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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cancer Immunol Immunother ; 69(5): 703-716, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32034426

RESUMEN

Cancer is one of the main causes of mortality worldwide and a major public health concern. Among various strategies, therapeutic vaccines have been developed to stimulate anti-tumoral immune responses. However, in spite of extensive studies, this approach suffers from a lack of efficacy. Recently, we designed the MAG-Tn3 vaccine, aiming to induce antibody responses against Tn, a tumor-associated carbohydrate antigen. The Tn antigen is of interest because it is expressed by several adenocarcinomas, but not normal cells. The fully synthetic glycopeptide vaccine MAG-Tn3 is composed of four arms built on three adjacent Tn moieties associated with the tetanus toxin-derived peptide TT830-844 CD4+ T-cell epitope. This promiscuous CD4+ T-cell epitope can bind to a wide range of HLA-DRB molecules and is thus expected to activate CD4+ T-cell responses in a large part of the human population. The MAG-Tn3 vaccine was formulated with the GSK-proprietary immunostimulant AS15, composed of CpG7909, MPL, and QS21, which has been shown to stimulate both innate and humoral responses, in addition to being well tolerated. Here, seven patients with localized breast cancer with a high-risk of relapse were immunized with the MAG-Tn3 vaccine formulated with AS15. The first results of phase I clinical trial demonstrated that all vaccinated patients developed high levels of Tn-specific antibodies. Moreover, these antibodies specifically recognized Tn-expressing human tumor cells and killed them through a complement-dependent cytotoxicity mechanism. Overall, this study establishes, for the first time, the capacity of a fully synthetic glycopeptide cancer vaccine to induce specific immune responses in humans.


Asunto(s)
Anticuerpos Antineoplásicos/sangre , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Neoplasias de la Mama/terapia , Vacunas contra el Cáncer/inmunología , Recurrencia Local de Neoplasia/prevención & control , Adyuvantes Inmunológicos/administración & dosificación , Adulto , Anciano , Animales , Anticuerpos Antineoplásicos/inmunología , Antígenos de Carbohidratos Asociados a Tumores/administración & dosificación , Antígenos de Carbohidratos Asociados a Tumores/genética , Neoplasias de la Mama/sangre , Neoplasias de la Mama/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/genética , Femenino , Glicopéptidos/administración & dosificación , Glicopéptidos/genética , Glicopéptidos/inmunología , Humanos , Inmunogenicidad Vacunal , Inyecciones Intramusculares , Células Jurkat , Persona de Mediana Edad , Recurrencia Local de Neoplasia/inmunología , Resultado del Tratamiento , Vacunación/métodos , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología
2.
Cancer Immunol Immunother ; 65(3): 315-25, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26847142

RESUMEN

Malignant transformations are often associated with aberrant glycosylation processes that lead to the expression of new carbohydrate antigens at the surface of tumor cells. Of these carbohydrate antigens, the Tn antigen is particularly highly expressed in many carcinomas, especially in breast carcinoma. We designed MAG-Tn3, a fully synthetic vaccine based on three consecutive Tn moieties that are O-linked to a CD4+ T cell epitope, to induce anti-Tn antibody responses that could be helpful for therapeutic vaccination against cancer. To ensure broad coverage within the human population, the tetanus toxoid-derived peptide TT830-844 was selected as a T-helper epitope because it can bind to various HLA-DRB molecules. We showed that the MAG-Tn3 vaccine, which was formulated with the GSK proprietary immunostimulant AS15 and designed for human cancer therapy, is able to induce an anti-Tn antibody response in mice of various H-2 haplotypes, and this response correlates with the ability to induce a specific T cell response against the TT830-844 peptide. The universality of the TT830-844 peptide was extended to new H-2 and HLA-DRB molecules that were capable of binding this T cell epitope. Finally, the MAG-Tn3 vaccine was able to induce anti-Tn antibody responses in cynomolgus monkeys, which targeted Tn-expressing tumor cells and mediated tumor cell death both in vitro and in vivo. Thus, MAG-Tn3 is a highly promising anticancer vaccine that is currently under evaluation in a phase I clinical trial.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/inmunología , Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/inmunología , Epítopos de Linfocito T/inmunología , Glicoproteína Asociada a Mielina/inmunología , Fragmentos de Péptidos/inmunología , Toxoide Tetánico/inmunología , Secuencia de Aminoácidos , Animales , Femenino , Antígenos H-2/genética , Cadenas HLA-DRB1/inmunología , Haplotipos , Humanos , Macaca fascicularis , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Vacunación , Vacunas Sintéticas/inmunología
3.
Org Biomol Chem ; 15(1): 114-123, 2016 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-27812586

RESUMEN

Herein, we report a new process that enables the gram-scale production of a fully synthetic anti-cancer vaccine for human use. This therapeutic vaccine candidate, named MAG-Tn3, is a high-molecular-weight tetrameric glycopeptide encompassing carbohydrate tumor-associated Tn antigen clusters and peptidic CD4+ T-cell epitopes. The synthetic process involves (i) the stepwise solid-phase assembly of protected amino acids, including the high value-added Tn building blocks with only 1.5 equivalents, (ii) a single isolated intermediate, and (iii) the simultaneous deprotection of 36 hindered protective groups. The resulting MAG-Tn3 was unambiguously characterized using a combination of techniques, including a structural analysis by nuclear magnetic resonance spectroscopy. The four peptidic chains are flexible in solution, with a more constrained but extended conformation at the Tn3 antigen motif. Finally, we demonstrate that, when injected into HLA-DR1-expressing transgenic mice, this vaccine induces Tn-specific antibodies that mediate the killing of human Tn-positive tumor cells. These studies led to a clinical batch of the MAG-Tn3, currently investigated in breast cancer patients (phase I clinical trial). The current study demonstrates the feasibility of the multigram-scale synthesis of a highly pure complex glycopeptide, and it opens new avenues for the use of synthetic glycopeptides as drugs in humans.


Asunto(s)
Vacunas contra el Cáncer/química , Dendrímeros/química , Glicopéptidos/química , Neoplasias/prevención & control , Vacunas Sintéticas/química , Secuencia de Aminoácidos , Animales , Antígenos de Carbohidratos Asociados a Tumores/química , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Linfocitos T CD4-Positivos/inmunología , Vacunas contra el Cáncer/síntesis química , Vacunas contra el Cáncer/uso terapéutico , Dendrímeros/síntesis química , Dendrímeros/uso terapéutico , Glicopéptidos/síntesis química , Glicopéptidos/uso terapéutico , Humanos , Ratones , Ratones Transgénicos , Neoplasias/inmunología , Vacunas Sintéticas/uso terapéutico
4.
Therapie ; 79(1): 35-45, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38105120

RESUMEN

This round table is the result of an observation. The observation being that controlled human infection clinical trials (also called "infectious challenge" trials or "Controlled Human Infection Models", "CHIM") recommended or even encouraged in the context of vaccine developments in particular, are not carried out in France. However, there are no formal prohibitions within regulations or ethical principles, which point to the prior assessment of risks and benefits for individuals and for society. The participants in this Round Table thus wished to examine, through the prism of their respective disciplines, the scientific and medical relevance of conducting such trials in France and, if possible, to imagine the conditions under which they would be carried out, thus resulting in recommendations on (1) the advisability of their conduct in France (2), the conditions under which they would be implemented in terms of logistics and regulations, and (3) their social acceptability. The recommendations on which the participants of the Round Table came to an agreement are presented as the analysis progresses.


Asunto(s)
Ensayos Clínicos como Asunto , Infecciones , Humanos , Francia , Ensayos Clínicos como Asunto/ética , Ensayos Clínicos como Asunto/legislación & jurisprudencia
5.
EBioMedicine ; 75: 103810, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35045362

RESUMEN

BACKGROUND: V591 (TMV-083) is a live recombinant measles vector-based vaccine candidate expressing a pre-fusion stabilized SARS-CoV-2 spike protein. METHODS: We performed a randomized, placebo-controlled Phase I trial with an unblinded dose escalation and a double-blind treatment phase at 2 sites in France and Belgium to evaluate the safety and immunogenicity of V591. Ninety healthy SARS-CoV-2 sero-negative adults (18-55 years of age) were randomized into 3 cohorts, each comprising 24 vaccinees and 6 placebo recipients. Participants received two intramuscular injections of a low dose vaccine (1 × 105 median Tissue Culture Infectious Dose [TCID50]), one or two injections of a high dose vaccine (1 × 106 TCID50), or placebo with a 28 day interval. Safety was assessed by solicited and unsolicited adverse events. Immunogenicity was measured by SARS-CoV-2 spike protein-binding antibodies, neutralizing antibodies, spike-specific T cell responses, and anti-measles antibodies. ClinicalTrials.gov, NCT04497298. FINDINGS: Between Aug 10 and Oct 13, 2020, 148 volunteers were screened of whom 90 were randomized. V591 showed a good safety profile at both dose levels. No serious adverse events were reported. At least one treatment-related adverse event was reported by 15 (20.8%) participants receiving V591 vs. 6 (33.3%) of participants receiving placebo. Eighty-one percent of participants receiving two injections of V591 developed spike-binding antibodies after the second injection. However, neutralizing antibodies were detectable on day 56 only in 17% of participants receiving the low dose and 61% receiving the high dose (2 injections). Spike-specific T cell responses were not detected. Pre-existing anti-measles immunity had a statistically significant impact on the immune response to V591, which was in contrast to previous results with the measles vector-based chikungunya vaccine. INTERPRETATION: While V591 was generally well tolerated, the immunogenicity was not sufficient to support further development. FUNDING: Themis Bioscience GmbH, a subsidiary of Merck & Co. Inc., Kenilworth, NJ, USA; Coalition for Epidemic Preparedness Innovations (CEPI).


Asunto(s)
Vacunas contra la COVID-19/administración & dosificación , COVID-19/inmunología , Vectores Genéticos , Inmunogenicidad Vacunal , Virus del Sarampión , SARS-CoV-2/inmunología , Adolescente , Adulto , COVID-19/genética , COVID-19/prevención & control , Vacunas contra la COVID-19/genética , Vacunas contra la COVID-19/inmunología , Método Doble Ciego , Femenino , Humanos , Masculino , Persona de Mediana Edad , SARS-CoV-2/genética
6.
Front Immunol ; 12: 655478, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34040605

RESUMEN

Mucopolysaccharidosis type IIIB syndrome (Sanfilippo disease) is a rare autosomic recessif disorder caused by mutations in the α-N-acetylglucosaminidase (NAGLU) gene coding for a lysosomal enzyme, leading to neurodegeneration and progressive deterioration of cognitive abilities in affected children. To supply the missing enzyme, several recent human gene therapy trials relied on the deposit of adeno-associated virus (AAV) vectors directly into the brain. We reported safety and efficacy of an intracerebral therapy in a phase 1/2 clinical trial (https://clinicaltrials.gov/ct2/show/NCT03300453), with a recombinant AAV serotype 2/5 (rAAV2/5) coding human NAGLU in four children with MPS IIIB syndrome receiving immunosuppression. It was reported that AAV-mediated gene therapies might elicit a strong host immune response resulting in decreased transgene expression. To address this issue, we performed a comprehensive analysis of cellular immunity and cytokine patterns generated against the therapeutic enzyme in the four treated children over 5.5 years of follow-up. We report the emergence of memory and polyfunctional CD4+ and CD8+ T lymphocytes sensitized to the transgene soon after the start of therapy, and appearing in peripheral blood in waves throughout the follow-up. However, this response had no apparent impact on CNS transgene expression, which remained stable 66 months after surgery, possibly a consequence of the long-term immunosuppressive treatment. We also report that gene therapy did not trigger neuroinflammation, evaluated through the expression of cytokines and chemokines in patients' CSF. Milder disease progression in the youngest patient was found associated with low level and less differentiated circulating NAGLU-specific T cells, together with the lack of proinflammatory cytokines in the CSF. Findings in this study support a systematic and comprehensive immunomonitoring approach for understanding the impact immune reactions might have on treatment safety and efficacy of gene therapies.


Asunto(s)
Acetilglucosaminidasa/inmunología , Terapia Genética/efectos adversos , Vectores Genéticos/efectos adversos , Inmunidad Celular , Mucopolisacaridosis III/complicaciones , Transgenes/inmunología , Acetilglucosaminidasa/genética , Niño , Citocinas/metabolismo , Vías de Administración de Medicamentos , Terapia Genética/métodos , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Humanos , Memoria Inmunológica , Activación de Linfocitos , Mucopolisacaridosis III/genética , Mucopolisacaridosis III/terapia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transgenes/genética
7.
Lancet Infect Dis ; 21(4): 546-558, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33186516

RESUMEN

BACKGROUND: Shigella remains in the top four pathogens responsible for moderate to severe diarrhoea in children below 5 years of age. The shigella O-specific polysaccharide (O-SP) is a promising vaccine target. We developed a conjugate vaccine prototype incorporating a unique well defined synthetic oligosaccharide hapten, chemically designed for optimal antigenic, conformational, structural, and functional mimicry of the O-SP from Shigella flexneri 2a (SF2a). We aimed to assess the safety, tolerability, and immunogenicity of this original synthetic oligosaccharide-based vaccine candidate, SF2a-TT15, conceived to drive the antibody response towards the key protective determinants of the native lipopolysaccharide antigen, in a first-in-human phase 1 study. METHODS: We did a first-in-human, dose-escalating, single-blind, observer-masked, randomised, placebo-controlled study at the Clinical Research Center of Tel Aviv Sourasky Medical Center (Israel). Participants were healthy adults aged 18-45 years with low titres of serum SF2a-specific IgG antibodies. 64 eligible participants were assigned to one of two cohorts. 32 participants in each of the two cohorts were randomly assigned via computer-generated algorithm in a stepwise manner to receive the 2 µg (cohort 1) and 10 µg oligosaccharide dose (cohort 2) of the SF2a-TT15 vaccine candidate non-adjuvanted or adjuvanted with aluminium hydroxide (alum) or matching placebos. The vaccine was administered as three single intramuscular injections into the arm, 28 days apart. The primary outcome was the incidence and severity of adverse events, which were assessed in the intention-to-treat safety population analysis including all participants who were randomly assigned and received at least one vaccine or placebo injection. The immunogenicity endpoints were secondary outcomes and were analysed in all participants who were randomly assigned, received all of the assigned injections before the time of the immunogenicity assessment, and provided blood samples for immunological follow-up (per-protocol immunogenicity analysis). The study is registered with ClinicalStudies.gov, NCT02797236 and is completed. FINDINGS: Of 203 volunteers initially screened, 64 participants were enrolled between Sept 20, 2016, and Sept 26, 2017. In each of the two cohorts, 12 participants received the adjuvanted vaccine, 12 received the non-adjuvanted vaccine and eight received the matching placebo (four each). The SF2a-TT15 glycoconjugate was well tolerated at both doses. No serious or severe adverse events occurred. Overall, seven (88%) of eight to 12 (100%) of 12 in each group of volunteers had one adverse event or more after receiving the study agents with the majority of adverse events, 300 (98%) of 307, considered mild in intensity. Of the seven adverse events defined as moderate in severity, one (nausea) was suspected to be related to the vaccine candidate. At all post-immunisation days and for both oligosaccharide doses, whether adjuvanted or not, SF2a-TT15 induced significantly higher serum IgG anti-SF2a lipopolysaccharide geometric mean titres (GMTs) as compared with baseline or with the corresponding GMTs in placebo recipients (p<0·01). After one injection, the non-adjuvanted 10 µg oligosaccharide dose induced a 27-times increase in IgG GMT (5080 vs 189) and the non-adjuvanted 2 µg oligosaccharide dose induced a five-times increase (1411 vs 283), compared with baseline. Alum enhanced the specific IgG response at 2 µg oligosaccharide dose after the third injection (GMTs 3200 vs 1176, p=0.045). INTERPRETATION: SF2a-TT15 was safe and well tolerated and induced high titres of anti-SF2a LPS IgG antibodies. These results support further evaluation of this original synthetic oligosaccharide-protein conjugate vaccine candidate for safety, immunogenicity, and protective efficacy in target populations. FUNDING: The European Union Seventh Framework Programme.


Asunto(s)
Disentería Bacilar/prevención & control , Inmunogenicidad Vacunal , Vacunas contra la Shigella/efectos adversos , Shigella flexneri/inmunología , Adyuvantes Inmunológicos/administración & dosificación , Adyuvantes Inmunológicos/efectos adversos , Adolescente , Adulto , Hidróxido de Aluminio/administración & dosificación , Hidróxido de Aluminio/efectos adversos , Hidróxido de Aluminio/inmunología , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Relación Dosis-Respuesta Inmunológica , Disentería Bacilar/inmunología , Disentería Bacilar/microbiología , Femenino , Voluntarios Sanos , Humanos , Inyecciones Intramusculares , Masculino , Persona de Mediana Edad , Antígenos O/genética , Antígenos O/inmunología , Vacunas contra la Shigella/administración & dosificación , Vacunas contra la Shigella/genética , Vacunas contra la Shigella/inmunología , Método Simple Ciego , Vacunas Conjugadas/administración & dosificación , Vacunas Conjugadas/efectos adversos , Vacunas Conjugadas/genética , Vacunas Conjugadas/inmunología , Vacunas Sintéticas/administración & dosificación , Vacunas Sintéticas/efectos adversos , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Adulto Joven
8.
Methods Mol Biol ; 2013: 165-176, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31267501

RESUMEN

Vaccines are biological pharmaceutical products prescribed as prevention of a hypothetical infection. Development of a new vaccine is the result of a long process involving several stages. During all developmental phases, priority is the safety of the new product, which is often used in young infants. The initial research phase lasts 1-5 years and is followed by a clinical and pharmaceutical development phase (preclinical and clinical phases), which can last from 15 to 20 years on average before licensure is obtained. There are, however, exceptions, like the malaria vaccine for which research has been going on for more than 30 years and at least 30 candidate vaccines have been assessed. This chapter summarizes the different phases of vaccine candidate development from preclinical studies to phase 2 vaccine trials.


Asunto(s)
Vacunas contra la Malaria/efectos adversos , Vacunas contra la Malaria/uso terapéutico , Ensayos Clínicos Fase I como Asunto , Ensayos Clínicos Fase II como Asunto , Humanos
10.
Trials ; 19(1): 666, 2018 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-30514364

RESUMEN

BACKGROUND: The aim of this open-label, randomized controlled trial conducted in four African countries (Madagascar, Niger, Central African Republic, and Senegal) is to compare three strategies of renutrition for moderate acute malnutrition (MAM) in children based on modulation of the gut microbiota with enriched flours alone, enriched flours with prebiotics or enriched flours coupled with antibiotic treatment. METHODS: To be included, children aged between 6 months and 2 years are preselected based on mid-upper-arm circumference (MUAC) and are included based on a weight-for-height Z-score (WHZ) between - 3 and - 2 standard deviations (SD). As per current protocols, children receive renutrition treatment for 12 weeks and are assessed weekly to determine improvement. The primary endpoint is recovery, defined by a WHZ ≥ - 1.5 SD after 12 weeks of treatment. Data collected include clinical and socioeconomic characteristics, side effects, compliance and tolerance to interventions. Metagenomic analysis of gut microbiota is conducted at inclusion, 3 months, and 6 months. The cognitive development of children is evaluated in Senegal using only the Developmental Milestones Checklist II (DMC II) questionnaire at inclusion and at 3, 6, and 9 months. The data will be correlated with renutrition efficacy and metagenomic data. DISCUSSION: This study will provide new insights for the treatment of MAM, as well as original data on the modulation of gut microbiota during the renutrition process to support (or not) the microbiota hypothesis of malnutrition. TRIAL REGISTRATION: ClinicalTrials.gov, ID: NCT03474276 Last update 28 May 2018.


Asunto(s)
Antibacterianos/administración & dosificación , Azitromicina/administración & dosificación , Harina , Alimentos Fortificados , Microbioma Gastrointestinal/efectos de los fármacos , Trastornos de la Nutrición del Lactante/terapia , Fenómenos Fisiológicos Nutricionales del Lactante , Desnutrición/terapia , Estado Nutricional , Prebióticos/administración & dosificación , Enfermedad Aguda , África , Factores de Edad , Albendazol/administración & dosificación , Antibacterianos/efectos adversos , Antiparasitarios/administración & dosificación , Azitromicina/efectos adversos , Desarrollo Infantil , Preescolar , Femenino , Harina/efectos adversos , Alimentos Fortificados/efectos adversos , Humanos , Lactante , Trastornos de la Nutrición del Lactante/diagnóstico , Trastornos de la Nutrición del Lactante/microbiología , Trastornos de la Nutrición del Lactante/fisiopatología , Masculino , Desnutrición/diagnóstico , Desnutrición/microbiología , Desnutrición/fisiopatología , Estudios Multicéntricos como Asunto , Prebióticos/efectos adversos , Ensayos Clínicos Controlados Aleatorios como Asunto , Factores de Tiempo , Resultado del Tratamiento
11.
Lancet Neurol ; 16(9): 712-720, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28713035

RESUMEN

BACKGROUND: Mucopolysaccharidosis type IIIB syndrome (also known as Sanfilippo type B syndrome) is a lysosomal storage disease resulting in progressive deterioration of cognitive acquisition after age 2-4 years. No treatment is available for the neurological manifestations of the disease. We sought to assess the safety and efficacy of a novel intracerebral gene therapy. METHODS: Local regulatory authorities in France allowed inclusion of up to four children in this phase 1/2 study. Treatment was 16 intraparenchymal deposits (four in the cerebellum) of a recombinant adenoassociated viral vector serotype 2/5 (rAAV2/5) encoding human α-N-acetylglucosaminidase (NAGLU) plus immunosuppressive therapy. We assessed tolerance, neurocognitive progression, brain growth, NAGLU enzymatic activity in CSF, and specific anti-NAGLU immune response for 30 months after surgery. This trial is registered with EudraCT, number 2012-000856-33, and the International Standard Clinical Trial Registry, number ISRCTN19853672. FINDINGS: Of seven eligible children, the four youngest, from France (n=2), Italy (n=1), and Greece (n=1), aged 20, 26, 30, and 53 months, were included between February, 2012, and February, 2014. 125 adverse events were recorded, of which 117 were treatment emergent and included six classified as severe, but no suspected unexpected serious adverse drug reactions were seen. Vector genomes were detected in blood for 2 days after surgery. Compared with the natural history of mucopolysaccharidosis type III syndromes, neurocognitive progression was improved in all patients, with the youngest patient having function close to that in healthy children. Decrease in developmental quotient was -11·0 points in patient one, -23·0 in patient two, -29·0 in patient three, and -17·0 in patient four, compared with -37·7 in the natural history of the disease. NAGLU activity was detected in lumbar CSF and was 15-20% of that in unaffected children. Circulating T lymphocytes that proliferated and produced tumour necrosis factor α upon ex-vivo exposure to NAGLU antigens were detectable at 1-12 months and 3-12 months, respectively, but not at 30 months in three of four patients. INTERPRETATION: Intracerebral rAVV2/5 was well tolerated and induced sustained enzyme production in the brain. The initial specific anti-NAGLU immune response that later subsided suggested acquired immunological tolerance. The best results being obtained in the youngest patient implies a potential window of opportunity. Longer follow-up is needed to further assess safety outcomes and persistence of improved cognitive development. FUNDING: Association Française Contre les Myopathies, Vaincre les Maladies Lysosomales, Institut Pasteur, and UniQure.


Asunto(s)
Acetilglucosaminidasa , Encéfalo/enzimología , Dependovirus/genética , Terapia Genética/métodos , Vectores Genéticos/farmacología , Mucopolisacaridosis III/terapia , Evaluación de Resultado en la Atención de Salud , Acetilglucosaminidasa/genética , Preescolar , Terapia Genética/efectos adversos , Vectores Genéticos/administración & dosificación , Humanos , Inmunosupresores/uso terapéutico , Lactante , Mucopolisacaridosis III/tratamiento farmacológico , Síndrome
12.
Int J Mol Med ; 17(6): 1045-52, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16685414

RESUMEN

Carbohydrate mimetic peptides of tumor associated carbohydrate antigens (TACA) are T-cell-dependent antigens and, therefore, immunization with these surrogates is predicted to overcome the low immunogenicity of carbohydrate antigens. Consistent with this hypothesis, we show that among the potential immune cells involved, peptide immunization led to an increase in T-cell populations. While peptide mimetics may also function as TLR binding ligands, we did not observe evidence of involvement of NK cells. Examining tumor challenged animals, we observed that peptide immunization and not tumor cells rendered IL-12 responsiveness to T-cells, as T-cells from peptide-immunized mice produced IFN-gamma upon stimulation with IL-12. Cyclophosphamide administration enhanced the anti-tumor efficacy of the vaccine, which was achieved by enhancing T-cell responses with no effect on NK cell population. Prophylactic immunization of mice with a DNA construct encoding carbohydrate mimetic peptides indicated a specific role for the mimotope vaccine in anti-tumor immune responses. These data suggest a role for both CD4(+) and CD8(+) T-cells induced by mimotopes of TACA in protective immunity against tumor cells.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas contra el Cáncer/inmunología , Péptidos/inmunología , Sarcoma/prevención & control , Secuencia de Aminoácidos , Animales , Antígenos de Carbohidratos Asociados a Tumores/genética , Antineoplásicos Alquilantes/administración & dosificación , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD8-positivos/efectos de los fármacos , Vacunas contra el Cáncer/uso terapéutico , Carbohidratos/inmunología , Ciclofosfamida/administración & dosificación , Femenino , Interleucina-12/farmacología , Ratones , Ratones Endogámicos BALB C , Imitación Molecular/inmunología , Datos de Secuencia Molecular , Péptidos/genética , Vacunas de ADN/inmunología , Vacunas de ADN/uso terapéutico , Vacunas de Subunidad/inmunología , Vacunas de Subunidad/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cancer Res ; 64(6): 2162-6, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15026358

RESUMEN

Tumor-associated carbohydrate antigens are typically perceived as inadequate targets for generating tumor-specific cellular responses. Lectin profile reactivity and crystallographic studies demonstrate that MHC class I molecules can present to the immune system posttranslationally modified cytosolic peptides carrying O-beta-linked N-acetylglucosamine (GlcNAc). Here we report that a peptide surrogate of GlcNAc can facilitate an in vivo tumor-specific cellular response to established Meth A tumors that display native O-GlcNAc glycoproteins on the tumor cell surface. Peptide immunization of tumor-bearing mice had a moderate effect on tumor regression. Inclusion of interleukin 12 in the immunization regimen stimulated complete elimination of tumor cells in all of the mice tested, whereas interleukin 12 administration alone afforded no tumor growth inhibition. Adoptive transfer of immune T cells into tumor-bearing nude mice indicates a role for CD8+ T cells in tumor regression. This work postulates that peptide mimetics of glycosylated tumor rejection antigens might be further developed for immune therapy of cancer.


Asunto(s)
Acetilglucosamina/inmunología , Antígenos de Carbohidratos Asociados a Tumores/uso terapéutico , Antígenos de Histocompatibilidad Clase I/metabolismo , Imitación Molecular , Fragmentos de Péptidos/uso terapéutico , Sarcoma Experimental/prevención & control , Animales , Presentación de Antígeno , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Antígenos de Carbohidratos Asociados a Tumores/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Citosol/metabolismo , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Inmunización , Inmunoterapia , Interferón gamma/metabolismo , Interleucina-12/uso terapéutico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Fragmentos de Péptidos/inmunología , Fragmentos de Péptidos/metabolismo , Sarcoma Experimental/inmunología , Sarcoma Experimental/patología , Bazo/inmunología , Linfocitos T Citotóxicos/efectos de los fármacos , Linfocitos T Citotóxicos/inmunología
14.
Int J Oncol ; 27(1): 257-63, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15942667

RESUMEN

End-products of glycolysis as well as phospholipid precursors and catabolites have been suggested as metabolic indicators of tumor progression. To test the hypothesis that increased levels of such indicators can distinguish metastatic phenotypes, we determined a limited cellular 1H-NMR metabolic profile of subpopulations of murine mammary 4T1 cells that differ in their metastatic potential. Subpopulations with differing metastatic phenotypes were identified by sorting for the expression of the cell surface adhesion oligosaccharide sialylated Lewis x (sLeX). The sLeX-negative subpopulation metastasizes to the lung of syngeneic mice more rapidly than the sLeX-positive subpopulations. The metabolic profile of the sLeX-negative subpopulation indicated higher levels of lactate and total choline metabolites than the sLeX-positive subpopulation, suggesting that altered metabolism is a critical component of the malignant phenotype. Analysis of shed cellular material from the sLeX-negative subpopulation displayed an increased ratio of phosphocholine to glycerophosphocholine when compared to the parental line and sLeX-positive subpopulation. Serum obtained from mice inoculated with either sLeX-negative or sLeX-positive tumor cells contained broader methylene resonances (P = 0.0002; P = 0.0003) and narrower methyl resonances (P = 0.0013; P < 0.0001) when compared to serum of naive mice. However, line widths of methylene and methyl resonances were not useful for distinguishing between the two tumor phenotypes. Results of this study further support the notion that metabolic indicators of malignancy can correlate with in vivo metastatic behavior.


Asunto(s)
Espectroscopía de Resonancia Magnética/métodos , Neoplasias Mamarias Animales/patología , Animales , Antígenos de Neoplasias , Biomarcadores , Biomarcadores de Tumor , Adhesión Celular , Proliferación Celular , Separación Celular , Citometría de Flujo , Glucólisis , Humanos , Hidrocarburos , Neoplasias Mamarias Animales/metabolismo , Metano/análogos & derivados , Metano/química , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Oligosacáridos/química , Fenotipo , Fosforilcolina/química , Pronóstico , Antígeno Sialil Lewis X , Factores de Tiempo
15.
Cancers (Basel) ; 3(4): 4151-69, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-24213131

RESUMEN

Among the most challenging of clinical targets for cancer immunotherapy are Tumor Associated Carbohydrate Antigens (TACAs). To augment immune responses to TACA we are developing carbohydrate mimetic peptides (CMPs) that are sufficiently potent to activate broad-spectrum anti-tumor reactivity. However, the activation of immune responses against terminal mono- and disaccharide constituents of TACA raises concerns regarding the balance between "tumor destruction" and "tissue damage", as mono- and disaccharides are also expressed on normal tissue. To support the development of CMPs for clinical trial testing, we demonstrate in preclinical safety assessment studies in mice that vaccination with CMPs can enhance responses to TACAs without mediating tissue damage to normal cells expressing TACA. BALB/c mice were immunized with CMPs that mimic TACAs reactive with Griffonia simplicifolia lectin 1 (GS-I), and tissue reactivity of serum antibodies were compared with the tissue staining profile of GS-I. Tissues from CMP immunized mice were analyzed using hematoxylin and eosin stain, and Luxol-fast blue staining for myelination. Western blots of membranes from murine mammary 4T1 cells, syngeneic with BALB/c mice, were also compared using GS-I, immunized serum antibodies, and naive serum antibodies. CMP immunization enhanced glycan reactivities with no evidence of pathological autoimmunity in any immunized mice demonstrating that tissue damage is not an inevitable consequence of TACA reactive responses.

16.
Int J Oncol ; 37(3): 615-22, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20664930

RESUMEN

Aberrant glycosylation is a universal feature of cancer cells, and certain glycan structures are well-known markers for tumor progression. Availability and composition of sugars in the microenvironment may affect cell glycosylation. Recent studies of human breast tumor cell lines indicate their ability to take up and utilize fructose. Here we tested the hypothesis that adding fructose to culture as a carbon source induces phenotypic changes in cultured human breast tumor cells that are associated with metastatic disease. MDA-MB-468 cells were adapted to culture media in which fructose was substituted for glucose. Changes in cell surface glycan structures, expression of genes related to glycan assembly, cytoskeleton F-actin, migration, adhesion and invasion were determined. Cells cultured in fructose expressed distinct cell-surface glycans. The addition of fructose affected sialylation and fucosylation patterns. Fructose feeding also increased binding of leukoagglutinating Phaseolus vulgaris isolectin, suggesting a possible rise in expression of branching beta-1, 6 GlcNAc structures. Rhodamine-phalloidin staining revealed an altered F-actin cytoskeletal system. Fructose accelerated cellular migration and increased invasion. These data suggest that changing the carbon source of the less aggressive MDA-MB-468 cell line induced characteristics associated with more aggressive phenotypes. These data could be of fundamental importance due to the markedly increased consumption of sweeteners containing free fructose in recent years, as they suggest that the presence of fructose in nutritional microenvironment of tumor cells may negatively affect the outcome for some breast cancer patients.


Asunto(s)
Neoplasias de la Mama/metabolismo , Carbono/metabolismo , Fructosa/metabolismo , Actinas/genética , Actinas/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Movimiento Celular/fisiología , Femenino , Fructosa/administración & dosificación , Glicosilación , Humanos , Microscopía de Contraste de Fase , Invasividad Neoplásica , Fenotipo , Lectinas de Plantas/química , Lectinas de Plantas/metabolismo , Especificidad por Sustrato , Células Tumorales Cultivadas
17.
Int J Cancer ; 120(6): 1179-91, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-17154173

RESUMEN

The metastatic breast cancer cell line, 4T1, abundantly expresses the oligosaccharide sialylated Lewis x (sLe(x)). SLe(x) oligosaccharide on tumor cells can be recognized by E- and P-selectin, contributing to tumor metastatic process. We observed that both selectins reacted with this cell line. However, contrary to the E-selectin reactivity, which was sLe(x) dependent, P-selectin reactivity with this cell line was sLe(x)-independent. The sLe(x)-Neg variant of the 4T1 cell line with markedly diminished expression of sLe(x) and lack of sLe(a), provided a unique opportunity to characterize P-selectin ligands and their contribution to metastasis in the absence of overlapping selectin ligands and E-selectin binding. We observed that P-selectin binding was Ca(2+)-independent and sulfation-dependent. We found that P-selectin reacted primarily with cell surface chondroitin sulfate (CS) proteoglycans, which were abundantly and stably expressed on the surface of the 4T1 cell line. P-selectin binding to the 4T1 cells was inhibited by heparin and CS glycosaminoglycans (GAGs). Moreover, Heparin administration significantly inhibited experimental lung metastasis. In addition, the data suggest that surface CS GAG chains were involved in P-selectin mediated adhesion of the 4T1 cells to murine platelets and human umbilical vein endothelial cells. The data suggest that CS GAGs are also the major P-selectin-reactive ligands on the surface of human MDA-MET cells. The results warrant conducting clinical studies on the involvement of cell surface CS chains in breast cancer metastasis and evaluation of various CS types and their biosynthetic pathways as target for development of treatment strategies for antimetastatic therapy of this disease.


Asunto(s)
Neoplasias de la Mama/patología , Sulfatos de Condroitina/metabolismo , Neoplasias Pulmonares/secundario , Selectina-P/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Calcio/metabolismo , Línea Celular Tumoral , Membrana Celular , Sulfatos de Condroitina/análisis , Fucosiltransferasas/genética , Glicosaminoglicanos/análisis , Glicosaminoglicanos/metabolismo , Heparina/farmacología , Humanos , Ligandos , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Oligosacáridos/metabolismo , Proteoglicanos/análisis , Proteoglicanos/metabolismo , Antígeno Sialil Lewis X , Transfección
18.
Vaccine ; 25(16): 3022-31, 2007 Apr 20.
Artículo en Inglés | MEDLINE | ID: mdl-17303294

RESUMEN

Limited immune responses to tumor-associated carbohydrate antigens (TACA) are due in part to their being self-antigens. Immunization with xenoantigens of TACA provides an approach to break tolerance and augment responses to TACA. Carbohydrate mimetic peptides (CMPs) as xenoantigens can induce serum antibodies that target shared carbohydrate residues on differing carbohydrate structures. In preclinical studies, we observe that CMP immunization in mice induce immune responses that are effective in inhibiting the in vitro and in vivo growth of breast cancer and melanoma tumor cells expressing self-target antigens. CMPs of TACA can be further defined that induce IgM antibodies with broadened responses to both breast and melanoma cells. Consequently, CMPs are effective at generating a multifaceted carbohydrate-reactive immune response that should be clinically evaluated for their ability to amplify carbohydrate immune responses against circulating or disseminated tumor cells.


Asunto(s)
Antígenos de Carbohidratos Asociados a Tumores/administración & dosificación , Neoplasias de la Mama/prevención & control , Vacunas contra el Cáncer/administración & dosificación , Melanoma/prevención & control , Animales , Antígenos de Neoplasias , Neoplasias de la Mama/inmunología , Vacunas contra el Cáncer/química , Vacunas contra el Cáncer/inmunología , Melanoma/inmunología , Ratones , Imitación Molecular , Péptidos/genética , Péptidos/inmunología
19.
J Immunol ; 174(11): 7057-65, 2005 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-15905549

RESUMEN

The selective targeting of tumor-associated carbohydrate Ags by the induction of serum Abs that trigger apoptosis of tumor cells as a means to reduce circulating tumor cells and micrometastases would be an advantage in cancer vaccine development. Some plant lectins like Griffonia simplicifolia lectin I and wheat germ agglutinin mediate the apoptosis of tumor cells. We investigated the possibility of using these lectins as templates to select peptide mimotopes of tumor-associated carbohydrate Ags as immunogens to generate cross-reactive Abs capable of mediating apoptosis of tumor cells. In this study, we show that immunization with a mimotope selected based on its reactivity with Griffonia simplicifolia lectin I and wheat germ agglutinin induced serum IgM Abs in mice that mediated the apoptosis of murine 4T1 and human MCF7 cell lines in vitro, paralleling the apoptotic activity of the lectins. Vaccine-induced anti-carbohydrate Abs reduced the outgrowth of micrometastases in the 4T1 spontaneous tumor model, significantly increasing survival time of tumor-bearing animals. This finding parallels suggestions that carbohydrate-reactive IgM with apoptotic activity may have merit in the adjuvant setting if the right carbohydrate-associated targets are identified.


Asunto(s)
Anticuerpos Antineoplásicos/biosíntesis , Anticuerpos Antineoplásicos/uso terapéutico , Antígenos de Carbohidratos Asociados a Tumores/inmunología , Apoptosis/inmunología , Neoplasias Hepáticas/secundario , Neoplasias Hepáticas/terapia , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Secuencia de Aminoácidos , Animales , Anticuerpos Antineoplásicos/sangre , Antígenos de Carbohidratos Asociados a Tumores/genética , Antineoplásicos/administración & dosificación , Antineoplásicos/inmunología , Línea Celular Tumoral , Reacciones Cruzadas , Citotoxicidad Inmunológica/inmunología , Femenino , Inhibidores de Crecimiento/administración & dosificación , Inhibidores de Crecimiento/genética , Inhibidores de Crecimiento/inmunología , Humanos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Neoplasias Mamarias Experimentales/inmunología , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/terapia , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Vacunas de ADN/administración & dosificación , Vacunas de ADN/inmunología , Vacunas de Subunidad/administración & dosificación , Vacunas de Subunidad/genética , Vacunas de Subunidad/inmunología , Aglutininas del Germen de Trigo/farmacología
20.
Int J Cancer ; 117(3): 398-408, 2005 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-15906360

RESUMEN

Expression of sialyl Lewis(x) (sLe(x)) and sLe(a) on tumor cells is thought to facilitate metastasis by promoting cell adhesion to selectins on vascular endothelial cells. Experiments supporting this concept usually bypass the early steps of the metastatic process by employing tumor cells that are injected directly into the blood. We investigated the relative role of sLe(x) oligosaccharide in the dissemination of breast carcinoma, employing a spontaneous murine metastasis model. An sLe(x) deficient subpopulation of the 4T1 mammary carcinoma cell line was produced by negative selection using the sLe(x)-reactive KM93 MAb. This subpopulation was negative for E-selectin binding but retained P-selectin binding. Both sLe(x)-negative and -positive cells grew at the same rate; however, sLe(x)-negative cells spread more efficiently on plates and had greater motility in wound-scratch assays. Mice inoculated in the mammary fat pad with sLe(x)-negative and -positive variants produced lung metastases. However, the number of lung metastases was significantly increased in the group inoculated with the sLe(x)-negative variant (p = 0.0031), indicating that negative selection for the sLe(x) epitope resulted in enrichment for a subpopulation of cells with a high metastatic phenotype. Cell variants demonstrated significant differences in cellular morphology and pattern of tumor growth in primary and secondary tumor sites. These results strongly suggest that loss of sLe(x) may facilitate the metastatic process by contributing to escape from the primary tumor mass.


Asunto(s)
Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/genética , Oligosacáridos/genética , Animales , Selectina E/genética , Femenino , Ligandos , Neoplasias Pulmonares/genética , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Antígeno Sialil Lewis X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA