Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
1.
BMC Oral Health ; 24(1): 525, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38702623

RESUMO

OBJECTIVE: To evaluate the antibacterial effectiveness of a combination of ε-poly-L-lysine (ε-PL), funme peptide (FP) as well as domiphen against oral pathogens, and assess the efficacy of a BOP® mouthwash supplemented with this combination in reducing halitosis and supragingival plaque in a clinical trial. MATERIALS AND METHODS: The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of the compound against Fusobacterium nucleatum, Porphyromonas gingivalis, Streptococcus mutans, and Aggregatibacter actinomycetemcomitans were determined by the gradient dilution method. Subsequently, the CCK-8 assay was used to detect the toxicity of mouthwash on human gingival fibroblastst, and the effectiveness in reducing halitosis and supragingival plaque of the mouthwash supplemented with the combination was analyzed by a randomized, double-blind, parallel-controlled clinical trial. RESULTS: The combination exhibited significant inhibitory effects on tested oral pathogens with the MIC < 1.56% (v/v) and the MBC < 3.13% (v/v), and the mouthwash containing this combination did not inhibit the viability of human gingival fibroblasts at the test concentrations. The clinical trial showed that the test group displayed notably lower volatile sulfur compounds (VSCs) at 0, 10, 24 h, and 7 d post-mouthwash (P < 0.05), compared with the baseline. After 7 days, the VSC levels of the and control groups were reduced by 50.27% and 32.12%, respectively, and notably cutting severe halitosis by 57.03% in the test group. Additionally, the Plaque Index (PLI) of the test and control group decreased by 54.55% and 8.38%, respectively, and there was a significant difference in PLI between the two groups after 7 days (P < 0.01). CONCLUSIONS: The combination of ε-PL, FP and domiphen demonstrated potent inhibitory and bactericidal effects against the tested oral pathogens, and the newly formulated mouthwash added with the combination exhibited anti-dental plaque and anti-halitosis properties in a clinical trial and was safe. TRIAL REGISTRATION: The randomized controlled clinical trial was registered on Chinese Clinical Trial Registry (No. ChiCTR2300073816, Date: 21/07/2023).


Assuntos
Placa Dentária , Halitose , Antissépticos Bucais , Polilisina , Humanos , Halitose/prevenção & controle , Halitose/tratamento farmacológico , Halitose/microbiologia , Antissépticos Bucais/uso terapêutico , Placa Dentária/microbiologia , Placa Dentária/prevenção & controle , Método Duplo-Cego , Masculino , Feminino , Polilisina/uso terapêutico , Adulto , Testes de Sensibilidade Microbiana , Adulto Jovem , Antibacterianos/uso terapêutico , Antibacterianos/farmacologia , Porphyromonas gingivalis/efeitos dos fármacos , Fusobacterium nucleatum/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Peptídeos/uso terapêutico , Peptídeos/farmacologia , Aggregatibacter actinomycetemcomitans/efeitos dos fármacos , Streptococcus mutans/efeitos dos fármacos
2.
J Exp Clin Cancer Res ; 40(1): 213, 2021 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-34172082

RESUMO

Immunotherapy is currently under intensive investigation as a potential breakthrough treatment option for glioblastoma. Given the anatomical and immunological complexities surrounding glioblastoma, lymphocytes that infiltrate the brain to develop durable immunity with memory will be key. Polyinosinic:polycytidylic acid, or poly(I:C), and its derivative poly-ICLC could serve as a priming or boosting therapy to unleash lymphocytes and other factors in the (immuno)therapeutic armory against glioblastoma. Here, we present a systematic review on the effects and efficacy of poly(I:C)/poly-ICLC for glioblastoma treatment, ranging from preclinical work on cellular and murine glioblastoma models to reported and ongoing clinical studies. MEDLINE was searched until 15 May 2021 to identify preclinical (glioblastoma cells, murine models) and clinical studies that investigated poly(I:C) or poly-ICLC in glioblastoma. A systematic review approach was conducted according to PRISMA guidelines. ClinicalTrials.gov was queried for ongoing clinical studies. Direct pro-tumorigenic effects of poly(I:C) on glioblastoma cells have not been described. On the contrary, poly(I:C) changes the immunological profile of glioblastoma cells and can also kill them directly. In murine glioblastoma models, poly(I:C) has shown therapeutic relevance as an adjuvant therapy to several treatment modalities, including vaccination and immune checkpoint blockade. Clinically, mostly as an adjuvant to dendritic cell or peptide vaccines, poly-ICLC has been demonstrated to be safe and capable of eliciting immunological activity to boost therapeutic responses. Poly-ICLC could be a valuable tool to enhance immunotherapeutic approaches for glioblastoma. We conclude by proposing several promising combination strategies that might advance glioblastoma immunotherapy and discuss key pre-clinical aspects to improve clinical translation.


Assuntos
Neoplasias Encefálicas/tratamento farmacológico , Carboximetilcelulose Sódica/análogos & derivados , Glioblastoma/tratamento farmacológico , Poli I-C/uso terapêutico , Polilisina/análogos & derivados , Animais , Neoplasias Encefálicas/imunologia , Vacinas Anticâncer/uso terapêutico , Carboximetilcelulose Sódica/uso terapêutico , Ensaios Clínicos como Assunto , Glioblastoma/imunologia , Humanos , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunoterapia , Camundongos , Polilisina/uso terapêutico
3.
Cancer Immunol Immunother ; 70(11): 3081-3091, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33751208

RESUMO

The nanoparticle complex of cholesteryl pullulan (CHP) and NY-ESO-1 antigen protein (CHP-NY-ESO-1) presents multiple epitope peptides to MHC class I and II pathways, leading to CD8+ and CD4+ T cell responses. Poly-ICLC is a synthetic, double-stranded RNA, an agonist of toll-like receptor (TLR)-3, and a cytoplasmic receptor of melanoma differentiation-associated gene (MDA)-5. It should be a suitable immune adjuvant of cancer vaccine to overcome the inhibitory tumor microenvironment. We conducted a phase 1 clinical trial of CHP-NY-ESO-1 with poly-ICLC in patients with advanced or recurrent esophageal cancer. CHP-NY-ESO-1/poly-ICLC (µg/mg) was administered at a dose of 200/0.5 or 200/1.0 (cohorts 1 and 2, respectively) every 2 weeks for a total of six doses. The primary endpoints were safety and immune response. The secondary endpoint was tumor response. In total, 16 patients were enrolled, and six patients in each cohort completed the trial. The most common adverse event (AE) was injection site skin reaction (86.7%). No grade 3 or higher drug-related AEs were observed. No tumor responses were observed, and three patients (30%) had stable disease. The immune response was comparable between the two cohorts, and all patients (100%) achieved antibody responses with a median of 2.5 vaccinations. Comparing CHP-NY-ESO-1 alone to the poly-ICLC combination, all patients in both groups exhibited antibody responses, but the titers were higher in the combination group. In a mouse model, adding anti-PD-1 antibody to the combination of CHP-NY-ESO-1/poly-ICLC suppressed the growth of NY-ESO-1-expressing tumors. Combining the vaccine with PD-1 blockade holds promise in human trials.


Assuntos
Antígenos de Neoplasias/uso terapêutico , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Carboximetilcelulose Sódica/análogos & derivados , Neoplasias Esofágicas/tratamento farmacológico , Glucanos/uso terapêutico , Proteínas de Membrana/uso terapêutico , Poli I-C/uso terapêutico , Polilisina/análogos & derivados , Adjuvantes Imunológicos/uso terapêutico , Idoso , Idoso de 80 Anos ou mais , Animais , Antígenos de Neoplasias/imunologia , Carboximetilcelulose Sódica/uso terapêutico , Neoplasias Esofágicas/imunologia , Feminino , Glucanos/imunologia , Humanos , Indutores de Interferon/imunologia , Indutores de Interferon/uso terapêutico , Masculino , Proteínas de Membrana/imunologia , Camundongos , Pessoa de Meia-Idade , Nanopartículas , Poli I-C/imunologia , Polilisina/imunologia , Polilisina/uso terapêutico
4.
J Mater Chem B ; 8(41): 9492-9501, 2020 10 28.
Artigo em Inglês | MEDLINE | ID: mdl-33001118

RESUMO

Developing new strategies to enhance drug accumulation in the tumor and therapeutic efficacy is of great importance in the field of tumor therapy. Herein, a peanut-like multifunctional nanomedicine (CuS-PGH NMs) made of CuS nanoparticles encapsulated in poly(l-lysine)(PLL)/glucose oxidase (GOx)-hyaluronic acid (HA) shells has been constructed via layer-by-layer (LbL) assembly, and shows good biocompatibility and effective multi-gradient therapy. Because of the enhanced permeability and retention (EPR) effect, the CuS-PGH NMs could significantly enhance the cellular uptake by tumors overexpressing CD44 receptors, which respond to hyaluronidase (HAase)-triggered surface charge conversion. Once internalized by the tumor, GOx was the first to be exposed and could effectively deplete endogenous glucose for starvation therapy, and the excess H2O2 was then converted into highly toxic hydroxyl radicals (˙OH) via a Cu+-mediated Fenton-like reaction for chemodynamic therapy (CDT). Meanwhile, the as-obtained Cu+ ions accompanied the regenerated less-active Cu2+ ions. Interestingly, the high content of H2O2 could, in turn, accelerate Cu2+/Cu+ conversion to promote the Cu+-H2O2 reaction for enhanced chemodynamic therapy (CDT), thereby achieving efficient tumor growth suppression via synergistic starvation/CDT therapy. Subsequently, owing to the strong NIR-II absorption capability of CuS-PGH NMs, effective photothermal tumor ablation of the weakened tumor cells could be realized with the precise guidance of NIR-II PAI. This multi-gradient therapeutic strategy has been demonstrated to have excellent antitumor activity with minimal nonspecific damages, and offers a new avenue to precise tumor therapy.


Assuntos
Neoplasias da Mama/terapia , Nanopartículas/uso terapêutico , Nanomedicina Teranóstica/métodos , Animais , Neoplasias da Mama/diagnóstico por imagem , Linhagem Celular Tumoral , Cobre/uso terapêutico , Feminino , Glucose Oxidase/uso terapêutico , Ácido Hialurônico/uso terapêutico , Camundongos , Técnicas Fotoacústicas , Polilisina/uso terapêutico
5.
Semin Immunol ; 49: 101414, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-33011064

RESUMO

Immunotherapies have become the first line of treatment for many cancer types. Unfortunately, only a small fraction of patients benefits from these therapies. This low rate of success can be attributed to 3 main barriers: 1) low frequency of anti-tumor specific T cells; 2) lack of infiltration of the anti-tumor specific T cells into the tumor parenchyma and 3) accumulation of highly suppressive cells in the tumor mass that inhibit the effector function of the anti-tumor specific T cells. Thus, the identification of immunomodulators that can increase the frequency and/or the infiltration of antitumor specific T cells while reducing the suppressive capacity of the tumor microenvironment is necessary to ensure the effectiveness of T cell immunotherapies. In this review, we discuss the potential of poly-ICLC as a multi-functional immune modulator for treating cancer and its impact on the 3 above mentioned barriers. We describe the unique capacity of poly-ICLC in stimulating 2 separate pattern recognition receptors, TLR3 and cytosolic MDA5 and the consequences of these activations on cytokines and chemokines production. We emphasize the role of poly-ICLC as an adjuvant in the setting of peptide-based cancer vaccines and in situ tumor vaccination by mimicking natural immune responses to infections. Finally, we summarize the impact of poly-ICLC in enhancing T infiltration into the tumor parenchyma and address the implication of this finding in the clinic.


Assuntos
Antineoplásicos/farmacologia , Carboximetilcelulose Sódica/análogos & derivados , Fatores Imunológicos/farmacologia , Imunomodulação , Poli I-C/imunologia , Poli I-C/farmacologia , Polilisina/análogos & derivados , Animais , Antineoplásicos/uso terapêutico , Carboximetilcelulose Sódica/farmacologia , Carboximetilcelulose Sódica/uso terapêutico , Citocinas/metabolismo , Humanos , Imunidade Inata/efeitos dos fármacos , Fatores Imunológicos/uso terapêutico , Imunomodulação/efeitos dos fármacos , Helicase IFIH1 Induzida por Interferon/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Neoplasias/tratamento farmacológico , Neoplasias/etiologia , Neoplasias/metabolismo , Neoplasias/patologia , Poli I-C/uso terapêutico , Polilisina/imunologia , Polilisina/farmacologia , Polilisina/uso terapêutico , Receptores de Reconhecimento de Padrão/metabolismo , Receptor 3 Toll-Like/metabolismo
6.
Urol Clin North Am ; 47(4S): e1-e8, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33446322

RESUMO

Relatively simple, synthetic, double-stranded RNAs can be powerful viral pathogen-associated molecular pattern (PAMP) mimics, inducing a panoply of antiviral and antitumor responses that act at multiple stages of host defense. Their mechanisms of action and uses are beginning to be understood, alone, in combination with other therapeutics, or as novel PAMP-adjuvants providing the critical danger signal that has been missing from most cancer and other modern vaccines. Dose, timing, route of administration combinations, and other clinical variables can have a critical impact on immunogenicity. This article reviews advances in the use of polyinosinic-polycytidylic acid and derivatives, in particular poly-ICLC.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Carboximetilcelulose Sódica/análogos & derivados , Fatores Imunológicos/uso terapêutico , Poli I-C/uso terapêutico , Polilisina/análogos & derivados , Neoplasias da Próstata/terapia , RNA de Cadeia Dupla/uso terapêutico , Adjuvantes Imunológicos/fisiologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Carboximetilcelulose Sódica/uso terapêutico , Ensaios Clínicos como Assunto , Humanos , Fatores Imunológicos/imunologia , Masculino , Moléculas com Motivos Associados a Patógenos/imunologia , Moléculas com Motivos Associados a Patógenos/uso terapêutico , Poli I-C/imunologia , Polilisina/imunologia , Polilisina/uso terapêutico , Neoplasias da Próstata/imunologia , RNA de Cadeia Dupla/imunologia
7.
Nano Lett ; 19(8): 5515-5523, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31362507

RESUMO

Designing simple-structured nanomedicine without lacking key functionalities, thereby avoiding incomplete damage or relapse of tumor with the administration of a safe dose, is pivotal for successful cancer nanotherapy. We herein presented a nanomedicine of photodynamic therapy (PDT) that simply assembled amphiphilic macromolecules of poly-l-lysine conjugating with photosensitizers onto hydrophobic upconverting nanoparticles. We demonstrated that the nanoformulation, despite its simple structure and synthesis, simultaneously possesses multiple features, including substantial payload of photosensitizers, avid cellular internalization both in vitro and in vivo, efficient diffusion and broad distribution in tumor lesion, and potent fatality for cancer stem cells that are refractory to other therapy modalities. Because of the combination of these functionalities, the tumors in mice were eradicated and no relapse was observed after at least 40 days, just with an extremely low intraperitoneal injection dose of 5.6 mg/kg. Our results suggested a strategy for designing multifunctional nanomedicines with simple construct and efficacious therapeutic response and presented the promising potential of PDT for a radical cure of cancer.


Assuntos
Nanoconjugados/uso terapêutico , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/uso terapêutico , Animais , Linhagem Celular Tumoral , Células HeLa , Humanos , Camundongos , Nanoconjugados/administração & dosagem , Nanoconjugados/química , Recidiva Local de Neoplasia/prevenção & controle , Células-Tronco Neoplásicas/efeitos dos fármacos , Fotoquimioterapia , Fármacos Fotossensibilizantes/administração & dosagem , Fármacos Fotossensibilizantes/química , Polilisina/administração & dosagem , Polilisina/análogos & derivados , Polilisina/uso terapêutico
8.
Nat Med ; 25(5): 814-824, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30962585

RESUMO

Indolent non-Hodgkin's lymphomas (iNHLs) are incurable with standard therapy and are poorly responsive to checkpoint blockade. Although lymphoma cells are efficiently killed by primed T cells, in vivo priming of anti-lymphoma T cells has been elusive. Here, we demonstrate that lymphoma cells can directly prime T cells, but in vivo immunity still requires cross-presentation. To address this, we developed an in situ vaccine (ISV), combining Flt3L, radiotherapy, and a TLR3 agonist, which recruited, antigen-loaded and activated intratumoral, cross-presenting dendritic cells (DCs). ISV induced anti-tumor CD8+ T cell responses and systemic (abscopal) cancer remission in patients with advanced stage iNHL in an ongoing trial ( NCT01976585 ). Non-responding patients developed a population of PD1+CD8+ T cells after ISV, and murine tumors became newly responsive to PD1 blockade, prompting a follow-up trial of the combined therapy. Our data substantiate that recruiting and activating intratumoral, cross-priming DCs is achievable and critical to anti-tumor T cell responses and PD1-blockade efficacy.


Assuntos
Vacinas Anticâncer , Linfoma de Células B/terapia , Adulto , Idoso , Animais , Apresentação de Antígeno , Linfócitos T CD8-Positivos/imunologia , Carboximetilcelulose Sódica/análogos & derivados , Carboximetilcelulose Sódica/uso terapêutico , Linhagem Celular Tumoral , Terapia Combinada , Células Dendríticas/imunologia , Feminino , Humanos , Imunoterapia Adotiva , Linfoma de Células B/imunologia , Linfoma de Células B/patologia , Masculino , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Poli I-C/uso terapêutico , Polilisina/análogos & derivados , Polilisina/uso terapêutico , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptor de Morte Celular Programada 1/imunologia , Receptor 3 Toll-Like/agonistas , Vacinação
9.
Front Immunol ; 10: 725, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31024557

RESUMO

Objective: Toll-like receptor-3 agonist Poly-ICLC has been known to activate immune cells and induce HIV replication in pre-clinical experiments. In this study we investigated if Poly-ICLC could be used for disrupting HIV latency while simultaneously enhancing innate immune responses. Design: This was a randomized, placebo-controlled, double-blinded trial in aviremic, cART-treated HIV-infected subjects. Participants (n = 15) were randomized 3:1 to receive two consecutive daily doses of Poly-ICLC (1.4 mg subcutaneously) vs. placebo. Subjects were observed for adverse events, immune activation, and viral replication. Methods: Besides primary outcomes of safety and tolerability, several longitudinal immune parameters were evaluated including immune cell phenotype and function via flowcytometry, ELISA, and transcriptional profiling. PCR assays for plasma HIV-1 RNA, CD4+ T cell-associated HIV-1 RNA, and proviral DNA were performed to measure HIV reservoirs and latency. Results: Poly-ICLC was overall safe and well-tolerated. Poly-ICLC-related adverse events were Grade 1/2, with the exception of one Grade 3 neutropenia which was short-lived. Mild Injection site reactions were observed in nearly all participants in the Poly-ICLC arm. Transcriptional analyses revealed upregulation of innate immune pathways in PBMCs following Poly-ICLC treatment, including strong interferon signaling accompanied by transient increases in circulating IP-10 (CXCL10) levels. These responses generally peaked by 24-48 h after the first injection and returned to baseline by day 8. CD4+ T cell number and phenotype were unchanged, plasma viral control was maintained and no significant effect on HIV reservoirs was observed. Conclusions: These finding suggest that Poly-ICLC could be safely used for inducing transient innate immune responses in treated HIV+ subjects indicating promise as an adjuvant for HIV therapeutic vaccines. Trial Registration: www.ClinicalTrials.gov, identifier: NCT02071095.


Assuntos
Carboximetilcelulose Sódica/análogos & derivados , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Imunidade Inata/efeitos dos fármacos , Poli I-C/uso terapêutico , Polilisina/análogos & derivados , Receptor 3 Toll-Like/metabolismo , Adulto , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Carboximetilcelulose Sódica/uso terapêutico , Método Duplo-Cego , Feminino , HIV/imunologia , Infecções por HIV/metabolismo , Humanos , Masculino , Pessoa de Meia-Idade , Polilisina/uso terapêutico
10.
BMB Rep ; 52(5): 324-329, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30293549

RESUMO

Recent progress in cellular reprogramming technology and lineage-specific cell differentiation has provided great opportunities for translational research. Because virus-based gene delivery is not a practical reprogramming protocol, protein-based reprogramming has been receiving attention as a safe way to generate reprogrammed cells. However, the poor efficiency of the cellular uptake of reprogramming proteins is still a major obstacle. Here, we reported key factors which improve the cellular uptake of these proteins. Purified red fluorescent proteins fused with 9xLysine (dsRED-9K) as a cell penetrating peptide were efficiently delivered into the diverse primary cells. Protein delivery was improved by the addition of amodiaquine. Furthermore, purified dsRED-9K was able to penetrate all cell lineages derived from mouse embryonic stem cells efficiently. Our data may provide important insights into the design of protein-based reprogramming or differentiation protocols [BMB Reports 2019; 52(5): 324-329].


Assuntos
Peptídeos Penetradores de Células/metabolismo , Técnicas de Reprogramação Celular/métodos , Polilisina/metabolismo , Amodiaquina/farmacologia , Animais , Técnicas de Cultura de Células , Diferenciação Celular/genética , Peptídeos Penetradores de Células/farmacologia , Reprogramação Celular/genética , Células-Tronco Embrionárias/citologia , Fibroblastos/metabolismo , Técnicas de Transferência de Genes , Células HEK293 , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Peptídeos/uso terapêutico , Polilisina/uso terapêutico , Fatores de Transcrição/metabolismo
11.
Kidney Blood Press Res ; 43(4): 1273-1284, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30078011

RESUMO

BACKGROUND/AIMS: Myocardial ischemia/reperfusion (I/R) injury (MI/RI) is a critical cause of death in patients with heart disease. However, the pharmaco-therapeutical outcome for MI/RI remains unsatisfactory. Innovative approaches for enhancing drug sensitivity and recovering myocardial function in MI/RI treatment are urgently needed. The purpose of this study was to evaluate the protective effects of exenatide-loaded poly(L-lysine)-poly(ethylene glycol)-poly(L-lysine) (PLL-PEG-PLL) nanoparticles (NPs) against MI/RI. METHODS: The size of PLL-PEG-PLL NPs and the loading and release rates of exenatide were determined. The in vitro NP cytotoxicity was evaluated using newborn rat cardiomyocytes. Rats pretreated with free exenatide or exenatide/PLL-PEG-PLL polyplexes were subjected to 0.5-h ischemia and 2-h reperfusion in the left anterior descending coronary artery. The histopathologic lesions were assessed using hematoxylin-eosin staining. The general physiological indices, including blood pressure (BP), heart rate (HR), the left ventricular ejection fraction (LVEF) and end-diastolic pressure (LEVDP), and the left ventricular pressure maximal rate of rising (dp/dtmax), were monitored using a non-invasive blood pressure analyzer and color Doppler echocardiography. The antioxidative activity in the myocardial tissue was measured. The myocardial enzymatic activity was further estimated by determining the serum levels of creatine kinase (CK), lactate dehydrogenase (LDH), cardiac troponin T (cTnT), and glucagon-like peptide-1 (GLP-1), as well as the expression of GLP-1R in the myocardial tissue. RESULTS: Exenatide preconditioning attenuated the oxidative stress injury and promoted the myocardial function in I/R-induced myocardial injury, while the application of block copolymer PLL-PEG-PLL as a potential exenatide nanocarrier with sustained release significantly enhanced the bioavailability of exenatide. CONCLUSION: The block copolymer PLL-PEG-PLL may function as a potent exenatide nanocarrier for augmenting pharmacotherapy against MI/RI with unprecedented clinical benefits. Further study is needed to better clarify the underlying mechanisms.


Assuntos
Precondicionamento Isquêmico Miocárdico/métodos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Nanopartículas/química , Peptídeos/farmacologia , Peçonhas/farmacologia , Animais , Animais Recém-Nascidos , Portadores de Fármacos/química , Exenatida , Hipoglicemiantes , Incretinas , Masculino , Estresse Oxidativo/efeitos dos fármacos , Peptídeos/farmacocinética , Peptídeos/uso terapêutico , Polietilenoglicóis/uso terapêutico , Polilisina/análogos & derivados , Polilisina/uso terapêutico , Ratos , Ratos Sprague-Dawley , Peçonhas/farmacocinética , Peçonhas/uso terapêutico
12.
Theranostics ; 8(11): 2974-2987, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29896297

RESUMO

Although there is ample evidence that the chemotherapeutic drugs trigger an immune response, the efficient tumor rejection or regression is not guaranteed probably due to the massive immunosuppression within the tumor microenvironment. Thus, a rational delivery platform that overcomes immunosuppression is needed to maximally achieve both cytotoxic and immune-modulatory functions of chemotherapeutics. Accumulating evidence suggests that platinum-based drugs might be suitable for this application. Methods: The dendrigraft polylysine (DGL) with its uniform size and multifunctional groups was employed as the polymeric core and conjugated with platinum-based compounds as therapeutics and WKYMVm peptide (Wpep) as a targeting ligand to construct the novel delivery platform Wpep-DGL/Pt. A series of in vitro and in vivo analyses, including physical and chemical characterizations, targeting property, biosafety, and antitumor efficacy of Wpep-DGL/Pt were systematically carried out. Results: Wpep-DGL/Pt showed potent antitumor efficacy in MDA-MB-231 cells tumor-bearing nude mice with a deficient immune system, demonstrating targeted delivery of chemotherapeutics and the resultant cytotoxicity. Furthermore, in immunocompetent mice bearing 4T1 cells tumors, Wpep-DGL/Pt activated immune cells and induced cell death proving their dual function of chemotherapeutic and immunomodulatory efficacy. Conclusion: This work represents a novel approach for cancer immunotherapy by integrating nanotechnology and platinum-based therapeutics which not only efficiently exerts the chemotherapeutic cytotoxic effect on tumor cell but also restores immune response of immunological cells within the tumor microenvironment.


Assuntos
Adjuvantes Imunológicos/uso terapêutico , Antineoplásicos/uso terapêutico , Neoplasias/terapia , Peptídeos/uso terapêutico , Platina/uso terapêutico , Adjuvantes Imunológicos/química , Animais , Antineoplásicos/química , Composição de Medicamentos , Sistemas de Liberação de Medicamentos , Feminino , Imunidade Humoral , Imunoterapia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Nanopartículas , Neoplasias/imunologia , Peptídeos/química , Platina/química , Polilisina/química , Polilisina/uso terapêutico , Microambiente Tumoral
13.
Biomed Pharmacother ; 102: 1064-1076, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29710523

RESUMO

PURPOSE: The present study, attempts to validate the molecular mechanism(s) of Poly-l-lysine (PLL) induced apoptosis, anti-proliferative and anti-tumorigenic properties in in-vitro HUVECs cells and Dalton's Ascitic Lymphoma (DAL) and in in-vivo DAL cell bearing BALB/c mice model. MATERIALS AND METHODS: The cell proliferation assay and morphological assay was carried out using the MTT assay and Giemsa staining method. The antitumor activity of PLL was evaluated in BALB/c mice at 20 and 40 mg/kg/b.w doses for 21 days for DAL solid tumor model. Several tumor evaluation endpoints, hematological and biochemical parameters were estimated. Additionally, the tumor apoptosis, anti-proliferative and anti-tumor angiogenesis effects were assessed using western blots and immunohistochemistry. RESULTS: PLL significantly decreased cell proliferation in in-vitro HUVECs and DAL cells without significant effects on normal cell growth. PLL also induced alteration in cellular morphology in DAL cells. Therafter, in the BALB/c mouse model, PLL had noticeable inhibition in DAL-induced tumorigenesis. This inhibition was evident through reduced solid tumor volume and weight versus the control group. However, PLL promoted tumor apoptosis and suppressed cell-proliferation and tumor-angiogenesis. PLL also increased hematological markers significantly compared to 5-flurouracil (5-FU). The amount of TdT in the nuclei of DAL cells in mice treated with PLL was significantly increased while in contrast decreases of anti-apoptotic protein Bcl-2 expression were observed. PLL also significantly upregulated the pro-apoptotic protein Bax and activated caspase-3. Measurable decreases of cyclin-D1 were observed through PLL treatments, an indicator of cell-cycle arrest. These studies also indicate PLL's induction and anti-proliferative effects through suppression of the c-Myc and Ki-67 proliferation-indices. Additionally, PLL inhibited tumor-angiogenesis through suppression of VEGF and CD34 protein expression levels and reduction ofmicrovesseldensityversus similar parameters in tumors from control mice. CONCLUSION: The present study offers opportunities and hopes for possible anti-tumortherapies with PLL in the near future and warrants further formulation developments.


Assuntos
Apoptose , Ascite/patologia , Carcinogênese/patologia , Regulação para Baixo , Linfoma/tratamento farmacológico , Linfoma/patologia , Neovascularização Patológica/tratamento farmacológico , Polilisina/uso terapêutico , Animais , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Apoptose/efeitos dos fármacos , Carcinogênese/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Forma Celular/efeitos dos fármacos , Ciclina D1/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Feminino , Células Endoteliais da Veia Umbilical Humana/efeitos dos fármacos , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Estimativa de Kaplan-Meier , Antígeno Ki-67/metabolismo , Linfoma/sangue , Camundongos Endogâmicos BALB C , Neovascularização Patológica/sangue , Polilisina/administração & dosagem , Polilisina/química , Polilisina/farmacologia , Análise de Sobrevida
14.
J Immunother Cancer ; 5(1): 92, 2017 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-29157306

RESUMO

BACKGROUND: Breast cancer remains a leading cause of cancer death worldwide. There is evidence that immunotherapy may play a role in the eradication of residual disease. Peptide vaccines for immunotherapy are capable of durable immune memory, but vaccines alone have shown sparse clinical activity against breast cancer to date. Toll-like receptor (TLR) agonists and helper peptides are excellent adjuvants for vaccine immunotherapy and they are examined in this human clinical trial. METHODS: A vaccine consisting of 9 MHC class I-restricted breast cancer-associated peptides (from MAGE-A1, -A3, and -A10, CEA, NY-ESO-1, and HER2 proteins) was combined with a TLR3 agonist, poly-ICLC, along with a helper peptide derived from tetanus toxoid. The vaccine was administered on days 1, 8, 15, 36, 57, 78. CD8+ T cell responses to the vaccine were assessed by both direct and stimulated interferon gamma ELIspot assays. RESULTS: Twelve patients with breast cancer were treated: five had estrogen receptor positive disease and five were HER2 amplified. There were no dose-limiting toxicities. Toxicities were limited to Grade 1 and Grade 2 and included mild injection site reactions and flu-like symptoms, which occurred in most patients. The most common toxicities were injection site reaction/induration and fatigue, which were experienced by 100% and 92% of participants, respectively. In the stimulated ELIspot assays, peptide-specific CD8+ T cell responses were detected in 4 of 11 evaluable patients. Two patients had borderline immune responses to the vaccine. The two peptides derived from CEA were immunogenic. No difference in immune response was evident between patients receiving endocrine therapy and those not receiving endocrine therapy during the vaccine series. CONCLUSIONS: Peptide vaccine administered in the adjuvant breast cancer setting was safe and feasible. The TLR3 adjuvant, poly-ICLC, plus helper peptide mixture provided modest immune stimulation. Further optimization is required for this multi-peptide vaccine/adjuvant combination. TRIAL REGISTRATION: ClinicalTrials.gov (posted 2/15/2012): NCT01532960. Registered 2/8/2012. https://clinicaltrials.gov/show/NCT01532960.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/imunologia , Vacinas Anticâncer/uso terapêutico , Carboximetilcelulose Sódica/análogos & derivados , Indutores de Interferon/uso terapêutico , Poli I-C/uso terapêutico , Polilisina/análogos & derivados , Adjuvantes Imunológicos , Adulto , Vacinas Anticâncer/farmacologia , Carboximetilcelulose Sódica/farmacologia , Carboximetilcelulose Sódica/uso terapêutico , Feminino , Humanos , Imunoterapia , Indutores de Interferon/farmacologia , Pessoa de Meia-Idade , Projetos Piloto , Poli I-C/farmacologia , Polilisina/farmacologia , Polilisina/uso terapêutico
15.
Kidney Blood Press Res ; 42(3): 495-508, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28854424

RESUMO

BACKGROUND/AIMS: The aim of this study was to assess the effect of human hepatocyte growth factor (hHGF)-loaded poly (ethylene glycol)-b-brush poly (l-lysine) (PEG-b-P(ELG-g-PLL)) copolymer on ischemia/reperfusion (I/R) injury to different organs. METHODS: The isoelectric point (pI) of hHGF is 5.5, and hHGF combined with PEG-b-P(ELG-g-PLL) copolymer via electrostatic interactions at pH 7.4. The synthesized PEG-b-P(ELG-g-PLL) copolymer was analyzed using 1H nuclear magnetic resonance (1H NMR) and gel permeation chromatography (GPC). The hHGF/PEG-b-P(ELG-g-PLL) complex was evaluated using a nanoparticle size instrument and transmission electron microscopy (TEM). In addition, vivo performance of hHGF/PEG-b-P(ELG-g-PLL) complex was evaluated using plasma hHGF concentration and different organs ischemia reperfusion injury in rats. RESULTS: An in vitro investigation showed that PEG-b-P(ELG-g-PLL) could serve as a potential hHGF nanocarrier with efficient encapsulation and sustained release. An additional in vivo investigation revealed that the hHGF/PEG-b-P(ELG-g-PLL) complex could prolong increases in plasma hHGF concentration and protect different organs (the brain, heart and kidney) against I/R injury. CONCLUSION: Poly (ethylene glycol)-block-brush poly (l-lysine) copolymer as an efficient nanocarrier for human hepatocyte growth factor with enhanced bioavailability and anti-ischemia reperfusion injury efficacy.


Assuntos
Portadores de Fármacos/química , Fator de Crescimento de Hepatócito/administração & dosagem , Polietilenoglicóis/uso terapêutico , Polilisina/análogos & derivados , Traumatismo por Reperfusão/tratamento farmacológico , Animais , Disponibilidade Biológica , Portadores de Fármacos/uso terapêutico , Fator de Crescimento de Hepatócito/farmacocinética , Humanos , Polilisina/uso terapêutico , Ratos
16.
Biomaterials ; 141: 210-222, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28689117

RESUMO

Magnetic hyperthermia was reported to increase the survival of patients with recurrent glioblastoma by 7 months. This promising result may potentially be further improved by using iron oxide nanoparticles, called magnetosomes, which are synthesized by magnetotactic bacteria, extracted from these bacteria, purified to remove most endotoxins and organic material, and then coated with poly-l-lysine to yield a stable and non-pyrogenic nanoparticle suspension. Due to their ferrimagnetic behavior, high crystallinity and chain arrangement, these magnetosomes coated with poly-l-lysine (M-PLL) are characterized by a higher heating power than their chemically synthesized counterparts currently used in clinical trials. M-PLL-enhanced antitumor efficacy was demonstrated by administering 500-700 µg in iron of M-PLL to intracranial U87-Luc tumors of 1.5 mm3 and by exposing mice to 27 magnetic sessions each lasting 30 min, during which an alternating magnetic field of 202 kHz and 27 mT was applied. Treatment conditions were adjusted to reach a typical hyperthermia temperature of 42 °C during the first magnetic session. In 100% of treated mice, bioluminescence due to living glioblastoma cells fully disappeared 68 days following tumor cell implantation (D68). These mice were all still alive at D350. Histological analysis of their brain tissues revealed an absence of tumor cells, suggesting that they were fully cured. In comparison, antitumor efficacy was less pronounced in mice treated by the administration of IONP followed by 23 magnetic sessions, leading to full tumor bioluminescence disappearance in only 20% of the treated mice.


Assuntos
Neoplasias Encefálicas/terapia , Materiais Revestidos Biocompatíveis/uso terapêutico , Óxido Ferroso-Férrico/uso terapêutico , Glioblastoma/terapia , Hipertermia Induzida/métodos , Magnetossomos/química , Polilisina/uso terapêutico , Células 3T3 , Animais , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Materiais Revestidos Biocompatíveis/química , Feminino , Óxido Ferroso-Férrico/química , Glioblastoma/patologia , Humanos , Campos Magnéticos , Magnetossomos/ultraestrutura , Magnetospirillum/química , Camundongos , Camundongos Nus , Polilisina/análogos & derivados
17.
Anticancer Res ; 37(5): 2195-2200, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28476782

RESUMO

BACKGROUND: Chemotherapy is an effective option to treat recurrent or metastatic cancer but its debilitating side-effects limit the dose and time of exposure. Prodrugs that can be activated locally by an activating enzyme can minimize collateral damage from chemotherapy. We previously demonstrated the efficacy of a poly-L-lysine-based theranostic nanoplex containing bacterial cytosine deaminase (bCD) that locally converted 5-fluorocytosine (5-FC) to the chemotherapeutic agent 5-fluorouracil in MDA-MB-231 primary tumor xenografts. MATERIALS AND METHODS: Here we used a more effective variant of bCD to target metastatic red fluorescence protein expressing MDA-MB-435 cells in the lungs. We used an intravenous injection of tumor cells and monitored tumor growth in the lungs for 5 weeks by which time metastatic nodules were detected with optical imaging. The animals were then treated with the bCD-nanoplex and 5-FC. RESULTS: We observed a significant decrease in metastatic burden with a single dose of the enzyme-nanoplex and two consecutive prodrug injections. CONCLUSION: These results are a first step towards the longitudinal evaluation of such a strategy with multiple doses. Additionally, the enzyme can be directly coupled to imaging reporters to time prodrug administration for the detection and treatment of aggressive metastatic cancer.


Assuntos
Antineoplásicos/administração & dosagem , Citosina Desaminase/administração & dosagem , Proteínas de Escherichia coli/administração & dosagem , Fluoruracila/administração & dosagem , Neoplasias Pulmonares/tratamento farmacológico , Pró-Fármacos/administração & dosagem , Animais , Antineoplásicos/química , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Citosina Desaminase/química , Citosina Desaminase/uso terapêutico , Progressão da Doença , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/química , Portadores de Fármacos/uso terapêutico , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/uso terapêutico , Feminino , Fluoruracila/química , Fluoruracila/uso terapêutico , Humanos , Neoplasias Pulmonares/patologia , Camundongos SCID , Nanoestruturas/administração & dosagem , Nanoestruturas/química , Nanoestruturas/uso terapêutico , Polietilenoimina/administração & dosagem , Polietilenoimina/química , Polietilenoimina/uso terapêutico , Polilisina/administração & dosagem , Polilisina/química , Polilisina/uso terapêutico , Pró-Fármacos/química , Pró-Fármacos/uso terapêutico
18.
Neuro Oncol ; 18(8): 1157-68, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26984745

RESUMO

BACKGROUND: Low-grade gliomas (LGGs) are the most common brain tumors of childhood. Although surgical resection is curative for well-circumscribed superficial lesions, tumors that are infiltrative or arise from deep structures are therapeutically challenging, and new treatment approaches are needed. Having identified a panel of glioma-associated antigens (GAAs) overexpressed in these tumors, we initiated a pilot trial of vaccinations with peptides for GAA epitopes in human leukocyte antigen-A2+ children with recurrent LGG that had progressed after at least 2 prior regimens. METHODS: Peptide epitopes for 3 GAAs (EphA2, IL-13Rα2, and survivin) were emulsified in Montanide-ISA-51 and administered subcutaneously adjacent to intramuscular injections of polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose every 3 weeks for 8 courses, followed by booster vaccines every 6 weeks. Primary endpoints were safety and T-lymphocyte responses against GAA epitopes. Treatment response was evaluated clinically and by MRI. RESULTS: Fourteen children were enrolled. Other than grade 3 urticaria in one child, no regimen-limiting toxicity was encountered. Vaccination induced immunoreactivity to at least one vaccine-targeted GAA in all 12 evaluable patients: to IL-13Rα2 in 3, EphA2 in 11, and survivin in 3. One child with a metastatic LGG had asymptomatic pseudoprogression noted 6 weeks after starting vaccination, followed by dramatic disease regression with >75% shrinkage of primary tumor and regression of metastatic disease, persisting >57 months. Three other children had sustained partial responses, lasting >10, >31, and >45 months, and one had a transient response. CONCLUSIONS: GAA peptide vaccination in children with recurrent LGGs is generally well tolerated, with preliminary evidence of immunological and clinical activity.


Assuntos
Antígenos de Neoplasias/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/imunologia , Carboximetilcelulose Sódica/análogos & derivados , Glioma/tratamento farmacológico , Glioma/imunologia , Indutores de Interferon/uso terapêutico , Poli I-C/uso terapêutico , Polilisina/análogos & derivados , Vacinação/métodos , Adolescente , Antígenos de Neoplasias/administração & dosagem , Antígenos de Neoplasias/efeitos adversos , Antígenos de Neoplasias/imunologia , Carboximetilcelulose Sódica/administração & dosagem , Carboximetilcelulose Sódica/efeitos adversos , Carboximetilcelulose Sódica/uso terapêutico , Criança , Pré-Escolar , Intervalo Livre de Doença , Epitopos , Feminino , Humanos , Lactente , Proteínas Inibidoras de Apoptose/imunologia , Indutores de Interferon/administração & dosagem , Indutores de Interferon/efeitos adversos , Indutores de Interferon/imunologia , Subunidade alfa2 de Receptor de Interleucina-13/imunologia , Masculino , Gradação de Tumores , Projetos Piloto , Poli I-C/administração & dosagem , Poli I-C/efeitos adversos , Poli I-C/imunologia , Polilisina/administração & dosagem , Polilisina/efeitos adversos , Polilisina/imunologia , Polilisina/uso terapêutico , Receptor EphA2/imunologia , Survivina , Resultado do Tratamento
19.
Stroke ; 47(1): 262-6, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26564103

RESUMO

BACKGROUND AND PURPOSE: Preconditioning with poly-l-lysine and carboxymethylcellulose (ICLC) provides robust neuroprotection from cerebral ischemia in a mouse stroke model. However, the receptor that mediates neuroprotection is unknown. As a synthetic double-stranded RNA, poly-ICLC may bind endosomal Toll-like receptor 3 or one of the cytosolic retinoic acid-inducible gene-I-like receptor family members, retinoic acid-inducible gene-I, or melanoma differentiation-associated protein 5. Activation of these receptors culminates in type I interferons (IFN-α/ß) induction-a response required for poly-ICLC-induced neuroprotection. In this study, we investigate the receptor required for poly-ICLC-induced neuroprotection. METHODS: Toll-like receptor 3, melanoma differentiation-associated protein 5-, and IFN-promoter stimulator 1-deficient mice were treated with poly-ICLC 24 hours before middle cerebral artery occlusion. Infarct volume was measured 24 hours after stroke to identify the receptor signaling pathways involved in protection. IFN-α/ß induction was measured in plasma samples collected 6 hours after poly-ICLC treatment. IFN-ß-deficient mice were used to test the requirement of IFN-ß for poly-ICLC-induced neuroprotection. Mice were treated with recombinant IFN-α-A to test the role of IFN-α as a potential mediator of neuroprotection. RESULTS: Poly-ICLC induction of both neuroprotection and systemic IFN-α/ß requires the cytosolic receptor melanoma differentiation-associated protein 5 and the adapter molecule IFN-promoter stimulator 1, whereas it is independent of Toll-like receptor 3. IFN-ß is not required for poly-ICLC-induced neuroprotection. IFN-α treatment protects against stroke. CONCLUSIONS: Poly-ICLC preconditioning is mediated by melanoma differentiation-associated protein 5 and its adaptor molecule IFN-promoter stimulator 1. This is the first evidence that a cytosolic receptor can mediate neuroprotection, providing a new target for the development of therapeutic agents to protect the brain from ischemic injury.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , RNA Helicases DEAD-box/metabolismo , Precondicionamento Isquêmico/métodos , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/prevenção & controle , Animais , Carboximetilcelulose Sódica/análogos & derivados , Carboximetilcelulose Sódica/metabolismo , Carboximetilcelulose Sódica/uso terapêutico , Helicase IFIH1 Induzida por Interferon , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fármacos Neuroprotetores/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Poli I-C/metabolismo , Poli I-C/uso terapêutico , Polilisina/análogos & derivados , Polilisina/metabolismo , Polilisina/uso terapêutico
20.
Pharmacol Ther ; 146: 120-31, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25281915

RESUMO

Although cancer vaccination has yielded promising results in patients, the objective response rates are low. The right choice of adjuvant might improve the efficacy. Here, we review the biological rationale, as well as the preclinical and clinical results of polyinosinic:polycytidylic acid and its derivative poly-ICLC as cancer vaccine adjuvants. These synthetic immunological danger signals enhanced vaccine-induced anti-tumor immune responses and contributed to tumor elimination in animal tumor models and patients. Supported by these results, poly-ICLC-containing cancer vaccines are currently extensively studied in the ongoing trials, making it highly plausible that poly-ICLC will be part of the future approved cancer immunotherapies.


Assuntos
Adjuvantes Imunológicos , Vacinas Anticâncer , Carboximetilcelulose Sódica/análogos & derivados , Poli I-C , Polilisina/análogos & derivados , Adjuvantes Imunológicos/farmacologia , Adjuvantes Imunológicos/uso terapêutico , Animais , Vacinas Anticâncer/farmacologia , Vacinas Anticâncer/uso terapêutico , Carboximetilcelulose Sódica/farmacologia , Carboximetilcelulose Sódica/uso terapêutico , Humanos , Poli I-C/farmacologia , Poli I-C/uso terapêutico , Polilisina/farmacologia , Polilisina/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA