Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 300
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Br J Cancer ; 124(11): 1795-1802, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33762716

RESUMEN

BACKGROUND: Preoperative chemotherapy containing anthracyclines and taxanes is well established in early-stage breast cancer. Previous studies have suggested that the chemotherapy sequence may matter but definitive evidence is missing. ABCSG trial 34 evaluated the activity of the MUC1 vaccine tecemotide when added to neoadjuvant treatment; the study provided the opportunity for the second randomisation to compare two different anthracycline/taxane sequences. METHODS: HER2-negative early-stage breast cancer patients were recruited to this randomised multicentre Phase 2 study. Patients in the chemotherapy cohort (n = 311) were additionally randomised to a conventional or reversed sequence of epirubicin/cyclophosphamide and docetaxel. Residual cancer burden (RCB) with/without tecemotide was defined as primary study endpoint; RCB in the two chemotherapy groups was a key secondary endpoint. RESULTS: No significant differences in terms of RCB 0/I (40.1% vs. 37.2%; P = 0.61) or pathologic complete response (pCR) rates (24.3% vs. 25%, P = 0.89) were observed between conventional or reverse chemotherapy sequence. No new safety signals were reported, and upfront docetaxel did not result in decreased rates of treatment delay or discontinuation. CONCLUSION: Upfront docetaxel did not improve chemotherapy activity or tolerability; these results suggest that upfront neoadjuvant treatment with anthracyclines remains a valid option.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/administración & dosificación , Glicoproteínas de Membrana/administración & dosificación , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/mortalidad , Neoplasias de la Mama/patología , Ciclofosfamida/administración & dosificación , Ciclofosfamida/efectos adversos , Docetaxel/administración & dosificación , Docetaxel/efectos adversos , Esquema de Medicación , Epirrubicina/administración & dosificación , Epirrubicina/efectos adversos , Femenino , Humanos , Persona de Mediana Edad , Terapia Neoadyuvante/métodos , Estadificación de Neoplasias , Neoplasia Residual , Carga Tumoral
2.
Hum Mol Genet ; 27(17): 3079-3098, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29878115

RESUMEN

Niemann-Pick type C disease (NP-C) is a fatal neurodegenerative lysosomal storage disorder. It is caused in 95% of cases by a mutation in the NPC1 gene that encodes NPC1, an integral transmembrane protein localized to the limiting membrane of the lysosome. There is no cure for NP-C but there is a disease-modifying drug (miglustat) that slows disease progression but with associated side effects. Here, we demonstrate in a well-characterized mouse model of NP-C that a single administration of AAV-mediated gene therapy to the brain can significantly extend lifespan, improve quality of life, prevent or ameliorate neurodegeneration, reduce biochemical pathology and normalize or improve various indices of motor function. Over-expression of human NPC1 does not cause adverse effects in the brain and correctly localizes to late endosomal/lysosomal compartments. Furthermore, we directly compare gene therapy to licensed miglustat. Even at a low dose, gene therapy has all the benefits of miglustat but without adverse effects. On the basis of these findings and on-going ascendency of the field, we propose intracerebroventricular gene therapy as a potential therapeutic option for clinical use in NP-C.


Asunto(s)
Adenoviridae/genética , Proteínas Portadoras/administración & dosificación , Modelos Animales de Enfermedad , Trastornos Neurológicos de la Marcha/prevención & control , Terapia Genética , Longevidad/genética , Glicoproteínas de Membrana/administración & dosificación , Enfermedad de Niemann-Pick Tipo C/prevención & control , Animales , Proteínas Portadoras/fisiología , Trastornos Neurológicos de la Marcha/genética , Trastornos Neurológicos de la Marcha/patología , Humanos , Inflamación/genética , Inflamación/patología , Inflamación/prevención & control , Péptidos y Proteínas de Señalización Intracelular , Glicoproteínas de Membrana/fisiología , Ratones , Ratones Transgénicos , Mutación , Proteína Niemann-Pick C1 , Enfermedad de Niemann-Pick Tipo C/genética , Enfermedad de Niemann-Pick Tipo C/patología
3.
Eur Radiol ; 30(6): 3188-3197, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32060711

RESUMEN

OBJECTIVES: When increasing the PET acquisition time to match the longer MRI protocol in simultaneous PET/MR, the injected PET tracer dose can possibly be lowered to reduce radiation exposure. Moreover, applying new commercially available time-of-flight (TOF) block sequential regularized expectation maximization (BSREM)-based reconstruction algorithms could allow for further dose reductions. The purpose of this study was to find the minimal dose of the tracer targeting the prostate specific membrane antigen (68Ga-PSMA-11) for a dedicated 15-min pelvic PET/MR scan that still matches the image quality of a reference 3-min scan at 100% (150 MBq) dose. METHODS: In this retrospective analysis, 25 patients were included. PET emission datasets were edited to simulate stepwise reductions of injected tracer dose. Reference TOF ordered subset expectation maximum (OSEM) and new TOF BSREM reconstructions were performed and differences in the resulting PET images were visually and quantitatively assessed. RESULTS: Visually, TOF BSREM reconstructions with relatively high regularization parameter (ß) values are preferred. Quantitatively, however, high ß-values result in lower lesion maximum standardized uptake values (SUVmax) compared to the reference. A ß-value of 550 was considered the optimal compromise for the lowest possible 10% dose reconstructions, resulting in comparable visual assessment and lesion SUVmax. CONCLUSIONS: This study indicates that the injected 68Ga-PSMA-11 tracer dose for a standard 3-min PET scan can be reduced to approximately 10% (15 MBq) when the PET acquisition time is matched to the 15-min pelvic MRI protocol, and when reconstructed with TOF BSREM using ß = 550. This decreases the effective dose from 3.54 to 0.35 mSv. KEY POINTS: • Low-dose dedicated pelvic68Ga-PSMA-11 PET/MR reduces radiation exposure for patients. • Retrospective study investigating the minimal dose needed for adequate image quality for 15-min PET frames over the pelvis showed using quantitative and qualitative analysis that a substantial dose reduction is possible without significant loss of image quality when using the TOF BSREM reconstruction algorithm. • With the introduction of low-dose pelvic68Ga-PSMA-11 PET/MR, new potential applications of68Ga-PSMA-11 PET for local staging or investigation of equivocal MRI findings could become applicable, even for patients without confirmed prostate cancer.


Asunto(s)
Radioisótopos de Galio/administración & dosificación , Glicoproteínas de Membrana/administración & dosificación , Compuestos Organometálicos/administración & dosificación , Pelvis/diagnóstico por imagen , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Próstata/diagnóstico por imagen , Anciano , Algoritmos , Antígenos de Superficie , Isótopos de Galio , Glutamato Carboxipeptidasa II , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Próstata/diagnóstico por imagen , Dosis de Radiación , Estudios Retrospectivos
4.
J Virol ; 92(19)2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30021899

RESUMEN

An effective human immunodeficiency virus (HIV) vaccine has yet to be developed, and defining immune correlates of protection against HIV infection is of paramount importance to inform future vaccine design. The complement system is a component of innate immunity that can directly lyse pathogens and shape adaptive immunity. To determine if complement lysis of simian immunodeficiency virus (SIV) and/or SIV-infected cells represents a protective immune correlate against SIV infection, sera from previously vaccinated and challenged rhesus macaques were analyzed for the induction of antibody-dependent complement-mediated lysis (ADCML). Importantly, the vaccine regimen, consisting of a replication-competent adenovirus type 5 host-range mutant SIV recombinant prime followed by a monomeric gp120 or oligomeric gp140 boost, resulted in overall delayed SIV acquisition only in females. Here, sera from all vaccinated animals induced ADCML of SIV and SIV-infected cells efficiently, regardless of sex. A modest correlation of SIV lysis with a reduced infection rate in males but not females, together with a reduced peak viremia in all animals boosted with gp140, suggested a potential for influencing protective efficacy. Gag-specific IgG and gp120-specific IgG and IgM correlated with SIV lysis in females, while Env-specific IgM correlated with SIV-infected cell lysis in males, indicating sex differences in vaccine-induced antibody characteristics and function. In fact, gp120/gp140-specific antibody functional correlates between antibody-dependent cellular cytotoxicity, antibody-dependent phagocytosis, and ADCML as well as the gp120-specific IgG glycan profiles and the corresponding ADCML correlations varied depending on the sex of the vaccinees. Overall, these data suggest that sex influences vaccine-induced antibody function, which should be considered in the design of globally effective HIV vaccines in the future.IMPORTANCE An HIV vaccine would thwart the spread of HIV infection and save millions of lives. Unfortunately, the immune responses conferring universal protection from HIV infection are poorly defined. The innate immune system, including the complement system, is an evolutionarily conserved, basic means of protection from infection. Complement can prevent infection by directly lysing incoming pathogens. We found that vaccination against SIV in rhesus macaques induces antibodies that are capable of directing complement lysis of SIV and SIV-infected cells in both sexes. We also found sex differences in vaccine-induced antibody species and their functions. Overall, our data suggest that sex affects vaccine-induced antibody characteristics and function and that males and females might require different immune responses to protect against HIV infection. This information could be used to generate highly effective HIV vaccines for both sexes in the future.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Anticuerpos Antivirales/biosíntesis , Proteínas del Sistema Complemento/inmunología , Vacunas contra el SIDAS/administración & dosificación , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/efectos de los fármacos , Adenovirus de los Simios/genética , Adenovirus de los Simios/inmunología , Animales , Proteínas del Sistema Complemento/agonistas , Proteínas del Sistema Complemento/genética , Citotoxicidad Inmunológica , Femenino , Regulación de la Expresión Génica , Productos del Gen env/administración & dosificación , Productos del Gen env/genética , Productos del Gen env/inmunología , Sueros Inmunes/química , Inmunización Secundaria/métodos , Inmunoglobulina G/biosíntesis , Inmunoglobulina M/biosíntesis , Macaca mulatta , Masculino , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Vacunas contra el SIDAS/genética , Vacunas contra el SIDAS/inmunología , Factores Sexuales , Transducción de Señal , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Vacunas Sintéticas , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
5.
Protein Expr Purif ; 153: 7-17, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30081196

RESUMEN

A transmission-blocking vaccine targeting the sexual stages of Plasmodium species could play a key role in eradicating malaria. Multiple studies have identified the P. falciparum proteins Pfs25 and Pfs48/45 as prime targets for transmission-blocking vaccines. Although significant advances have been made in recombinant expression of these antigens, they remain difficult to produce at large scale and lack strong immunogenicity as subunit antigens. We linked a self-assembling protein, granule lattice protein 1 (Grl1p), from the ciliated protozoan, Tetrahymena thermophila, to regions of the ectodomains of either Pfs25 or Pfs48/45. We found that resulting protein chimera could be produced in E. coli as nanoparticles that could be readily purified in soluble form. When produced in the E. coli SHuffle strain, fusion to Grl1p dramatically increased solubility of target antigens while at the same time directing the formation of particles with diameters centering on 38 and 25 nm depending on the antigen. In a number of instances, co-expression with chaperone proteins and induction at a lower temperature further increased expression and solubility. Based on Western blotting and ELISA analysis, Pfs25 and Pfs48/45 retained their transmission-blocking epitopes within E. coli-derived particles, and the particles themselves elicited strong antibody responses in rabbits when given with an aluminum-based adjuvant. Antibodies against Pfs25-containing nanoparticles blocked parasite transmission in standard membrane-feeding assays. In conclusion, fusion to Grl1p can act as a solubility enhancer for proteins with limited solubility while retaining correct folding, which may be useful for applications such as the production of vaccines and other biologics.


Asunto(s)
Anticuerpos Antiprotozoarios/biosíntesis , Proteínas de Unión al Calcio/genética , Vacunas contra la Malaria/genética , Malaria Falciparum/prevención & control , Glicoproteínas de Membrana/genética , Plasmodium falciparum/química , Proteínas Protozoarias/genética , Tetrahymena thermophila/química , Animales , Antígenos de Protozoos/administración & dosificación , Antígenos de Protozoos/química , Antígenos de Protozoos/genética , Antígenos de Protozoos/inmunología , Bioensayo , Proteínas de Unión al Calcio/administración & dosificación , Proteínas de Unión al Calcio/química , Proteínas de Unión al Calcio/inmunología , Clonación Molecular , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Inmunogenicidad Vacunal , Vacunas contra la Malaria/administración & dosificación , Malaria Falciparum/inmunología , Malaria Falciparum/parasitología , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/inmunología , Mosquitos Vectores/parasitología , Nanopartículas , Plasmodium falciparum/inmunología , Pliegue de Proteína , Proteínas Protozoarias/administración & dosificación , Proteínas Protozoarias/química , Proteínas Protozoarias/inmunología , Conejos , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/inmunología , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Solubilidad , Tetrahymena thermophila/inmunología
6.
J Immunol ; 198(4): 1595-1605, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-28077601

RESUMEN

Among innovative adjuvants conferring a Th1-shift, RNAdjuvant is a promising candidate. This adjuvant consists of a 547-nt uncapped noncoding ssRNA containing polyU repeats that is stabilized by a cationic carrier peptide. Whereas vaccination of mice with an influenza subunit vaccine induced moderate virus-specific IgG1, vaccination together with RNAdjuvant significantly enhanced this IgG1 and additionally promoted the formation of IgG2b/c, which is indicative of Th1 responses. Furthermore, such sera neutralized influenza virus, whereas this effect was not detected upon vaccination with the subunit vaccine alone. Similarly, upon vaccination with virus-like particles displaying vesicular stomatitis virus G protein, RNAdjuvant promoted the formation of virus-specific IgG2b/c and enhanced neutralizing IgG responses to an extent that mice were protected against lethal virus infection. RNAdjuvant induced dendritic cells to upregulate activation markers and produce IFN-I. Although these effects were strictly TLR7 dependent, RNAdjuvant-mediated augmentation of vaccine responses needed concurrent TLR and RIG-I-like helicase signaling. This was indicated by the absence of the adjuvant effect in vaccinated MyD88-/-Cardif-/- mice, which are devoid of TLR (with the exception of TLR3) and RIG-I-like helicase signaling, whereas in vaccinated MyD88-/- mice the adjuvant effect was reduced. Notably, i.m. RNAdjuvant injection induced local IFN-I responses and did not induce systemic effects, implying good tolerability and a favorable safety profile for RNAdjuvant.


Asunto(s)
Adyuvantes Inmunológicos , Inmunoglobulina G/sangre , Vacunas contra la Influenza/inmunología , Glicoproteínas de Membrana/inmunología , ARN no Traducido/inmunología , Receptor Toll-Like 7/inmunología , Vacunas de Partículas Similares a Virus/inmunología , Adyuvantes Inmunológicos/efectos adversos , Animales , Anticuerpos Antivirales/sangre , Proteína 58 DEAD Box/inmunología , Proteína 58 DEAD Box/metabolismo , Inmunoglobulina G/inmunología , Vacunas contra la Influenza/administración & dosificación , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/metabolismo , Células TH1/inmunología , Receptor Toll-Like 7/metabolismo , Vacunación , Vacunas de Subunidad/inmunología , Vacunas de Partículas Similares a Virus/administración & dosificación , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/inmunología
7.
Int J Urol ; 26(8): 804-811, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31083784

RESUMEN

OBJECTIVE: To evaluate the clinical impact of 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography on the planned management of prostate cancer patients with biochemical recurrence after surgery. METHODS: We enrolled 276 prostate cancer patients referred to 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography due to biochemical recurrence after surgery (two consecutive prostate-specific antigen assays ≥0.2 ng/mL). First, the detection rate of 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography was assessed according to different prostate-specific antigen levels. Second, the independent predictors of 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography positive results were assessed. Finally, the intended treatment before revision of 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography was assessed by a multidisciplinary team based on the European Association of Urology guidelines, patient clinical condition and clinical parameters. Then, re-assessment of the treatment plan was prospectively recorded by the same board after revision of 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography. The effective clinical impact of 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography was rated as major (change in therapeutic approach), minor (same treatment, but modified therapeutic strategy) or none. RESULTS: The overall detection rate of 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography was 47.5%. Prostate-specific antigen at 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography (odds ratio 3.52) and prostate-specific antigen doubling time <3 months (odds ratio 3.98) were independent predictors of positive 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography results (all P ≤ 0.03). 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography led to a major treatment change in 177 cases (64.1%), with a minor clinical impact of 2.5%. The overall clinical impact of 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography was 42.4%, 27.7%, 21.2% and 8.7% in men with prostate-specific antigen at 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography of 0.2-0.4, 0.5-1, 1.1-2 and >2 ng/mL, respectively. CONCLUSIONS: 68 Ga-prostate-specific membrane antigen positron emission tomography/computed tomography allows clinicians to radically change the intended treatment approach before imaging evaluation, in roughly two out three individuals.


Asunto(s)
Glicoproteínas de Membrana/administración & dosificación , Recurrencia Local de Neoplasia/diagnóstico , Compuestos Organometálicos/administración & dosificación , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Próstata/diagnóstico por imagen , Neoplasias de la Próstata/terapia , Anciano , Antagonistas de Andrógenos/uso terapéutico , Quimioradioterapia Adyuvante/métodos , Toma de Decisiones Clínicas/métodos , Estudios de Factibilidad , Isótopos de Galio , Radioisótopos de Galio , Humanos , Calicreínas/sangre , Masculino , Persona de Mediana Edad , Recurrencia Local de Neoplasia/sangre , Selección de Paciente , Estudios Prospectivos , Próstata/cirugía , Antígeno Prostático Específico/sangre , Prostatectomía , Neoplasias de la Próstata/sangre , Neoplasias de la Próstata/diagnóstico , Radiofármacos/administración & dosificación
8.
Biochem Biophys Res Commun ; 495(1): 1227-1232, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29170125

RESUMEN

The S-layer is a (glyco)-proteinaceous envelope constituted by self-assembled subunits that form a two-dimensional lattice covering the surface of different species of Bacteria and Archaea. It could be considered as one of the most abundant biopolymers in our planet. Because of their unique self-assembly features, exhibiting repetitive identical physicochemical properties down to the subnanometer scale, as well as their involvement in specific interactions with host cells, the S-layer proteins (SLPs) show a high potential application in different areas of biotechnology, including the development of antigen carriers or new adjuvants. The presence of a glycosylated SLP on potentially probiotic Lactobacillus kefiri strains was previously described by our research group. In this study, we aim to investigate the role of carbohydrates present in the SLP from L. kefiri CIDCA 8348 (SLP-8348) in their internalization by murine macrophages, as well as to analyze their immunomodulatory capacity and their effect on LPS-stimulated macrophages. RAW 264.7 cells internalized the SLP-8348 in a process that was mediated by carbohydrate-receptor interactions since it was inhibited by glucose, mannose or EGTA, a Ca+2 chelating agent. These results correlated with the recognition of SLP-8348 by ConA lectin. We further show that while SLP-8348 was not able to induce the activation of macrophages by itself, it favored the LPS-induced response, since there was a significant increase in the expression of surface cell markers MHC-II, CD86 and CD40, as well as in IL-6 and IL-10 expression at both transcript and protein levels, in comparison with LPS-stimulated cells. The presence of EGTA completely abrogated this synergistic effect. Taken together, these results strongly suggest the involvement of both glycosidic residues and Ca+2 ions in the recognition of SLP-8348 by cellular receptors on murine macrophages. Moreover, these results suggest the potentiality of the SLP-8348 for the development of new adjuvants capable of stimulating antigen presenting cells by interaction with glycan receptors.


Asunto(s)
Calcio/inmunología , Lactobacillus/metabolismo , Lipopolisacáridos/administración & dosificación , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Glicoproteínas de Membrana/administración & dosificación , Animales , Señalización del Calcio/efectos de los fármacos , Señalización del Calcio/inmunología , Células Cultivadas , Sinergismo Farmacológico , Lactobacillus/clasificación , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Células RAW 264.7
9.
Mol Pharm ; 15(8): 3502-3511, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-29920108

RESUMEN

68Ga-PSMA-11 is currently the most popular prostate-specific membrane antigen (PSMA) radioligand used in the clinic to detect prostate cancer and metastases. However, the high uptake of 68Ga-PSMA-11 in kidneys can create halo-artifacts resulting in lower detection sensitivity for lesions adjacent to the kidneys. In this study, we developed two 68Ga-labeled PSMA-targeted tracers, 68Ga-HTK01166 and 68Ga-HTK01167, based on 68Ga-PSMA-617 with the goal of improving tumor-to-kidney ratio compared to 68Ga-PSMA-11. The 2-naphthylalanine (2-Nal) in PSMA-617 was replaced with 2-indanylglycine (Igl) or 3,3-diphenylalanine (Dip) to synthesize HTK01166 and HTK01167, respectively. Binding affinities ( Ki) of Ga-PSMA-11, Ga-PSMA-617, Ga-HTK01166, and Ga-HTK01167 to PSMA were 3.13 ± 0.40, 1.23 ± 0.08, 5.74 ± 2.48, and 25.7 ± 9.84 nM, respectively, as determined by in vitro competition binding assays. 68Ga labeling was performed in HEPES buffer with microwave heating, and 68Ga-labeled PSMA-11, PSMA-617, HTK01166, and HTK01167 were obtained in 46-69% average decay-corrected radiochemical yield with >99% radiochemical purity and 62.9-152 GBq/µmol average specific activity. PET imaging and biodistribution studies were performed in mice bearing PSMA-expressing LNCap prostate cancer xenografts. All tracers enabled clear visualization of tumors in PET images with excellent tumor-to-background contrast. The uptake values (%ID/g) for tumor and kidneys at 1 h postinjection were 8.91 ± 0.86 and 204 ± 70.6 for 68Ga-PSMA-11, 16.7 ± 2.30 and 29.2 ± 5.14 for 68Ga-PSMA-617, 14.1 ± 4.40 and 147 ± 59.6 for 68Ga-HTK01166, and 7.79 ± 1.65 and 4.30 ± 1.80 for 68Ga-HTK01167. The tumor-to-kidney ratios for 68Ga-labeled PSMA-11, PSMA-617, HTK01166, and HTK01167 were 0.05 ± 0.02, 0.63 ± 0.10, 0.10 ± 0.02, and 1.98 ± 0.63, respectively. Compared with 68Ga-PSMA-617, 68Ga-HTK01166 showed comparable tumor uptake and almost 5-fold higher kidney uptake, whereas 68Ga-HTK01167 exhibited lower tumor and kidney uptake. Compared with 68Ga-PSMA-11, 68Ga-HTK01167 had similar tumor uptake and tumor-to-blood contrast ratio (23.8 ± 6.71 vs 20.4 ± 4.98) but higher tumor-to-background contrast ratios for other background organs especially for kidneys. Our data indicate that substitution of 2-Nal in PSMA-617 with other lipophilic amino acid can modulate PSMA binding affinity and their pharmacokinetics in vivo.


Asunto(s)
Antígenos de Superficie/metabolismo , Radioisótopos de Galio/farmacocinética , Glutamato Carboxipeptidasa II/metabolismo , Glicoproteínas de Membrana/farmacocinética , Compuestos Organometálicos/farmacocinética , Neoplasias de la Próstata/diagnóstico por imagen , Animales , Línea Celular Tumoral , Isótopos de Galio , Radioisótopos de Galio/administración & dosificación , Radioisótopos de Galio/química , Humanos , Riñón/metabolismo , Masculino , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/química , Ratones , Compuestos Organometálicos/administración & dosificación , Compuestos Organometálicos/química , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones/métodos , Próstata/diagnóstico por imagen , Próstata/patología , Neoplasias de la Próstata/patología , Distribución Tisular , Microtomografía por Rayos X/métodos , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Cell Physiol ; 232(3): 526-539, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27255231

RESUMEN

Malignant glioma continues to be a clinical challenge with an urgent need for developing curative therapeutic intervention. Apoptosis induction in tumor-associated endothelial cells represent a central mechanism that counteracts angiogenesis in glioma and other solid tumors. We previously demonstrated that intraperitoneal administration of sheep erythrocyte membrane glycopeptide T11-target structure (T11TS) in rodent glioma model inhibits PI3K/Akt pathway and Raf/MEK/ERK signaling in glioma-associated brain endothelial cells. In the present study, we investigated whether T11TS treatment influence apoptosis signaling in vivo in glioma-associated brain endothelial cells. Annexin-V/PI staining showed that T11TS treatment in glioma-induced rats increases apoptosis of glioma-associated endothelial cells within glioma milieu compared to brain endothelial cells in glioma induced and control groups. Flowcytometric JC-1 assay revealed that T11TS administration triggers loss of mitochondrial membrane potential in glioma-associated brain endothelial cells. Flowcytometry, immunoblotting, and in situ immunofluoresecnt imaging were employed to investigate the effect of T11TS on apoptotic regulatory proteins in brain endothelial cells. T11TS treatment-upmodulated expression of p53, Bax, Fas, FasL, and FADD in glioma associated endothelial cells and downregulated Bcl-2 protein. T11TS therapy induced cytochrome-c release into cytosol, activated caspase -9, 8, 3, and cleaved Bid in glioma associated brain endothelial cells. The study demonstrates that T11TS induces apoptosis in glioma-associated brain endothelial cells via p53 accumulation and activation of intrinsic as well as Fas-dependent extrinsic pathway. The pro-apoptotic action of T11TS on glioma-associated endothelial cells provides crucial insight into how T11TS exerts its anti-angiogenic function in glioma. J. Cell. Physiol. 232: 526-539, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Inhibidores de la Angiogénesis/uso terapéutico , Apoptosis/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Células Endoteliales/patología , Glioma/patología , Glicopéptidos/farmacología , Glicopéptidos/uso terapéutico , Glicoproteínas de Membrana/farmacología , Glicoproteínas de Membrana/uso terapéutico , Neovascularización Patológica/tratamiento farmacológico , Inhibidores de la Angiogénesis/farmacología , Animales , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Neoplasias Encefálicas/patología , Caspasas/metabolismo , Citocromos c/metabolismo , Citosol/metabolismo , Células Endoteliales/efectos de los fármacos , Activación Enzimática , Proteína Ligando Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Femenino , Glicopéptidos/administración & dosificación , Masculino , Glicoproteínas de Membrana/administración & dosificación , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Modelos Biológicos , Neovascularización Patológica/patología , Ratas , Ovinos , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Receptor fas/metabolismo
11.
J Immunol ; 190(6): 2495-9, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23401588

RESUMEN

Vaccine-induced protection against infection by HIV or highly pathogenic and virulent SIV strains has been limited. In a proof-of-concept study, we show that a novel vaccine approach significantly protects rhesus macaques from mucosal infection by the highly pathogenic strain SIVmac251. We vaccinated three cohorts of 12 macaques each with live, irradiated vaccine cells secreting the modified endoplasmic reticulum chaperone gp96-Ig. Cohort 1 was vaccinated with cells secreting gp96(SIV)Ig carrying SIV peptides. In addition, Cohort 2 received recombinant envelope protein SIV-gp120. Cohort 3 was injected with cells secreting gp96-Ig (no SIV Ags) vaccines. Cohort 2 was protected from infection. After seven rectal challenges with highly pathogenic SIVmac251, the hazard ratio was 0.27, corresponding to a highly significant, 73% reduced risk for viral acquisition. The apparent success of the novel vaccine modality recommends further study.


Asunto(s)
Mucosa Intestinal/inmunología , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/uso terapéutico , Vacunas contra el SIDAS/administración & dosificación , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/prevención & control , Virus de la Inmunodeficiencia de los Simios/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antivirales/biosíntesis , Estudios de Cohortes , Femenino , Células HEK293 , Humanos , Inyecciones Intraperitoneales , Mucosa Intestinal/patología , Mucosa Intestinal/virología , Macaca mulatta , Masculino , Membrana Mucosa/inmunología , Membrana Mucosa/virología , Enfermedades del Recto/inmunología , Enfermedades del Recto/prevención & control , Enfermedades del Recto/virología , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/patología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Linfocitos T Citotóxicos/virología
12.
J Immunol ; 191(4): 1965-75, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23851696

RESUMEN

Modulation of immune responses is one of the main research aims in transplant immunology. In this study, we investigate the local immunomodulatory properties of soluble CD83 (sCD83) at the graft-host interface using the high-risk corneal transplantation model. In this model, which mimics the inflammatory status and the preexisting vascularization of high-risk patients undergoing corneal transplantation, allogeneic donor corneas are transplanted onto sCD83-treated recipient animals. This model allows the direct and precise application of the immune modulator at the transplantation side. Interestingly, sCD83 was able to prolong graft survival after systemic application as well as after topical application, which is therapeutically more relevant. The therapeutic effect was accompanied by an increase in the frequency of regulatory T cells and was mediated by the immune-regulatory enzyme IDO and TGF-ß. In vitro, sCD83 induced long-term IDO expression in both conventional and plasmacytoid dendritic cells via autocrine or paracrine production of TGF-ß, a cytokine previously shown to be an essential mediator of IDO-dependent, long-term tolerance. These findings open new treatment avenues for local immune modulation after organ and tissue transplantation.


Asunto(s)
Antígenos CD/uso terapéutico , Trasplante de Córnea , Refuerzo Inmunológico de Injertos , Inmunoglobulinas/uso terapéutico , Factores Inmunológicos/uso terapéutico , Indolamina-Pirrol 2,3,-Dioxigenasa/fisiología , Glicoproteínas de Membrana/uso terapéutico , Linfocitos T Reguladores/efectos de los fármacos , Tolerancia al Trasplante/efectos de los fármacos , Administración Oftálmica , Aloinjertos , Animales , Antígenos CD/administración & dosificación , Antígenos CD/inmunología , Células de la Médula Ósea/inmunología , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/inmunología , Evaluación Preclínica de Medicamentos , Inducción Enzimática/efectos de los fármacos , Femenino , Factores de Transcripción Forkhead/análisis , Supervivencia de Injerto , Inmunoglobulinas/administración & dosificación , Inmunoglobulinas/inmunología , Factores Inmunológicos/administración & dosificación , Factores Inmunológicos/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Inyecciones Intraperitoneales , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Premedicación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/uso terapéutico , Solubilidad , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Factor de Crecimiento Transformador beta/administración & dosificación , Factor de Crecimiento Transformador beta/fisiología , Factor de Crecimiento Transformador beta/uso terapéutico , Antígeno CD83
13.
BMC Vet Res ; 11: 238, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26374102

RESUMEN

BACKGROUND: Neonatal and post-weaning colibacillosis caused by enterotoxigenic E. coli is responsible for substantial economic losses encountered by the pork industry. Intestinal colonization of young piglets by E. coli depends on the efficiency of bacterial attachment to host gastrointestinal epithelium that is mediated by fimbriae. We tested the effect of porcine individual milk fat globule membrane (MFGM) proteins on F4ac positive E. coli attachment to porcine enterocytes in vitro. RESULTS: Butyrophilin, lactadherin and fatty acid binding protein inhibited fimbriae-dependent adherence of E. coli to enterocytes in vitro, while xanthine dehydrogenase did not. The inhibiting activity was dose-dependent for all three proteins, but the inhibiting efficiency was different. CONCLUSIONS: The results indicate that MFGM proteins may interfere with attachment of E. coli to porcine neonatal intestinal mucosa.


Asunto(s)
Antígenos Bacterianos/metabolismo , Adhesión Bacteriana/efectos de los fármacos , Escherichia coli Enterotoxigénica/fisiología , Proteínas de Escherichia coli/metabolismo , Proteínas de Unión a Ácidos Grasos/farmacología , Proteínas Fimbrias/metabolismo , Glicoproteínas de Membrana/farmacología , Proteínas de la Leche/farmacología , Xantina Deshidrogenasa/farmacología , Animales , Antígenos Bacterianos/genética , Butirofilinas , Línea Celular , Enterocitos , Escherichia coli Enterotoxigénica/efectos de los fármacos , Proteínas de Escherichia coli/genética , Proteínas de Unión a Ácidos Grasos/administración & dosificación , Proteínas Fimbrias/genética , Glicoproteínas de Membrana/administración & dosificación , Proteínas de la Leche/administración & dosificación , Porcinos , Xantina Deshidrogenasa/administración & dosificación
14.
Parasite Immunol ; 36(7): 303-12, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24749785

RESUMEN

Schistosoma mansoni is a blood fluke parasite responsible for schistosomiasis. The best long-term strategy to control schistosomiasis is through immunization combined with drug treatment. In this study, we cloned, expressed and purified SmTSP-2 fused to the N- and C-terminal halves of Sm29 and tested these chimeras as vaccine candidates using an adjuvant approved to be used in humans. The results demonstrated that vaccination with SmTSP-2 fused to N- or C-terminus of Sm29-induced reduction in worm burden and liver pathology when compared to control animals. Additionally, we detected high levels of mouse-specific IgG, IgG1 and IgG2a against both chimeras and significant amounts of IFN-γ and TNF-α and no IL-4. Finally, studies with sera from patients resistant to infection and living in schistosomiasis endemic areas revealed high levels of specific IgG to both chimeras when compared to healthy individuals. In conclusion, SmTSP-2/Sm29 chimeras tested here induced partial protection against infection and might be a potential vaccine candidate.


Asunto(s)
Antígenos Bacterianos/inmunología , Antígenos Helmínticos/inmunología , Proteínas Bacterianas/inmunología , Proteínas del Helminto/inmunología , Glicoproteínas de Membrana/inmunología , Schistosoma mansoni , Esquistosomiasis mansoni/inmunología , Esquistosomiasis mansoni/prevención & control , Tetraspaninas/inmunología , Vacunas/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Animales , Anticuerpos Antihelmínticos/sangre , Antígenos Bacterianos/administración & dosificación , Antígenos Helmínticos/administración & dosificación , Proteínas Bacterianas/administración & dosificación , Islas de CpG , Citocinas/sangre , Femenino , Proteínas del Helminto/administración & dosificación , Humanos , Inmunoglobulina G/sangre , Hígado/patología , Glicoproteínas de Membrana/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Oligodesoxirribonucleótidos/administración & dosificación , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/inmunología , Tetraspaninas/administración & dosificación , Vacunas/administración & dosificación
15.
J Control Release ; 370: 66-81, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38631490

RESUMEN

Renal ischemia-reperfusion injury (IRI) is one of the most important causes of acute kidney injury (AKI). Interleukin (IL)-37 has been suggested as a novel anti-inflammatory factor for the treatment of IRI, but its application is still limited by its low stability and delivery efficiency. In this study, we reported a novel engineered method to efficiently and easily prepare neutrophil membrane-derived vesicles (N-MVs), which could be utilized as a promising vehicle to deliver IL-37 and avoid the potential side effects of neutrophil-derived natural extracellular vesicles. N-MVs could enhance the stability of IL-37 and targetedly deliver IL-37 to damaged endothelial cells of IRI kidneys via P-selectin glycoprotein ligand-1 (PSGL-1). In vitro and in vivo evidence revealed that N-MVs encapsulated with IL-37 (N-MV@IL-37) could inhibit endothelial cell apoptosis, promote endothelial cell proliferation and angiogenesis, and decrease inflammatory factor production and leukocyte infiltration, thereby ameliorating renal IRI. Our study establishes a promising delivery vehicle for the treatment of renal IRI and other endothelial damage-related diseases.


Asunto(s)
Células Endoteliales , Interleucina-1 , Riñón , Neutrófilos , Daño por Reperfusión , Animales , Daño por Reperfusión/tratamiento farmacológico , Interleucina-1/administración & dosificación , Masculino , Humanos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Riñón/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/metabolismo , Ratones Endogámicos C57BL , Apoptosis/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana , Glicoproteínas de Membrana/administración & dosificación , Proliferación Celular/efectos de los fármacos , Lesión Renal Aguda , Ratones , Nanopartículas/administración & dosificación , Nanopartículas/química
16.
J Virol ; 86(23): 12591-604, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22973034

RESUMEN

Human immunodeficiency virus (HIV)/simian immunodeficiency virus (SIV) infection causes B-cell dysregulation and the loss of memory B cells in peripheral blood mononuclear cells (PBMC). These effects are not completely reversed by antiretroviral treatment (ART). To further elucidate B-cell changes during chronic SIV infection and treatment, we investigated memory B-cell subpopulations and plasma cells/plasmablasts (PC/PB) in blood, bone marrow, and lymph nodes of rhesus macaques during ART and upon release from ART. Macaques previously immunized with SIV recombinants and the gp120 protein were included to assess the effects of prior vaccination. ART was administered for 11 weeks, with or without gp120 boosting at week 9. Naïve and resting, activated, and tissue-like memory B cells and PC/PB were evaluated by flow cytometry. Antibody-secreting cells (ASC) and serum antibody titers were assessed. No lasting changes in B-cell memory subpopulations occurred in bone marrow and lymph nodes, but significant decreases in numbers of activated memory B cells and increases in numbers of tissue-like memory B cells persisted in PBMC. Macaque PC/PB were found to be either CD27(+) or CD27(-) and therefore were defined as CD19(+) CD38(hi) CD138(+). The numbers of these PC/PB were transiently increased in both PBMC and bone marrow following gp120 boosting of the unvaccinated and vaccinated macaque groups. Similarly, ASC numbers in PBMC and bone marrow of the two macaque groups also transiently increased following envelope boosting. Nevertheless, serum binding titers against SIVgp120 remained unchanged. Thus, even during chronic SIV infection, B cells respond to antigen, but long-term memory does not develop, perhaps due to germinal center destruction. Earlier and/or prolonged treatment to allow the generation of virus-specific long-term memory B cells should benefit ART/therapeutic vaccination regimens.


Asunto(s)
Antirretrovirales/uso terapéutico , Linfocitos B/inmunología , Células de la Médula Ósea/inmunología , Memoria Inmunológica/inmunología , Macaca mulatta , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Análisis de Varianza , Animales , Antirretrovirales/inmunología , Linfocitos B/citología , Linfocitos T CD4-Positivos/inmunología , Citometría de Flujo , Modelos Lineales , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Masculino , Glicoproteínas de Membrana/administración & dosificación , Células Plasmáticas/citología , Células Plasmáticas/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/tratamiento farmacológico , Proteínas del Envoltorio Viral/administración & dosificación , Carga Viral
17.
J Immunol ; 186(6): 3309-16, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21289306

RESUMEN

A main goal of cancer immunology research is the formation of Ag-specific memory T cell immunity capable of activation upon tumor re-encounter. The requirements necessary to overcome the inhibitory signals present in the tumor microenvironment and form such memory T cell responses are unknown. In contrast to previous studies targeting tumors expressing highly immunogenic model Ags, we demonstrate that alleviating tumor-induced suppression along with vaccination against authentic Ags during the perioperative period provides long-lasting protection against a highly suppressive and poorly immunogenic melanoma. In this study, we employed DNA vaccination with an immunologically optimized mouse melanoma-shared Ag, Trp1ee/ng, combined with systemic TGF-ß blockade during the perioperative period of primary tumor resection, to confer protection against B16 melanoma, and against JBRH, an independently derived melanoma unrelated to B16. Importantly, we demonstrate that correlative to memory responses, perioperative immunotherapy increases the formation of tumor-infiltrating and tumor-reactive CD8(+) T cells expressing low levels of the transcription factor T-bet, defined as memory precursor effector cells. We show that conditions for an immunologically fertile environment are met when TGF-ß blockade and vaccination are applied during the perioperative period of primary tumor resection. These findings address limitations of current CD8(+) T cell immunotherapies against cancer by generating effective CD8(+) T cell memory recall responses.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/administración & dosificación , Memoria Inmunológica , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/cirugía , Melanoma Experimental/terapia , Glicoproteínas de Membrana/uso terapéutico , Oxidorreductasas/uso terapéutico , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/uso terapéutico , Carcinoma Pulmonar de Lewis/inmunología , Carcinoma Pulmonar de Lewis/patología , Carcinoma Pulmonar de Lewis/cirugía , Quimioterapia Combinada , Inmunización Secundaria/métodos , Memoria Inmunológica/genética , Inmunoterapia Adoptiva/métodos , Linfocitos Infiltrantes de Tumor/patología , Masculino , Melanoma Experimental/patología , Glicoproteínas de Membrana/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Oxidorreductasas/administración & dosificación , Periodo Perioperatorio/métodos , Células Madre/inmunología , Células Madre/patología , Factor de Crecimiento Transformador beta/fisiología , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética , Vacunas de ADN/uso terapéutico
18.
Curr Treat Options Oncol ; 14(4): 580-94, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23934510

RESUMEN

OPINION STATEMENT: Non-small-cell lung cancer (NSCLC) remains the most common cause of cancer-related death worldwide. Traditional cytotoxic agents and their attendant toxicities have remained the mainstay of systemic therapy for this disease, until now. With the identification of novel molecular and immune cancer-specific aberrancies, molecular agents and immunotherapies have garnered increasing attention as attractive targets, with the potential for improved outcomes while mitigating systemic toxicities seen with traditional cytotoxic agents. Despite a longstanding interest in immunotherapy for the treatment of NSCLC, results of prior studies of therapeutic vaccines have failed to show durable or convincingly meaningful clinical responses. However, newer trials of therapeutic vaccines and checkpoint inhibitors have yielded more promising results. In particular, the checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) and the programmed death-1 (PD-1) pathway have shown meaningful clinical responses with manageable toxicities. Large phase III studies are underway, the results of which have the potential to revolutionize the way in which we care for patients with NSCLC. More studies also are needed to investigate the potentially synergistic effects of traditional and immune-based therapies. Given their unique antineoplastic effects, novel immune-specific clinical endpoints also are actively being investigated.


Asunto(s)
Antineoplásicos/administración & dosificación , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Inmunoterapia , Neoplasias Pulmonares/tratamiento farmacológico , Terapia Molecular Dirigida , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/inmunología , Antígenos de Neoplasias/administración & dosificación , Antígenos de Neoplasias/inmunología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Antígeno CTLA-4/inmunología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Ensayos Clínicos como Asunto , Supervivencia sin Enfermedad , Receptores ErbB/inmunología , Femenino , Humanos , Inmunoterapia/métodos , Ipilimumab , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Masculino , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/inmunología , Proteínas de Neoplasias/administración & dosificación , Proteínas de Neoplasias/inmunología , Nivolumab , Receptor de Muerte Celular Programada 1/inmunología
19.
Virol J ; 9: 215, 2012 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-22995185

RESUMEN

BACKGROUND: Sublingual (s.l.) administration of soluble protein antigens, inactivated viruses, or virus-like particles has been shown to induce broad immune responses in mucosal and extra-mucosal tissues. Recombinant replication-defective adenovirus vectors (rADVs) infect mucosa surface and therefore can serve as a mucosal antigen delivery vehicle. In this study we examined whether s.l. immunization with rADV encoding spike protein (S) (rADV-S) of severe acute respiratory syndrome-associated coronavirus (SARS-CoV) induces protective immunity against SARS-CoV and could serve as a safe mucosal route for delivery of rADV. RESULTS: Here, we show that s.l. administration of rADV-S induced serum SARS-CoV neutralizing and airway IgA antibodies in mice. These antibody responses are comparable to those induced by intranasal (i.n.) administration. In addition, s.l. immunization induced antigen-specific CD8+ T cell responses in the lungs that are superior to those induced by intramuscular immunization. Importantly, unlike i.n. administration, s.l. immunization with rADV did not redirect the rADV vector to the olfactory bulb. CONCLUSION: Our study indicates that s.l. immunization with rADV-S is safe and effective in induction of a broad spectrum of immune responses and presumably protection against infection with SARS-CoV.


Asunto(s)
Encéfalo/virología , Glicoproteínas de Membrana/inmunología , Síndrome Respiratorio Agudo Grave/inmunología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/inmunología , Proteínas del Envoltorio Viral/inmunología , Administración Sublingual , Animales , Anticuerpos Antivirales/inmunología , Encéfalo/inmunología , Femenino , Humanos , Inmunidad Mucosa , Inmunización , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/genética , Síndrome Respiratorio Agudo Grave/virología , Glicoproteína de la Espiga del Coronavirus , Proteínas del Envoltorio Viral/administración & dosificación , Proteínas del Envoltorio Viral/genética , Vacunas Virales/administración & dosificación , Vacunas Virales/genética , Vacunas Virales/inmunología
20.
Immunol Invest ; 41(3): 249-60, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22221010

RESUMEN

DNA vaccines have been widely used to induce immune responses against molecular targets. In this study, we explored the possibility of using DNA vaccine combined with the immunosuppressant FK506 (tacrolimus) to antigen-specifically suppress unwanted immune responses and prevent autoimmune ovarian disease. To that end, we immunized C57BL/6 mice with a DNA vaccine encoding mouse zona pellucida 3 (ZP3) together with FK506. The immunization induced ZP3-specific CD4(+)CD25(+)Foxp3(+) regulatory T cells (Treg), which suppressed the induction of ZP3-specific delayed-type hypersensitivity in the animals. Significantly, the immunization also protected the animals from experimentally induced autoimmune ovarian disease. These results suggest that DNA vaccination in the presence of FK506 may be used to induce Treg cells and prevent AOD.


Asunto(s)
Enfermedades Autoinmunes/prevención & control , Proteínas del Huevo/metabolismo , Hipersensibilidad Tardía/inmunología , Glicoproteínas de Membrana/metabolismo , Enfermedades del Ovario/prevención & control , Receptores de Superficie Celular/metabolismo , Linfocitos T Reguladores/metabolismo , Vacunas de ADN/administración & dosificación , Animales , Enfermedades Autoinmunes/inmunología , Antígenos CD4/metabolismo , Células Cultivadas , Proteínas del Huevo/administración & dosificación , Proteínas del Huevo/genética , Proteínas del Huevo/inmunología , Femenino , Factores de Transcripción Forkhead/metabolismo , Hipersensibilidad Tardía/inducido químicamente , Hipersensibilidad Tardía/prevención & control , Tolerancia Inmunológica , Inmunización , Inmunosupresores/administración & dosificación , Inmunosupresores/efectos adversos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Glicoproteínas de Membrana/administración & dosificación , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Ratones Endogámicos C57BL , Enfermedades del Ovario/inmunología , Receptores de Superficie Celular/administración & dosificación , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Tacrolimus/administración & dosificación , Tacrolimus/efectos adversos , Glicoproteínas de la Zona Pelúcida
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA