Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Parkinsons Dis ; 11(3): 1079-1089, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34092654

RESUMO

BACKGROUND: Immunotherapies targeting α-synuclein aim to limit its extracellular spread in the brain and prevent progression of pathology in Parkinson's disease (PD). PD03A is a specific active immunotherapy (SAIT) involving immunization with a short peptide formulation. OBJECTIVE: This phase 1 study characterized the safety and tolerability of PD03A in patients with early PD. A key secondary objective was to evaluate immunological activity following immunization. METHODS: This was a phase 1 study of two different doses of PD03A versus placebo in PD patients. Patients were randomized (1:1:1) to receive four priming plus one booster vaccination of PD03A 15µg, PD03A 75µg or placebo and were followed for 52 weeks. RESULTS: Overall, 36 patients were randomized, of which 35 received five immunizations and completed the study. All patients experienced at least one adverse event. Transient local injection site reactions affected all but two patients; otherwise most AEs were considered unrelated to study treatment. A substantial IgG antibody response against PD03 was observed with a maximum titer achieved at Week-12. Differences in titers between both active groups versus placebo were statistically significant from the second immunization at Week-8 until Week-52. CONCLUSION: The safety profile and positive antibody response of PD03A supports the further development of active immunotherapeutic approaches for the treatment of PD.


Assuntos
Doença de Parkinson , alfa-Sinucleína , Método Duplo-Cego , Humanos , Imunoterapia/efeitos adversos , Imunoterapia Ativa , Doença de Parkinson/tratamento farmacológico
2.
Eur J Clin Pharmacol ; 77(10): 1473-1484, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33969434

RESUMO

PURPOSE: AT04A and AT06A are two AFFITOPE® peptide vaccine candidates being developed for the treatment of hypercholesterolemia by inducing proprotein convertase subtilisin/kexin type 9 (PCSK9)-specific antibodies. This study aimed to investigate safety, tolerability, antibody development, and reduction of low-density lipoprotein cholesterol (LDLc) following four subcutaneous immunizations. METHODS: This phase I, single-blind, randomized, placebo-controlled study was conducted in a total of 72 healthy subjects with a mean fasting LDLc level at baseline of 117.1 mg/dL (range 77-196 mg/dL). Each cohort enrolled 24 subjects to receive three priming immunizations at weeks 0, 4, and 8 and to receive a single booster immunization at week 60 of either AT04A, AT06A, or placebo. In addition to safety (primary objective), the antigenic peptide- and PCSK9-specific antibody response and the impact on LDLc were evaluated over a period of 90 weeks. RESULTS: The most common systemic treatment-related adverse events (AEs) reported were fatigue, headache, and myalgia in 75% of subjects in the AT06A group and 58% and 46% of subjects in the placebo and AT04A groups, respectively. Injection site reactions (ISR) representing 63% of all treatment-emergent adverse events (TEAEs), were transient and mostly of mild or moderate intensity and rarely severe (3%). Both active treatments triggered a robust, long-lasting antibody response towards the antigenic peptides used for immunization that optimally cross-reacted with the target epitope on PCSK9. In the AT04A group, a reduction in serum LDLc was observed with a mean peak reduction of 11.2% and 13.3% from baseline compared to placebo at week 20 and 70 respectively, and over the whole study period, the mean LDLc reduction for the AT04A group vs. placebo was -7.2% (95% CI [-10.4 to -3.9], P < 0.0001). In this group, PCSK9 target epitope titers above 50 were associated with clinically relevant LDLc reductions with an individual maximal decrease of 39%. CONCLUSIONS: Although both AT04A and AT06 were safe and immunogenic, only AT04A demonstrated significant LDLc-lowering activity, justifying further development. TRIAL REGISTRATION: EudraCT: 2015-001719-11. ClinicalTrials.gov Identifier: NCT02508896.


Assuntos
Hipercolesterolemia/tratamento farmacológico , Pró-Proteína Convertase 9/imunologia , Vacinas de Subunidades Antigênicas/uso terapêutico , Adolescente , Adulto , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Método Simples-Cego , Vacinas de Subunidades Antigênicas/administração & dosagem , Vacinas de Subunidades Antigênicas/efeitos adversos , Adulto Jovem
3.
Mov Disord ; 35(11): 1957-1965, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32882100

RESUMO

Multiple system atrophy (MSA) is a rare and fatal neurodegenerative disease with limited symptomatic treatment options. Aggregation of α-synuclein in oligodendrocytes is believed to be a central mechanism of the neurodegenerative process. PD01A and PD03A are 2 novel therapeutic vaccine candidates containing short peptides as antigenic moieties that are designed to induce a sustained antibody response, specifically targeting pathogenic assemblies of α-synuclein. The objectives of the current study were to evaluate primarily the safety and tolerability of PD01A and PD03A in patients with early MSA. Thirty patients (11 women) were randomized to receive 5 subcutaneous injections of either PD01A (n = 12), PD03A (n = 12), or placebo (n = 6) in this patient- and examiner-blinded, placebo-controlled, 52-week phase 1 clinical trial (ClinicalTrial.gov identifier: NCT02270489). Immunogenicity and clinical scores were assessed as secondary objectives. Twenty-nine patients reported a total of 595 treatment-emergent adverse events (mild or moderate, n = 555; severe, n = 40). Treatment-related adverse events included 190 injection-site reactions typically observed in vaccination trials with similar per-subject incidence in the treatment groups over time. Sustained IgG titers were observed in the PD01A-treated group, and 89% of treated patients developed a PD01-specific antibody response after receiving all injections. Induced antibodies displayed clear reactivity to the α-synuclein target epitope. Titers and antibody responder rate (58%) were lower in the PD03A-treated group. In conclusion, both PD01A and PD03A were safe and well tolerated. PD01A triggered a rapid and long-lasting antibody response that specifically targeted the α-synuclein epitope. © 2020 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Assuntos
Atrofia de Múltiplos Sistemas , Doença de Parkinson , Feminino , Humanos , Masculino , Atrofia de Múltiplos Sistemas/tratamento farmacológico , Doença de Parkinson/tratamento farmacológico , Peptídeos , Vacinação , alfa-Sinucleína
4.
Lancet Neurol ; 19(7): 591-600, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32562684

RESUMO

BACKGROUND: Robust evidence supports the role of α-synuclein pathology as a driver of neuronal dysfunction in Parkinson's disease. PD01A is a specific active immunotherapy with a short peptide formulation targeted against oligomeric α-synuclein. This phase 1 study assessed the safety and tolerability of the PD01A immunotherapeutic in patients with Parkinson's disease. METHODS: We did a first-in-human, randomised, phase 1 study of immunisations with PD01A, followed by three consecutive study extensions. Patients aged 45-65 years with a clinical diagnosis of Parkinson's disease (≤4 years since diagnosis and Hoehn and Yahr Stage 1 to 2), imaging results (dopamine transporter single photon emission CT and MRI) consistent with their Parkinson's disease diagnosis, and on stable doses of Parkinson's disease medications for at least 3 months were recruited at a single private clinic in Vienna, Austria. Patients were randomly assigned (1:1), using a computer-generated sequence with varying block size, to receive four subcutaneous immunisations with either 15 µg or 75 µg PD01A injected into the upper arms and followed up initially for 52 weeks, followed by a further 39 weeks' follow-up. Patients were then randomly assigned (1:1) again to receive the first booster immunisation at 15 µg or 75 µg and were followed up for 24 weeks. All patients received a second booster immunisation of 75 µg and were followed up for an additional 52 weeks. Patients were masked to dose allocation. Primary (safety) analyses included all treated patients. These four studies were registered with EU Clinical Trials Register, EudraCT numbers 2011-002650-31, 2013-001774-20, 2014-002489-54, and 2015-004854-16. FINDINGS: 32 patients were recruited between Feb 14, 2012, and Feb 6, 2013, and 24 were deemed eligible and randomly assigned to receive four PD01A priming immunisations. One patient had a diagnosis change to multiple system atrophy and was withdrawn and two patients withdrew consent during the studies. 21 (87%) of 24 patients received all six immunisations and completed 221-259 weeks in-study (two patients in the 15 µg dose group and one patient in the 75 µg dose group discontinued). All patients experienced at least one adverse event, but most of them were considered unrelated to study treatment (except for transient local injection site reactions, which affected all but one patient). Serial MRI assessments also ruled out inflammatory processes. Systemic treatment-related adverse events were fatigue (n=4), headache (n=3), myalgia (n=3), muscle rigidity (n=2), and tremor (n=2). The geometric group mean titre of antibodies against the immunising peptide PD01 increased from 1:46 at baseline to 1:3580 at week 12 in the 15 µg dose group, and from 1:76 to 1:2462 at week 12 in the 75 µg dose group. Antibody titres returned to baseline over 2 years, but could be rapidly reactivated after booster immunisation from week 116 onwards, reaching geometric group mean titres up to 1:20218. INTERPRETATION: Repeated administrations of PD01A were safe and well tolerated over an extended period. Specific active immunotherapy resulted in a substantial humoral immune response with target engagement. Phase 2 studies are needed to further assess the safety and efficacy of PD01A for the treatment of Parkinson's disease. FUNDING: AFFiRiS, Michael J Fox Foundation.


Assuntos
Imunoterapia/métodos , Doença de Parkinson/tratamento farmacológico , Fragmentos de Peptídeos/imunologia , Fragmentos de Peptídeos/uso terapêutico , Peptídeos/imunologia , Peptídeos/uso terapêutico , alfa-Sinucleína/antagonistas & inibidores , Idoso , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Método Simples-Cego
5.
PLoS One ; 9(12): e114469, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25474576

RESUMO

BACKGROUND: Low Density Lipoprotein (LDL) hypercholesterolemia, and its associated cardiovascular diseases, are some of the leading causes of death worldwide. The ability of proprotein convertase subtilisin/kexin 9 (PCSK9) to modulate circulating LDL cholesterol (LDLc) concentrations made it a very attractive target for LDLc management. To date, the most advanced approaches for PCSK9 inhibition are monoclonal antibody (mAb) therapies. Although shown to lower LDLc significantly, mAbs face functional limitations because of their relatively short in vivo half-lives necessitating frequent administration. Here, we evaluated the long-term efficacy and safety of PCSK9-specific active vaccines in different preclinical models. METHODS AND FINDING: PCSK9 peptide-based vaccines were successfully selected by our proprietary technology. To test their efficacy, wild-type (wt) mice, Ldlr+/- mice, and rats were immunized with highly immunogenic vaccine candidates. Vaccines induced generation of high-affine PCSK9-specific antibodies in all species. Group mean total cholesterol (TC) concentration was reduced by up to 30%, and LDLc up to 50% in treated animals. Moreover, the PCSK9 vaccine-induced humoral immune response persisted for up to one year in mice, and reduced cholesterol levels significantly throughout the study. Finally, the vaccines were well tolerated in all species tested. CONCLUSIONS: Peptide-based anti-PCSK9 vaccines induce the generation of antibodies that are persistent, high-affine, and functional for up to one year. They are powerful and safe tools for long-term LDLc management, and thus may represent a novel therapeutic approach for the prevention and/or treatment of LDL hypercholesterolemia-related cardiovascular diseases in humans.


Assuntos
Hipercolesterolemia/terapia , Pró-Proteína Convertases/antagonistas & inibidores , Vacinação , Animais , LDL-Colesterol/sangue , Feminino , Hipercolesterolemia/sangue , Hipercolesterolemia/imunologia , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/imunologia , Ratos Wistar , Serina Endopeptidases/imunologia , Vacinas de Subunidades Antigênicas/uso terapêutico
6.
J Immunol ; 173(5): 3051-61, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15322165

RESUMO

Among the different subsets of dendritic cells (DC) described in humans and mice, epidermal Langerhans cells and dermal DCs represent the only DC populations resident in normal skin. In this study we describe a population of CD4(+)CD3(-) plasmacytoid DC (pDC)-like cells that accumulate in the dermis and spleens of mice topically treated with imiquimod, a low m.w. immune response modifier with potent antiviral and antitumor activities. These CD4(+)CD3(-) cells coexpress GR-1, B220, MHC class II, and, to a lesser extent, CD11c and display the phenotypic features of pDCs described in lymphoid organs. The accumulation of pDC-like cells after imiquimod treatment was detected not only in normal skin, but also in intradermally induced melanomas. Imiquimod treatment leads either to complete regression or to a significant reduction of the tumors. The number of pDCs correlates well with the clinical response of the tumors to the drug, suggesting that the antitumor effects of imiquimod could be mediated at least in part by the recruitment of pDC-like cells to the skin. Therefore, strategies aimed at activating and directing these cells into neoplastic tissues may be a promising and novel approach for the immunotherapy of various types of cancer.


Assuntos
Aminoquinolinas/farmacologia , Antineoplásicos/farmacologia , Células Dendríticas/efeitos dos fármacos , Melanoma/tratamento farmacológico , Pele/efeitos dos fármacos , Animais , Movimento Celular/efeitos dos fármacos , Citometria de Fluxo , Imiquimode , Inflamação/induzido quimicamente , Melanoma/patologia , Camundongos , Baço/efeitos dos fármacos
7.
J Invest Dermatol ; 123(2): 371-9, 2004 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-15245438

RESUMO

DNA vaccines can induce impressive specific cellular immune response (IR) when taking advantage of their recognition as pathogen-associated molecular patterns (PAMP) through Toll-like receptors (TLR) expressed on/in cells of the innate immune system. Among the many types of PAMP, immunostimulatory DNA, so-called CpG motifs, was shown to interact specifically with TLR9, which is expressed in plasmacytoid dendritic cells (pDC), a key regulatory cell for the activation of innate and adaptive IR. We now report that CpG motifs, when introduced into the backbone, are a useful adjuvant for plasmid-based DNA (pDNA) vaccines to induce melanoma antigen-specific protective T cell responses in the Cloudman M3/DBA/2 model. The CpG-enriched pDNA vaccine induced protection against subsequent challenge with melanoma cells at significantly higher levels than its parental unmodified vector. Preferential induction of an antigen-specific, protective T cell response could be demonstrated by (i) induction of antigen-dependent tumor cell protection, (ii) complete loss of protection by in vivo CD4+/CD8+T cell- but not NK cell-depletion, and (iii) the detection of antigen-specific T cell responses but not of relevant NK cell activity in vitro. These results demonstrate that employing PAMP in pDNA vaccines improves the induction of protective, antigen-specific, T cell-mediated IR.


Assuntos
Adjuvantes Imunológicos/genética , Vacinas Anticâncer/genética , Ilhas de CpG/imunologia , Melanoma/prevenção & controle , Neoplasias Cutâneas/prevenção & controle , Vacinas de DNA/genética , Animais , Vacinas Anticâncer/imunologia , Epitopos , Feminino , Vetores Genéticos , Melanoma/imunologia , Camundongos , Camundongos Endogâmicos DBA , Plasmídeos , Neoplasias Cutâneas/imunologia , Transgenes/genética , Transgenes/imunologia , Vacinas de DNA/imunologia
8.
J Immunol ; 171(10): 5180-7, 2003 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-14607918

RESUMO

Subcutaneous injection of GM-CSF-expressing cancer cells into experimental animals results in protective cancer immunity. To delineate the mode of action of such vaccines, we used trinitrophenyl, the antigenic moiety of the contact allergen trinitrochlorobenzene, as surrogate Ag. Trinitrophenyl-derivatized bone marrow-derived dendritic cells were found to elicit a contact hypersensitivity response in syngeneic, but not in allogeneic recipients, compatible with their expected mode of direct Ag presentation. When expressing GM-CSF, haptenized M3 melanoma cells were also able to induce a contact hypersensitivity response but, in contrast to bone marrow-derived dendritic cells, not only in syngeneic but also in allogeneic recipients. This argues for a critical role of host APC. To identify their nature, we introduced the beta-galactosidase (betagal) gene into M3-GM cells. Their administration activated betagal-specific, L(d)-restricted CTL in syngeneic BALB/c mice. Evaluation of lymph nodes draining M3-GM-betagal injection sites revealed the presence of cells presenting the respective L(d)-binding betagal peptide epitope. Based on their capacity to activate betagal-specific CTL, they were identified as being CD11c(+) dendritic cells. These experiments provide a rational basis for the use of GM-CSF-based melanoma cell vaccines in an allogeneic setting.


Assuntos
Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Melanoma/imunologia , Transplante Homólogo/imunologia , Transplante Isogênico/imunologia , Administração Cutânea , Animais , Apresentação de Antígeno/genética , Transplante de Medula Óssea/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/genética , Linhagem Celular Tumoral , Citotoxicidade Imunológica/genética , Células Dendríticas/transplante , Dermatite de Contato/imunologia , Epitopos de Linfócito T/administração & dosagem , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/administração & dosagem , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Injeções Subcutâneas , Células de Langerhans/imunologia , Células de Langerhans/transplante , Ativação Linfocitária/genética , Melanoma/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Transplante de Neoplasias/imunologia , Cloreto de Picrila/administração & dosagem , Cloreto de Picrila/imunologia , Transfecção , Trinitrobenzenos/administração & dosagem , Trinitrobenzenos/imunologia , beta-Galactosidase/biossíntese , beta-Galactosidase/genética , beta-Galactosidase/imunologia
9.
Anticancer Res ; 23(6C): 4843-51, 2003.
Artigo em Inglês | MEDLINE | ID: mdl-14981934

RESUMO

BACKGROUND: In our previous study, we found complexes of the non-coding pMP6 plasmid and cationic liposomes which exerted antitumor activity in the C26 model. We now sought to unravel the underlying protective effector mechanism(s). MATERIALS AND METHODS: C26 recipients (i.p.; day 0) were injected once (day -2) or twice (days -2 and 2) with pMP6/liposome complexes. Thus treated mice were evaluated for tumor growth and the occurrence of innate and specific immune responses. RESULTS: A single pMP6/liposome injection prolonged the survival of the animals as compared to non-treated C26 recipients (median survival 28 vs. 19 days). Two injections not only prolonged the survival time (median survival 55 days) but completely prevented tumor development in 50% of C26 recipients. I.p. administration of pMP6/liposome complexes resulted in the expression of the proinflammatory cytokines IL-6, IFN-gamma and TNF-alpha. This was followed by the appearance of activated NK cells within the peritoneal cavity and, somewhat later, by the induction of C26-specific CTLs. T cell depletion studies demonstrated the latter to be critical for the protective effect to occur. CONCLUSION: Our data suggests that co-application of tumor cells and immunostimulatory pDNA/liposome complexes induces a protective and tumor-specific T cell response. Therefore these complexes may be considered promising agents in the immunological gene therapy of cancer.


Assuntos
Adenocarcinoma/genética , Divisão Celular/efeitos dos fármacos , Neoplasias do Colo/genética , Lipossomos , Plasmídeos/uso terapêutico , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Citocinas/genética , Portadores de Fármacos , Feminino , Lipossomos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Fatores de Tempo , Células Tumorais Cultivadas
10.
J Immunol ; 169(9): 5217-26, 2002 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-12391240

RESUMO

The s.c injection of tumor Ag-derived, MHC class I-binding peptides together with cationic poly-amino acids (e.g., poly-L-arginine; pR) has been shown to protect animals against a challenge with tumor cells expressing the respective peptide(s). Given our only restricted knowledge about immunogenic tumor-associated peptides, we sought to determine whether this pR-based vaccination protocol would also induce protective cancer immunity if large proteins were used instead of peptide epitopes. We found that the intracutaneous administration of the model Ag beta-galactosidase (beta-gal) together with pR (referred to as pR-based protein vaccine; pR-PV) was significantly more potent in protecting mice against the growth of beta-gal-expressing RENCA cells than the protein alone. Coadministration of pR enhanced both the beta-gal-induced specific humoral and CD8 response. The protective effect required CD8(+), but neither CD4(+) T lymphocytes nor beta-gal-specific Abs. beta-Gal priming of protective CD8(+) T lymphocytes was found to be CD4(+) T cell-independent, to take place within the draining lymph nodes, and to be accomplished by day 5 after vaccination. Ablation of the injection sites as early as 1.5 h after pR-PV administration still led to protection in a large proportion of the animals, indicating that certain protein Ags administered intradermally in the context of polycations are quickly transported to the draining nodes, where they induce molecular and cellular events resulting in the helper-independent priming and expansion of Tc1 cells. However, optimal protection required the prolonged presence of the injection site, suggesting that pR-PV injection facilitates the formation of a cutaneous depot of Ag-charged cells capable of migration and T cell activation.


Assuntos
Cátions/imunologia , Peptídeos/imunologia , Poliaminas/imunologia , Vacinas/imunologia , beta-Galactosidase/imunologia , Adjuvantes Imunológicos/administração & dosagem , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antineoplásicos/administração & dosagem , Antineoplásicos/imunologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Cátions/administração & dosagem , Células Clonais , Citotoxicidade Imunológica , Epitopos de Linfócito T/imunologia , Inibidores do Crescimento/administração & dosagem , Inibidores do Crescimento/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Injeções Intradérmicas , Injeções Subcutâneas , Neoplasias Renais/imunologia , Neoplasias Renais/prevenção & controle , Neoplasias Renais/terapia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Transplante de Neoplasias , Peptídeos/administração & dosagem , Poliaminas/administração & dosagem , Polieletrólitos , Células Tumorais Cultivadas , Vacinas/administração & dosagem , beta-Galactosidase/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA