Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 801
Filtrar
2.
Neuropharmacology ; 202: 108870, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34742741

RESUMO

Parkinson's disease (PD) is the second most common neurodegenerative disorder and is defined pathologically by the abnormal accumulation of the presynaptic protein alpha-synuclein (aSyn) in the form of Lewy bodies and Lewy neurites and loss of midbrain dopaminergic neurons in the substantia nigra pars compacta. Because of aSyn's involvement in both sporadic and familial forms of PD, it has become a key target for the development of novel therapeutics. Aberrant aSyn is associated with multiple mechanisms of neuronal dysfunction and degeneration including inflammation, impaired mitochondrial function, altered protein degradation systems, and oxidative stress. Inflammation, in particular, has emerged as a potential significant contributor early in the disease making it an attractive target for disease modification and neuroprotection. Thus, immunotherapies targeting aSyn are currently being investigated in pre-clinical and clinical trials. The focus of this review is to highlight the role of aSyn in neuroinflammation and discuss the current status of aSyn-directed immunotherapies in pre-clinical and clinical trials for PD.


Assuntos
Sistema Imunitário/imunologia , Imunoterapia Ativa/métodos , Imunoterapia/métodos , Terapia de Alvo Molecular/métodos , Doença de Parkinson/etiologia , Doença de Parkinson/terapia , alfa-Sinucleína/metabolismo , Animais , Ensaios Clínicos como Assunto , Neurônios Dopaminérgicos/patologia , Humanos , Imunoterapia/tendências , Imunoterapia Ativa/tendências , Corpos de Lewy/metabolismo , Camundongos Transgênicos , Terapia de Alvo Molecular/tendências , Doenças Neuroinflamatórias , Estresse Oxidativo , Doença de Parkinson/imunologia , Substância Negra/metabolismo , Substância Negra/patologia
3.
Ann Surg ; 275(1): e155-e162, 2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-33055588

RESUMO

OBJECTIVES: To elucidate the efficacy of adjuvant vaccine monotherapy using 3 Human Leukocyte Antigen (HLA)-A∗24-restricted tumor-specific peptide antigens for ESCC, upregulated lung cancer 10, cell division cycle associated 1, and KH domain-containing protein overexpressed in cancer 1. SUMMARY OF BACKGROUND DATA: ESCC patients with pathologically positive nodes (pN(+)) have a high risk for postoperative recurrence, despite curative resection after preoperative therapy. Subclinical micrometastases are an appropriate target for cancer vaccine. METHODS: This is a non-randomized prospective phase II clinical trial (UMIN000003557). ESCC patients curatively resected after preoperative therapy with pN(+) were allocated into the control and vaccine groups (CG and VG) according to the HLA-A status. One mg each of three epitope peptides was postoperatively injected 10 times weekly followed by 10 times biweekly to the VG. The primary and secondary endpoints were relapse-free survival (RFS) and esophageal cancer-specific survival (ECSS), respectively. RESULTS: Thirty were in the CG and 33 in the VG. No significant difference was observed in RFS between the CG and VG (5-year RFS: 32.5% vs 45.3%), but the recurrence rate significantly decreased with the number of peptides which induced antigen-specific cytotoxic T lymphocytes. The VG showed a significantly higher 5-year ECSS than the CG (60.0% vs 32.4%, P = 0.045) and this difference was more prominent in patients with CD8+ and programmed death-ligand 1 double negative tumor (68.0% vs 17.7%, P = 0.010). CONCLUSIONS: Our cancer peptide vaccine might improve the survival of ESCC patients, which is warranted to be verified in the phase III randomized controlled study.


Assuntos
Vacinas Anticâncer/uso terapêutico , Neoplasias Esofágicas/tratamento farmacológico , Esofagectomia , Imunoterapia Ativa/métodos , Linfonodos/patologia , Cuidados Pré-Operatórios/métodos , Microambiente Tumoral/imunologia , Adulto , Idoso , Antígenos de Neoplasias/imunologia , Intervalo Livre de Doença , Neoplasias Esofágicas/imunologia , Neoplasias Esofágicas/secundário , Feminino , Seguimentos , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante , Estadiamento de Neoplasias/métodos , Estudos Prospectivos
4.
Oncogene ; 40(21): 3655-3664, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33947958

RESUMO

Over the past 25 years, antibody therapeutics have emerged as clinically and commercially successful pharmaceuticals, rapidly approaching 100 Food and Drug Administration approvals with combined annual global sales exceeding $100 billion. Nearly half of the marketed antibody therapeutics are used in oncology. These antibody-based cancer therapies can be broken down into three categories based on their different mechanisms of action, i.e., (i) natural properties, (ii) engagement of cytotoxic T cells, and (iii) delivery of cytotoxic payloads. Both natural and engineered properties of the antibody molecule are founded on its highly stable and modular architecture. In this review we provide an overview and outlook of the rapidly evolving landscape of antibody-based cancer therapy.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Antineoplásicos Imunológicos/uso terapêutico , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia Ativa/métodos , Imunoterapia Adotiva/métodos , Neoplasias/terapia , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/metabolismo
5.
Curr Opin Immunol ; 69: 65-71, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33862306

RESUMO

Upon their activation, CD8+ T cells in the tumor micro-environment (TME) secrete cytokines such as IFNγ, TNFα, and IL-2. While over the past years a major interest has developed in the antigenic signals that induce such cytokine release, our understanding of the cells that subsequently sense these CD8+ T-cell secreted cytokines is modest. Here, we review the current insights into the spreading behavior of CD8+ T-cell-secreted cytokines in the TME. We argue for a model in which variation in the mode of cytokine secretion, cytokine half-life, receptor-mediated clearance, cytokine binding to extracellular components, and feedback or forward loops, between different cytokines or between individual tumors, sculpts the local tissue response to natural and therapy-induced T-cell activation in human cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Citocinas/metabolismo , Imunoterapia Ativa/métodos , Neoplasias/terapia , Animais , Humanos , Imunomodulação , Ativação Linfocitária , Neoplasias/imunologia , Comunicação Parácrina , Microambiente Tumoral
8.
Neurobiol Aging ; 101: 94-108, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33610062

RESUMO

Despite effective clearance of parenchymal amyloid-ß (Aß) in patients with Alzheimer's disease, Aß immunotherapy exacerbates the vascular Aß (VAß)-associated pathology in the brain. We have previously shown that BCG immunization facilitates protective monocyte recruitment to the brain of APP/PS1 mice. Here, we confirmed that the 4Aß1-15 vaccine exacerbates VAß deposits in this model, which coincides with a decrease in the number of cerebrovascular endothelial cells and pericytes, infiltration of neutrophils into the brain, and induction of cerebral microhemorrhage. Moreover, combined 4Aß1-15/BCG treatment abrogates the development of the VAß-associated pathology. In addition, BCG treatment is required for the upregulation of interleukin-10 in the brain. Notably, BCG treatment selectively enhances Aß phagocytosis by recruited macrophages. Furthermore, combined 4Aß1-15/BCG treatment is more effective than 4Aß1-15 monotherapy in synaptic preservation and the enhancement of the learning efficiency. Overall, our study suggests that the combination of Aß-targeted therapy with an immunomodulatory strategy may improve the efficacy of Aß vaccine in Alzheimer's disease.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/metabolismo , Vacina BCG/administração & dosagem , Encéfalo/metabolismo , Imunoterapia Ativa/métodos , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/psicologia , Animais , Vacina BCG/farmacologia , Encéfalo/patologia , Modelos Animais de Doenças , Feminino , Interleucina-10/metabolismo , Aprendizagem , Macrófagos/imunologia , Macrófagos/patologia , Camundongos Transgênicos , Fagocitose/efeitos dos fármacos
9.
Clin Epigenetics ; 13(1): 25, 2021 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-33531075

RESUMO

Epigenetic therapies may modulate the tumor microenvironment. We evaluated the safety and optimal sequence of combination DNA methyltransferase inhibitor guadecitabine with a granulocyte macrophage-colony-stimulating-factor (GM-CSF) secreting colon cancer (CRC) vaccine (GVAX) using a primary endpoint of change in CD45RO + T cells. 18 patients with advanced CRC enrolled, 11 underwent paired biopsies and were evaluable for the primary endpoint. No significant increase in CD45RO + cells was noted. Grade 3-4 toxicities were expected and manageable. Guadecitabine + GVAX was tolerable but demonstrated no significant immunologic activity in CRC. We report a novel trial design to efficiently evaluate investigational therapies with a primary pharmacodynamic endpoint.Trial registry Clinicaltrials.gov: NCT01966289. Registered 21 October, 2013.


Assuntos
Azacitidina/análogos & derivados , Vacinas Anticâncer/farmacologia , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , DNA (Citosina-5-)-Metiltransferase 1/antagonistas & inibidores , Adulto , Idoso , Idoso de 80 Anos ou mais , Azacitidina/administração & dosagem , Azacitidina/efeitos adversos , Azacitidina/farmacologia , Azacitidina/uso terapêutico , Biópsia , Vacinas Anticâncer/administração & dosagem , Vacinas Anticâncer/efeitos adversos , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Terapia Combinada/métodos , Metilação de DNA/efeitos dos fármacos , Epigenômica/métodos , Estudos de Viabilidade , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Imunoterapia/métodos , Imunoterapia Ativa/métodos , Antígenos Comuns de Leucócito/efeitos dos fármacos , Antígenos Comuns de Leucócito/metabolismo , Masculino , Pessoa de Meia-Idade , Segurança , Índice de Gravidade de Doença , Microambiente Tumoral
10.
J Infect Public Health ; 14(2): 221-226, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33493918

RESUMO

BACKGROUND: Rabies is found in many countries of the eastern Mediterranean and is one of the most important zoonotic diseases in the world. The study aims to describe rabies suspected exposures (RSE) and rabies prophylaxis practices in Antalya-Turkey between 2010 and 2013. METHODS: All 2513 RSE cases presenting to a rabies vaccination center in Antalya, southwestern Turkey, were retrospectively investigated. RESULTS: The mean age of the RSE cases was 30.04±19.63 years with male predominance (63.6%). The vast majority was from urban areas (91.7%), and a postexposure rabies vaccination program was applied to 79.7% of participants. Dogs were the primary source of RSE cases (61.2%). The 39.2% of animals were under observation, and 9.53% of them died. Forty-two animals (1.7%) were laboratory confirmed rabid; 61.9% of them were cows. The rabid animal rate in the rural area was significantly higher than the urban area (18.2% versus 0.2%; p=0.001). CONCLUSIONS: This study includes a large number of RSE cases and prophylaxis practices in southwestern Turkey. Most RSE cases had dog or cat contact. As most RSE cases were in urban areas; more focused efforts should be made for elimination and vaccination of feral dog and cat population in Turkey.


Assuntos
Mordeduras e Picadas/complicações , Imunoterapia Ativa/métodos , Profilaxia Pós-Exposição , Vacina Antirrábica/administração & dosagem , Raiva/epidemiologia , Raiva/prevenção & controle , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Mordeduras e Picadas/epidemiologia , Mordeduras e Picadas/virologia , Gatos , Bovinos , Criança , Cães , Feminino , Humanos , Incidência , Lactente , Recém-Nascido , Masculino , Saúde Pública , Estudos Retrospectivos , Turquia/epidemiologia , Adulto Jovem
11.
Nat Aging ; 1(6): 521-534, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-37117834

RESUMO

Alzheimer's disease (AD) pathology is partly characterized by accumulation of aberrant forms of tau protein. Here we report the results of ADAMANT, a 24-month double-blinded, parallel-arm, randomized phase 2 multicenter placebo-controlled trial of AADvac1, an active peptide vaccine designed to target pathological tau in AD (EudraCT 2015-000630-30). Eleven doses of AADvac1 were administered to patients with mild AD dementia at 40 µg per dose over the course of the trial. The primary objective was to evaluate the safety and tolerability of long-term AADvac1 treatment. The secondary objectives were to evaluate immunogenicity and efficacy of AADvac1 treatment in slowing cognitive and functional decline. A total of 196 patients were randomized 3:2 between AADvac1 and placebo. AADvac1 was safe and well tolerated (AADvac1 n = 117, placebo n = 79; serious adverse events observed in 17.1% of AADvac1-treated individuals and 24.1% of placebo-treated individuals; adverse events observed in 84.6% of AADvac1-treated individuals and 81.0% of placebo-treated individuals). The vaccine induced high levels of IgG antibodies. No significant effects were found in cognitive and functional tests on the whole study sample (Clinical Dementia Rating-Sum of the Boxes scale adjusted mean point difference -0.360 (95% CI -1.306, 0.589)), custom cognitive battery adjusted mean z-score difference of 0.0008 (95% CI -0.169, 0.172). We also present results from exploratory and post hoc analyses looking at relevant biomarkers and clinical outcomes in specific subgroups. Our results show that AADvac1 is safe and immunogenic, but larger stratified studies are needed to better evaluate its potential clinical efficacy and impact on disease biomarkers.


Assuntos
Doença de Alzheimer , Humanos , Doença de Alzheimer/terapia , Proteínas tau , Imunoterapia Ativa/métodos , Biomarcadores
12.
Pediatr Res ; 89(3): 476-482, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32311698

RESUMO

BACKGROUND: Neonatal hyperoxia increases oxidative stress and adversely disturbs glomerular and tubular maturity. Maternal Tn immunization induces anti-Tn antibody titer and attenuates hyperoxia-induced lung injury in neonatal rats. METHODS: We intraperitoneally immunized female Sprague-Dawley rats (6 weeks old) with Tn immunogen (50 µg/dose) or carrier protein five times at biweekly intervals on 8, 6, 4, 2, and 0 weeks before the delivery day. The pups were reared for 2 weeks in either room air (RA) or in 85% oxygen-enriched atmosphere (O2), thus generating four study groups, namely carrier protein + RA, Tn vaccine + RA, carrier protein + O2, and Tn vaccine + O2. On postnatal day 14, the kidneys were harvested for the oxidative stress marker 8-hydroxy-2'-deoxyguanosine (8-OHdG), nuclear factor-κB (NF-κB), and collagen expression and histological analyses. RESULTS: Hyperoxia reduced body weight, induced tubular and glomerular injuries, and increased 8-OHdG and NF-κB expression and collagen deposition in the kidneys. By contrast, maternal Tn immunization reduced kidney injury and collagen deposition in neonatal rats. Furthermore, kidney injury attenuation was accompanied by a reduction in 8-OHdG and NF-κB expression. CONCLUSION: Maternal Tn immunization protects against hyperoxia-induced kidney injury in neonatal rats by attenuating oxidative stress and NF-κB activity. IMPACT: Hyperoxia increased nuclear factor-κB (NF-κB) activity and collagen deposition in neonatal rat kidney. Maternal Tn immunization reduced kidney injury as well as collagen deposition in neonatal rats. Maternal Tn immunization reduced kidney injury and was associated with a reduction in 8-hydroxy-2'-deoxyguanosine and NF-κB activity. Tn vaccine can be a promising treatment modality against hyperoxia-induced kidney injury in neonates.


Assuntos
Injúria Renal Aguda/prevenção & controle , Antígenos Glicosídicos Associados a Tumores/imunologia , Hiperóxia/complicações , Imunoterapia Ativa/métodos , Injúria Renal Aguda/etiologia , Animais , Animais Recém-Nascidos , Peso Corporal , Colágeno/análise , Desoxiadenosinas/metabolismo , Feminino , Túbulos Renais/química , Túbulos Renais/patologia , NF-kappa B/metabolismo , Tamanho do Órgão , Estresse Oxidativo , Gravidez , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Vacinação , Vacúolos/ultraestrutura
13.
Curr Mol Med ; 21(1): 45-55, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-32338218

RESUMO

BACKGROUND: Alzheimer's disease (AD) is the most common progressive neurodegenerative disorder characterized by senile plaques and neurofibrillary tangles (NFTs). The amyloid-oligomer hypothesis indicates that the buildup of toxic oligomers in vivo is likely to impair memory and synaptic function. METHODS: In our study, a kind of novel recombinant chimeric 12×(Aß1-15-Th) antigen was developed as 12-mer Aß1-42-like assembly vaccine. We designed this 12×(Aß1-15- Th) antigen to mimic the assembly states of Aß1-42 using twelvefold Aß1-15 (B cell epitopes of human Aß1-42) and foreign human T helper (Th) epitopes (as the T cell epitopes of Aß1-42) constructs. Its immunogenicity as a subunit vaccine was tested on C57/BL6 mice, and the efficacy was shown by applying it to AD mice. RESULTS: This 12×(Aß1-15-Th) vaccine induced robust Aß-specific antibodies in 3×Tg- AD and C57/BL6 mice. As early immunotherapeutic agent of AD, the 12×(Aß1-15-Th) vaccine significantly improved the behavior performance of aged 3 × Tg-AD mice, and reduced the levels of soluble Aß oligomers and soluble Aß in the brain. In aged 3 × Tg- AD mice, immunotherapy with the 12×(Aß1-15-Th) vaccine could prevent Aß-induced decrease of synaptic proteins, which suggested that it had neuroprotective effects on the brain. CONCLUSION: The novel recombinant 12×(Aß1-15-Th) chimeric vaccine targeting of pathological conformations of Aß oligomers has shown obvious neuroprotective benefits in the preclinical AD model mouse, which indicates that it is a good candidate vaccine for the prophylaxis of AD.


Assuntos
Doença de Alzheimer/terapia , Peptídeos beta-Amiloides/imunologia , Cognição/efeitos dos fármacos , Modelos Animais de Doenças , Imunoterapia Ativa/métodos , Fragmentos de Peptídeos/imunologia , Linfócitos T/imunologia , Vacinas de Subunidades/administração & dosagem , Doença de Alzheimer/imunologia , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Animais , Citocinas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T/efeitos dos fármacos , Vacinas de Subunidades/imunologia
14.
Brasília; CONITEC; 2021.
Não convencional em Português | BRISA/RedTESA | ID: biblio-1444046

RESUMO

A DOENÇA: Segundo dados da Organização Mundial da Saúde (OMS) de 2018, o câncer renal representa 2,2% de todos os diagnósticos de câncer, sendo o 15º mais incidente no mundo. Em termos de mortalidade, foi responsável por 1,8% do número de mortes mundiais por doença oncológica. No Brasil, o carcinoma renal tem incidência estimada de 7 a 10 casos para 100.000 habitantes, e representa 2% a 3% de todas as neoplasias malignas do adulto. O carcinoma de células renais (CCR) representa 80% a 90% de todos os cânceres renais. A maioria dos CCR é esporádica (não relacionado a fatores hereditários), sendo que alguns fatores estão relacionados a um risco aumentado de desenvolver a doença. Os principais fatores de risco conhecidos são idade (entre 60 e 70 anos), sexo (predomínio no sexo masculino), obesidade, tabagismo, hipertensão, doença renal crônica ou cística, exposição ocupacional, uso inadequado de medicamentos, sobretudo analgésicos, fatores genéticos, anemia falciforme, cálculos renais


Assuntos
Humanos , Carcinoma de Células Renais/tratamento farmacológico , Citocinas/uso terapêutico , Imunoterapia Ativa/métodos , Receptores Proteína Tirosina Quinases/uso terapêutico , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias Renais/tratamento farmacológico , Brasil , Eficácia , Análise Custo-Benefício , Inibidores da Angiogênese/uso terapêutico , Projetos de Desenvolvimento Tecnológico e Inovação
15.
Mol Pharm ; 17(12): 4603-4615, 2020 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-33175556

RESUMO

Cancer nanovaccines have been widely explored to enhance immunotherapy efficiency, in which the significant irritation of antigen-specific cytotoxic T cells (CTLs) is the critical point. In this study, we developed a pH and reduction dual-sensitive nanovaccine (PMSN@OVA-MPN) composed of two parts. The inner part was made up of polyethyleneimine (PEI)-modified mesoporous silica nanoparticles (MSNs) loaded with model antigen ovalbumin (OVA) and the outer part was made up of disulfide bond-involved metal-phenolic networks (MPNs) as a protective corona. In vitro release experiments proved that PMSN@OVA-MPN could intelligently release OVA in the presence of reductive glutathione, but not in neutral phosphate-buffered saline (PBS). Moreover, in vitro cell assays indicated that the nanovaccine promoted not only the OVA uptake efficiency by DC2.4 cells but also antigen lysosome escape due to the proton sponge effect of PEI. Furthermore, in vivo animal experiments indicated that PMSN@OVA-MPN induced a large tumor-specific cellular immune response so as to effectively inhibit the growth of an existing tumor. Finally, the immune memory effect caused by the nanovaccine afforded conspicuous prophylaxis efficacy in neonatal tumors. Hence, the multifunctional vaccine delivery system prepared in this work exhibits a great application potential in cancer immunotherapy and offers a platform for the development of nanovaccines.


Assuntos
Vacinas Anticâncer/administração & dosagem , Portadores de Fármacos/química , Imunoterapia Ativa/métodos , Nanosferas/química , Neoplasias/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/farmacocinética , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Modelos Animais de Doenças , Composição de Medicamentos/métodos , Liberação Controlada de Fármacos , Feminino , Humanos , Concentração de Íons de Hidrogênio , Imunogenicidade da Vacina , Memória Imunológica , Estruturas Metalorgânicas/química , Camundongos , Neoplasias/imunologia , Polietilenoimina/química , Dióxido de Silício/química
16.
Front Immunol ; 11: 1855, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32973764

RESUMO

Current treatments for chronic immune-mediated diseases such as psoriasis, rheumatoid arthritis, or Crohn's disease commonly rely on cytokine neutralization using monoclonal antibodies; however, such approaches have drawbacks. Frequent repeated dosing can lead to the formation of anti-drug antibodies and patient compliance issues, and it is difficult to identify a single antibody that is broadly efficacious across diverse patient populations. As an alternative to monoclonal antibody therapy, anti-cytokine immunization is a potential means for long-term therapeutic control of chronic inflammatory diseases. Here we report a supramolecular peptide-based approach for raising antibodies against IL-17 and demonstrate its efficacy in a murine model of psoriasis. B-cell epitopes from IL-17 were co-assembled with the universal T-cell epitope PADRE using the Q11 self-assembling peptide nanofiber system. These materials, with or without adjuvants, raised antibody responses against IL-17. Exploiting the modularity of the system, multifactorial experimental designs were used to select formulations maximizing titer and avidity. In a mouse model of psoriasis induced by imiquimod, unadjuvanted nanofibers had therapeutic efficacy, which could be enhanced with alum adjuvant but reversed with CpG adjuvant. Measurements of antibody subclass induced by adjuvanted and unadjuvanted formulations revealed strong correlations between therapeutic efficacy and titers of IgG1 (improved efficacy) or IgG2b (worsened efficacy). These findings have important implications for the development of anti-cytokine active immunotherapies and suggest that immune phenotype is an important metric for eliciting therapeutic anti-cytokine antibody responses.


Assuntos
Desenho de Fármacos , Interleucina-17/antagonistas & inibidores , Psoríase/imunologia , Vacinas de Subunidades/imunologia , Vacinas de Subunidades/farmacologia , Animais , Modelos Animais de Doenças , Feminino , Imunoterapia Ativa/métodos , Camundongos , Camundongos Endogâmicos C57BL
17.
Br J Radiol ; 93(1113): 20200112, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32706978

RESUMO

Interventional oncology (IO) has proven to be highly efficient in the local therapy of numerous malignant tumors in addition to surgery, chemotherapy, and radiotherapy. Due to the advent of immune-oncology with the possibility of tumor control at the molecular and cellular levels, a system change is currently emerging. This will significantly rule oncology in the coming decades. Therefore, one cannot think about IO in the 21st century without considering immunology. For IO, this means paying much more attention to the immunomodulatory effects of the interventional techniques, which have so far been neglected, and to explore the synergistic possibilities with immuno-oncology. It can be expected that the combined use of IO and immuno-oncology will help to overcome the limitations of the latter, such as limited local effects and a high rate of side-effects. To do this, however, sectoral boundaries must be removed and interdisciplinary research efforts must be strengthened. In case of success, IO will face an exciting future.


Assuntos
Imunoterapia/tendências , Oncologia/tendências , Neoplasias/terapia , Vacinas Anticâncer/uso terapêutico , Quimioembolização Terapêutica/métodos , Quimioembolização Terapêutica/tendências , História do Século XXI , Humanos , Imunização Passiva/métodos , Imunomodulação , Imunoterapia/métodos , Imunoterapia Ativa/métodos , Neoplasias/imunologia , Radioterapia (Especialidade)/tendências , Ablação por Radiofrequência/métodos , Ablação por Radiofrequência/tendências , Radioisótopos de Ítrio/uso terapêutico
18.
Immunopharmacol Immunotoxicol ; 42(4): 346-357, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32515626

RESUMO

BACKGROUND: Human epidermal growth factor receptor 2 (HER2) is overexpressed in a subset of cancers including 25% of breast cancers. Since combination therapy consisting of multiple therapeutic approaches is considered a promising regimen, we examined combination treatment modalities in a xenograft model in Balb/c mice injected with 4T1-HER2 cells. We used HER2/neu-loaded bone marrow-derived dendritic cells (BM-DC's) along with anti-PD-L1 monoclonal antibody in a new combination immunotherapy model. METHODS: The combination was composed of an active immunotherapy (i.e. BM-DC-based vaccine) designed to boost the immune response against target antigen and was augmented by using anti-PD-L1 mAb to prevent immune evasion by the xenografted tumors. The vaccine combination was further supported using a QS-21 saponin adjuvant and the immune response was evaluated. RESULTS: Mice treated with HER2/neu-loaded BM-DCs, combined with QS-21 and anti-PD-L1 mAb had significantly decreased tumor sizes and their splenocytes had enhanced cytotoxic activity, based on the lactate dehydrogenase (LDH) assay, compared to vaccine and adjuvant groups alone. The same vaccination group demonstrated a remarkable increase in IFN-γ secreting CD8+ T-cells analyzed by flow cytometry. ELISA data also revealed a significant increase in the serum anti-HER2 IgG1 response; in addition, there was significant splenocyte proliferation upon stimulation with antigen compared to vaccine and adjuvant groups. Consistently, a significant infiltration of CD4+, CD8+ immune cells in and around the tumors was observed. CONCLUSIONS: Our data suggest that the BM-DC + HER2/neu + QS-21 + anti-PD-L1 vaccine combination paradigm synergistically generates anti-tumor activity and immune responses against HER2 overexpressing breast cancer in mice.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Células Dendríticas/transplante , Fragmentos de Peptídeos/administração & dosagem , Receptor ErbB-2/administração & dosagem , Receptor ErbB-2/genética , Saponinas/administração & dosagem , Adjuvantes Imunológicos/administração & dosagem , Animais , Antineoplásicos Imunológicos/administração & dosagem , Neoplasias da Mama/patologia , Vacinas Anticâncer/administração & dosagem , Linhagem Celular Tumoral , Terapia Combinada/métodos , Feminino , Humanos , Imunoterapia Ativa/métodos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
19.
J Immunother Cancer ; 8(1)2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32554612

RESUMO

BACKGROUND: While prophylactic human papillomavirus (HPV) vaccines will certainly reduce the incidence of HPV-associated cancers, these malignancies remain a major health issue. PDS0101 is a liposomal-based HPV therapeutic vaccine consisting of the immune activating cationic lipid R-DOTAP and HLA-unrestricted HPV16 peptides that has shown in vivo CD8+ T cell induction and safety in a phase I study. In this report, we have employed the PDS0101 vaccine with two immune modulators previously characterized in preclinical studies and which are currently in phase II clinical trials. Bintrafusp alfa (M7824) is a first-in-class bifunctional fusion protein composed of the extracellular domains of the transforming growth factor-ß receptor type II (TGFßRII) fused to a human IgG1 monoclonal antibody blocking programmed cell death protein-1 ligand (PDL1), designed both as a checkpoint inhibitor and to bring the TGFßRII 'trap' to the tumor microenvironment (TME). NHS-interleukin-12 (NHS-IL12) is a tumor targeting immunocytokine designed to bring IL-12 to the TME and thus enhance the inflammatory Th1 response. METHODS: We employed TC-1 carcinoma (expressing HPV16 E6 and E7 and devoid of PDL1 expression) in a syngeneic mouse model in monotherapy and combination therapy studies to analyze antitumor effects and changes in immune cell types in the spleen and the TME. RESULTS: As a monotherapy, the PDS0101 vaccine generated HPV-specific T cells and antitumor activity in mice bearing HPV-expressing mEER oropharyngeal and TC-1 lung carcinomas. When used as a monotherapy in the TC-1 model, NHS-IL12 elicited antitumor effects as well as an increase in CD8+ T cells in the TME. When used as a monotherapy, bintrafusp alfa did not elicit antitumor effects or any increase in T cells in the TME. When all three agents were used in combination, maximum antitumor effects were observed, which correlated with increases in T cells and T-cell clonality in the TME. CONCLUSION: These studies provide the rationale for the potential clinical use of combinations of agents that can (1) induce tumor-associated T-cell responses, (2) potentiate immune responses in the TME and (3) reduce immunosuppressive entities in the TME.


Assuntos
Vacinas Anticâncer/imunologia , Carcinoma/terapia , Inibidores de Checkpoint Imunológico/administração & dosagem , Infecções por Papillomavirus/terapia , Vacinas contra Papillomavirus/imunologia , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer/administração & dosagem , Carcinoma/imunologia , Carcinoma/patologia , Carcinoma/virologia , Linhagem Celular Tumoral/transplante , Modelos Animais de Doenças , Feminino , Papillomavirus Humano 16/imunologia , Humanos , Imunoconjugados/administração & dosagem , Imunogenicidade da Vacina , Imunoglobulina G/administração & dosagem , Imunoterapia Ativa/métodos , Interleucina-12/administração & dosagem , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Proteínas Oncogênicas Virais/imunologia , Proteínas E7 de Papillomavirus/imunologia , Infecções por Papillomavirus/imunologia , Infecções por Papillomavirus/patologia , Infecções por Papillomavirus/virologia , Vacinas contra Papillomavirus/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Repressoras/imunologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia , Vacinas de Subunidades/administração & dosagem , Vacinas de Subunidades/imunologia
20.
J Immunol Res ; 2020: 1705187, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32411788

RESUMO

Signal regulatory protein α (SIRPα), a transmembrane protein that is predominantly expressed in dendritic cells (DCs) or macrophages, interacts with CD47 that is overexpressed in almost all types of tumor cells. The interaction between SIRPα and CD47 leads to a negative signal that prevents the phenotypic and functional maturation of DC and inhibits phagocytosis. The SIRPα knockdown in DCs that were pulsed with a modified HPV16E7 (HPV16mE7) protein with enhanced antigenicity and reduced transformation activity results in increased cytokine (TNF-α/IL-12/IL-6) secretion, IFN-γ secretion by T lymphocytes, and in vitro/in vivo tumoricidal activity against cervical cancer cells. Taken together, these results suggest that SIRPα-silenced DC vaccination presented potential therapeutic implications against cervical cancer.


Assuntos
Antígenos de Diferenciação/genética , Vacinas Anticâncer/administração & dosagem , Células Dendríticas/imunologia , Imunoterapia Ativa/métodos , Receptores Imunológicos/genética , Neoplasias do Colo do Útero/terapia , Animais , Antígenos de Diferenciação/metabolismo , Vacinas Anticâncer/genética , Vacinas Anticâncer/imunologia , Linhagem Celular Tumoral , Células Dendríticas/metabolismo , Feminino , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Camundongos , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/metabolismo , Receptores Imunológicos/metabolismo , Neoplasias do Colo do Útero/imunologia , Neoplasias do Colo do Útero/virologia , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...