome, PBMC from 32 chronically untreated HIV-1-infected individuals were evaluated in response to peptide pools, representing the complete HIV-1 Gag, Nef, and Rev proteins, by PHPC and IFN-gamma ELISPOT assay, which instead identifies effector T cells with low proliferative capacity. High magnitude of Gag-specific PHPC, but not ELISPOT, responses significantly correlated with low plasma viremia, due to responses directed toward p17 and p15 subunits. Only Gag p17-specific PHPC response significantly correlated with high CD4 counts. Analysis of 20 additional PBMC samples from an independent cohort of chronically untreated HIV-1-infected individuals confirmed the correlation between Gag p17-specific PHPC response and either plasma viremia (inverse correlation) or CD4 counts (direct correlation). Our results indicate that the PHPC assay is quantitatively and qualitatively different from the ELISPOT assay, supporting that different T cell populations are being evaluated. The PHPC assay might be an attractive option for individual patient management and for the design and testing of therapeutic and prophylactic vaccines.


Asunto(s)
Recuento de Linfocito CD4 , Proliferación Celular , Infecciones por VIH/inmunología , VIH-1/inmunología , Células Precursoras de Linfocitos T/inmunología , Viremia/inmunología , Vacunas contra el SIDA/inmunología , Vacunas contra el SIDA/metabolismo , Animales , Enfermedad Crónica , Estudios de Cohortes , Femenino , Proteína p24 del Núcleo del VIH/inmunología , Proteína p24 del Núcleo del VIH/metabolismo , Infecciones por VIH/sangre , Infecciones por VIH/prevención & control , VIH-1/metabolismo , Humanos , Memoria Inmunológica , Interferón gamma/inmunología , Interferón gamma/metabolismo , Macaca , Masculino , Células Precursoras de Linfocitos T/metabolismo , Células Precursoras de Linfocitos T/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/sangre , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Virus de la Inmunodeficiencia de los Simios/metabolismo , Viremia/prevención & control
17.
Curr Opin HIV AIDS ; 3(2): 104-11, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19372950

RESUMEN

PURPOSE OF REVIEW: In the next 5-10 years, neither antiviral drug treatment nor a preventive vaccine is expected to provide a solution to the HIV epidemic. To achieve a relevant public health benefit, it is time to change focus and develop immune therapies to achieve a beneficial viral load reduction in the absence of drugs. RECENT FINDINGS: Induction of long-term immune control with only limited rebounds of the wild-type HIV virus was found to be feasible and achievable, albeit in very limited cases. This review provides an analysis of the past achievements and an overview for the present therapeutic approaches under clinical development that could succeed to suppress viral replication after treatment interruption. SUMMARY: Interruption of antiretroviral treatment results in rapid viral load rebound and disease progression. There is no immune therapy on the market for the treatment of HIV that could lower the viral load set point by over 0.5 log to provide significant clinical benefit. A lower viral load maintained by the patient's own immune system following treatment interruption could offer a favorable prognosis and represent an alternative additional approach to antiretroviral drugs.

18.
J Infect Dis ; 196(9): 1409-15, 2007 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-17922407

RESUMEN

Human immunodeficiency virus (HIV) expression is boosted after T lymphocyte stimulation. It is not known, however, in which phase(s) of the cell cycle HIV is maximally expressed. We demonstrate here that cell activation induces limited HIV expression and that progression to cell proliferation is required for optimal HIV replication. We also show that the G1/S cell cycle transition is a critical checkpoint in this process and that limiting progression at this step with antiproliferative drugs suppresses HIV replication. These results identify a specific phase of the cell cycle progression that is critical for HIV expression and suggest a new discrete target for anti-HIV treatment.


Asunto(s)
Fármacos Anti-VIH/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/virología , Ciclo Celular/efectos de los fármacos , VIH-1/efectos de los fármacos , VIH-1/fisiología , Replicación Viral/efectos de los fármacos , Linfocitos T CD4-Positivos/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Inhibidores Enzimáticos/farmacología , Regulación Viral de la Expresión Génica , Humanos , Hidroxiurea/farmacología , Ácido Micofenólico/análogos & derivados , Ácido Micofenólico/farmacología
19.
Expert Opin Biol Ther ; 7(10): 1563-74, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17916048

RESUMEN

To improve the efficacy of DNA immunization epidermal Langerhans cells are attractive targets to deliver antigen-encoding plasmid DNA. Topical vaccination with naked plasmid DNA has been shown to induce immune responses, and their potency might be improved by chemical and physical methods aimed to enhance the efficiency of plasmid DNA delivery into the skin. Cytokines have also been evaluated as adjuvants for DNA vaccines because they influence the host immune response. This review focuses on the action of several cytokines tested as molecular adjuvants for DNA vaccines and the combination of them with the DermaVir Patch vaccine. DermaVir vaccine, topically administered under a patch, consists of a plasmid DNA that is chemically formulated into a nanoparticle to support vaccine delivery into epidermal Langerhans cells and to induce antigen-specific memory T cells.


Asunto(s)
Vacunas contra el SIDA/metabolismo , Adyuvantes Inmunológicos/metabolismo , Citocinas/metabolismo , Técnicas de Transferencia de Gen , Células de Langerhans/metabolismo , Absorción Cutánea/efectos de los fármacos , Vacunas de ADN/metabolismo , Vacunas contra el SIDA/inmunología , Adyuvantes Inmunológicos/genética , Animales , Citocinas/genética , Humanos , Células de Langerhans/inmunología , Nanopartículas , Linfocitos T/inmunología , Vacunas de ADN/inmunología
20.
Virology ; 366(1): 197-211, 2007 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-17499328

RESUMEN

Topical DNA vaccination (DermaVir) facilitates antigen presentation to naive T cells. DermaVir immunization in mice, using HIV-1 Env and Gag, elicited cellular immune responses. Boosting with HIV-1 gp120 Env and p41 Gag augmented Th1 cytokine levels. Intramuscular DNA administration was less efficient in priming antigen-specific cytokine production and memory T cells. In rhesus macaques, DermaVir immunization induced Gag- and Env-specific Th1 and Th2 cytokines and generation of memory T cells. Boosting of DermaVir-primed serum antibody levels was noted following gp140(SHIV89.6P)/p27(SIV) immunization. Rectal challenge with pathogenic R5-tropic SHIV162P3 resulted in control of plasma viremia (4/5 animals) that was reflected in jejunum, colon and mesenteric lymph nodes. An inverse correlation was found between Gag- and Env-specific central memory T cell responses on the day of challenge and plasma viremia at set point. Overall, the topical DermaVir/protein vaccination yields central memory T cell responses and facilitates control of pathogenic SHIV infection.


Asunto(s)
Vacunas contra el SIDA/inmunología , Síndrome de Inmunodeficiencia Adquirida/inmunología , VIH-1/inmunología , Vacunas contra el SIDA/uso terapéutico , Animales , Codón , Citocinas/análisis , Citocinas/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Genes env , Proteína gp120 de Envoltorio del VIH/inmunología , VIH-1/patogenicidad , Inmunización Secundaria , Macaca mulatta , Ratones , Virus de la Inmunodeficiencia de los Simios/inmunología , Linfocitos T Citotóxicos/inmunología , Células TH1/inmunología , Células Th2/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA