Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
2.
Nature ; 627(8004): 646-655, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38418879

RESUMO

Tiragolumab, an anti-TIGIT antibody with an active IgG1κ Fc, demonstrated improved outcomes in the phase 2 CITYSCAPE trial (ClinicalTrials.gov: NCT03563716 ) when combined with atezolizumab (anti-PD-L1) versus atezolizumab alone1. However, there remains little consensus on the mechanism(s) of response with this combination2. Here we find that a high baseline of intratumoural macrophages and regulatory T cells is associated with better outcomes in patients treated with atezolizumab plus tiragolumab but not with atezolizumab alone. Serum sample analysis revealed that macrophage activation is associated with a clinical benefit in patients who received the combination treatment. In mouse tumour models, tiragolumab surrogate antibodies inflamed tumour-associated macrophages, monocytes and dendritic cells through Fcγ receptors (FcγR), in turn driving anti-tumour CD8+ T cells from an exhausted effector-like state to a more memory-like state. These results reveal a mechanism of action through which TIGIT checkpoint inhibitors can remodel immunosuppressive tumour microenvironments, and suggest that FcγR engagement is an important consideration in anti-TIGIT antibody development.


Assuntos
Anticorpos Monoclonais , Antineoplásicos , Antígeno B7-H1 , Células Mieloides , Neoplasias , Receptores Imunológicos , Linfócitos T Reguladores , Animais , Humanos , Camundongos , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/uso terapêutico , Antineoplásicos/uso terapêutico , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Quimioterapia Combinada , Inibidores de Checkpoint Imunológico/imunologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Ativação de Macrófagos , Células Mieloides/imunologia , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Receptores de IgG/imunologia , Receptores Imunológicos/imunologia , Linfócitos T Reguladores/imunologia , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/imunologia
3.
Cancer Cell ; 40(3): 289-300.e4, 2022 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-35216676

RESUMO

Inhibitors of the programmed cell death-1 (PD-1/PD-L1) signaling axis are approved to treat non-small cell lung cancer (NSCLC) patients, based on their significant overall survival (OS) benefit. Using transcriptomic analysis of 891 NSCLC tumors from patients treated with either the PD-L1 inhibitor atezolizumab or chemotherapy from two large randomized clinical trials, we find a significant B cell association with extended OS with PD-L1 blockade, independent of CD8+ T cell signals. We then derive gene signatures corresponding to the dominant B cell subsets present in NSCLC from single-cell RNA sequencing (RNA-seq) data. Importantly, we find increased plasma cell signatures to be predictive of OS in patients treated with atezolizumab, but not chemotherapy. B and plasma cells are also associated with the presence of tertiary lymphoid structures and organized lymphoid aggregates. Our results suggest an important contribution of B and plasma cells to the efficacy of PD-L1 blockade in NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Antígeno B7-H1/genética , Antígeno B7-H1/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Humanos , Inibidores de Checkpoint Imunológico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Plasmócitos/patologia
4.
Breast Cancer Res Treat ; 172(1): 33-43, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30043096

RESUMO

PURPOSE: Sphingosine kinase 1 (SK1) is a protooncogenic enzyme expressed in many human tumours and is associated with chemoresistance and poor prognosis. It is a potent therapy target and its inhibition chemosensitises solid tumours. Despite recent advances in SK1 inhibitors synthesis and validation, their clinical safety and chemosensitising options are not well described. In this study, we have designed, synthesised and tested a new specific SK1 inhibitor with a low toxicity profile. METHODS: Field template molecular modelling was used for compound design. Lead compounds were tested in cell and mouse cancer models. RESULTS: Field template analysis of three known SK1 inhibitors, SKI-178, 12aa and SK1-I, was performed and compound screening identified six potential new SK1 inhibitors. SK1 activity assays in both cell-free and in vitro settings showed that two compounds were effective SK1 inhibitors. Compound SK-F has potently decreased cancer cell viability in vitro and sensitised mouse breast tumours to docetaxel (DTX) in vivo, without significant whole-body toxicity. CONCLUSION: Through field template screening, we have identified a new SK1 inhibitor, SK-F, which demonstrated antitumour activity in vitro and in vivo without overt toxicity when combined with DTX.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Docetaxel/farmacologia , Inibidores Enzimáticos/química , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Amino Álcoois/química , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sistema Livre de Células/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores Enzimáticos/administração & dosagem , Inibidores Enzimáticos/síntese química , Feminino , Humanos , Hidrazinas/química , Ligantes , Camundongos , Modelos Moleculares , Fosfotransferases (Aceptor do Grupo Álcool)/química , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Pirazóis/química , Ensaios Antitumorais Modelo de Xenoenxerto
5.
J Biol Chem ; 293(5): 1756-1766, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29237731

RESUMO

Acid-sensing ion channels (ASICs) form both homotrimeric and heterotrimeric ion channels that are activated by extracellular protons and are involved in a wide range of physiological and pathophysiological processes, including pain and anxiety. ASIC proteins can form both homotrimeric and heterotrimeric ion channels. The ASIC3 subunit has been shown to be of particular importance in the peripheral nervous system with pharmacological and genetic manipulations demonstrating a role in pain. Naked mole-rats, despite having functional ASICs, are insensitive to acid as a noxious stimulus and show diminished avoidance of acidic fumes, ammonia, and carbon dioxide. Here we cloned naked mole-rat ASIC3 (nmrASIC3) and used a cell-surface biotinylation assay to demonstrate that it traffics to the plasma membrane, but using whole-cell patch clamp electrophysiology we observed that nmrASIC3 is insensitive to both protons and the non-proton ASIC3 agonist 2-guanidine-4-methylquinazoline. However, in line with previous reports of ASIC3 mRNA expression in dorsal root ganglia neurons, we found that the ASIC3 antagonist APETx2 reversibly inhibits ASIC-like currents in naked mole-rat dorsal root ganglia neurons. We further show that like the proton-insensitive ASIC2b and ASIC4, nmrASIC3 forms functional, proton-sensitive heteromers with other ASIC subunits. An amino acid alignment of ASIC3s between 9 relevant rodent species and human identified unique sequence differences that might underlie the proton insensitivity of nmrASIC3. However, introducing nmrASIC3 differences into rat ASIC3 (rASIC3) produced only minor differences in channel function, and replacing the nmrASIC3 sequence with that of rASIC3 did not produce a proton-sensitive ion channel. Our observation that nmrASIC3 forms nonfunctional homomers may reflect a further adaptation of the naked mole-rat to living in an environment with high-carbon dioxide levels.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Membrana Celular/metabolismo , Multimerização Proteica , Subunidades Proteicas/metabolismo , Canais Iônicos Sensíveis a Ácido/genética , Animais , Membrana Celular/genética , Guanidinas/farmacologia , Ratos-Toupeira , Subunidades Proteicas/genética , Quinazolinas/farmacologia
6.
Breast Cancer Res Treat ; 165(3): 531-543, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28695300

RESUMO

PURPOSE: Combining molecular therapies with chemotherapy may offer an improved clinical outcome for chemoresistant tumours. Sphingosine-1-phosphate (S1P) receptor antagonist and sphingosine kinase 1 (SK1) inhibitor FTY720 (FTY) has promising anticancer properties, however, it causes systemic lymphopenia which impairs its use in cancer patients. In this study, we developed a nanoparticle (NP) combining docetaxel (DTX) and FTY for enhanced anticancer effect, targeted tumour delivery and reduced systemic toxicity. METHODS: Docetaxel, FTY and glucosamine were covalently conjugated to poly(lactic-co-glycolic acid) (PLGA). NPs were characterised by dynamic light scattering and electron microscopy. The cellular uptake, cytotoxicity and in vivo antitumor efficacy of CNPs were evaluated. RESULTS: We show for the first time that in triple negative breast cancer cells FTY provides chemosensitisation to DTX, allowing a four-fold reduction in the effective dose. We have encapsulated both drugs in PLGA complex NPs (CNPs), with narrow size distribution of ~ 100 nm and excellent cancer cell uptake providing sequential, sustained release of FTY and DTX. In triple negative breast cancer cells and mouse breast cancer models, CNPs had similar efficacy to systemic free therapies, but allowed an effective drug dose reduction. Application of CNPs has significantly reversed chemotherapy side effects such as weight loss, liver toxicity and, most notably, lymphopenia. CONCLUSIONS: We show for the first time the DTX chemosensitising effects of FTY in triple negative breast cancer. We further demonstrate that encapsulation of free drugs in CNPs can improve targeting, provide low off-target toxicity and most importantly reduce FTY-induced lymphopenia, offering potential therapeutic use of FTY in clinical cancer treatment.


Assuntos
Neoplasias da Mama/complicações , Neoplasias da Mama/patologia , Cloridrato de Fingolimode/administração & dosagem , Linfopenia/induzido quimicamente , Nanopartículas , Taxoides/administração & dosagem , Animais , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Modelos Animais de Doenças , Docetaxel , Feminino , Cloridrato de Fingolimode/efeitos adversos , Humanos , Camundongos , Metástase Neoplásica , Estadiamento de Neoplasias , Carga Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Sci Rep ; 7(1): 5901, 2017 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-28724986

RESUMO

Many prostate cancers relapse after initial chemotherapy treatment. Combining molecular and chemotherapy together with encapsulation of drugs in nanocarriers provides effective drug delivery and toxicity reduction. We developed core shell lipid-polymer hybrid nanoparticles (CSLPHNPs) with poly (lactic-co-glycolic acid) (PLGA) core and lipid layer containing docetaxel and clinically used inhibitor of sphingosine kinase 1 (SK1) FTY720 (fingolimod). We show for the first time that FTY720 (both free and in CSLPHNPs) re-sensitizes castrate resistant prostate cancer cells and tumors to docetaxel, allowing a four-fold reduction in effective dose. Our CSLPHNPs showed high serum stability and a long shelf life. CSLPHNPs demonstrated a steady uptake by tumor cells, sustained intracellular drug release and in vitro efficacy superior to free therapies. In a mouse model of human prostate cancer, CSLPHNPs showed excellent tumor targeting and significantly lower side effects compared to free drugs, importantly, reversing lymphopenia induced by FTY720. Overall, we demonstrate that nanoparticle encapsulation can improve targeting, provide low off-target toxicity and most importantly reduce FTY720-induced lymphopenia, suggesting its potential use in clinical cancer treatment.


Assuntos
Docetaxel/uso terapêutico , Lipídeos/química , Terapia de Alvo Molecular , Nanopartículas/química , Neoplasias da Próstata/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Docetaxel/farmacologia , Liberação Controlada de Fármacos , Humanos , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Nanopartículas/ultraestrutura , Metástase Neoplásica , Tamanho da Partícula , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química
8.
J Cell Biol ; 213(1): 65-79, 2016 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-27069021

RESUMO

Sigma1 receptors (σ1Rs) are expressed widely; they bind diverse ligands, including psychotropic drugs and steroids, regulate many ion channels, and are implicated in cancer and addiction. It is not known how σ1Rs exert such varied effects. We demonstrate that σ1Rs inhibit store-operated Ca(2+)entry (SOCE), a major Ca(2+)influx pathway, and reduce the Ca(2+)content of the intracellular stores. SOCE was inhibited by expression of σ1R or an agonist of σ1R and enhanced by loss of σ1R or an antagonist. Within the endoplasmic reticulum (ER), σ1R associated with STIM1, the ER Ca(2+)sensor that regulates SOCE. This interaction was modulated by σ1R ligands. After depletion of Ca(2+)stores, σ1R accompanied STIM1 to ER-plasma membrane (PM) junctions where STIM1 stimulated opening of the Ca(2+)channel, Orai1. The association of STIM1 with σ1R slowed the recruitment of STIM1 to ER-PM junctions and reduced binding of STIM1 to PM Orai1. We conclude that σ1R attenuates STIM1 coupling to Orai1 and thereby inhibits SOCE.


Assuntos
Canais de Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Receptores sigma/metabolismo , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Retículo Endoplasmático/metabolismo , Células HEK293 , Células HeLa , Humanos , Proteína ORAI1 , Molécula 1 de Interação Estromal , Receptor Sigma-1
9.
Mol Pharmacol ; 87(4): 561-71, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25583083

RESUMO

The acid-sensing ion channels (ASICs) are a family of ion channels expressed throughout the mammalian nervous system. The principal activator of ASICs is extracellular protons, and ASICs have been demonstrated to play a significant role in many physiologic and pathophysiologic processes, including synaptic transmission, nociception, and fear. However, not all ASICs are proton-sensitive: ASIC2a is activated by acid, whereas its splice variant ASIC2b is not. We made a series of chimeric ASIC2 proteins, and using whole-cell electrophysiology we have identified the minimal region of the ASIC2a extracellular domain that is required for ASIC2 proton activation: the first 87 amino acids after transmembrane domain 1. We next examined the function of different domains within the ASIC2b N-terminus and identified a region proximal to the first transmembrane domain that confers tachyphylaxis upon ASIC2a. We have thus identified domains of ASIC2 that are crucial to channel function and may be important for the function of other members of the ASIC family.


Assuntos
Canais Iônicos Sensíveis a Ácido/metabolismo , Canais Iônicos Sensíveis a Ácido/química , Canais Iônicos Sensíveis a Ácido/genética , Animais , Células CHO , Cricetulus , Técnicas de Patch-Clamp , Isoformas de Proteínas , Estrutura Terciária de Proteína , Ratos , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Taquifilaxia
10.
FEBS Lett ; 588(17): 2874-80, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-24996186

RESUMO

Depletion of Ca(2+) from the endoplasmic reticulum (ER) lumen triggers the opening of Ca(2+) release-activated Ca(2+) (CRAC) channels at the plasma membrane. CRAC channels are activated by stromal interaction molecule 1 (STIM1), an ER resident protein that senses Ca(2+) store depletion and interacts with Orai1, the pore-forming subunit of the channel. The subunit stoichiometry of the CRAC channel is controversial. Here we provide evidence, using atomic force microscopy (AFM) imaging, that Orai1 assembles as a hexamer, and that STIM1 binds to Orai1 with sixfold symmetry. STIM1 associates with Orai1 in the form of monomers, dimers, and multimeric string-like structures that form links between the Orai1 hexamers. Our results provide new insights into the nature of the interactions between STIM1 and Orai1.


Assuntos
Canais de Cálcio/química , Canais de Cálcio/metabolismo , Proteínas de Membrana/química , Proteínas de Membrana/metabolismo , Microscopia de Força Atômica , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Multimerização Proteica , Humanos , Proteína ORAI1 , Ligação Proteica , Estrutura Quaternária de Proteína , Molécula 1 de Interação Estromal
11.
Biochem Biophys Res Commun ; 450(4): 1452-7, 2014 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-25017909

RESUMO

Unlike GluN2-containing N-methyl-d-aspartate (NMDA) receptors, which require both glycine and glutamate for activation, receptors composed of GluN1 and GluN3 subunits are activated by glycine alone. Here, we used atomic force microscopy (AFM) imaging to examine the response to activation of the GluN1/GluN3A excitatory glycine receptor. GluN1 and GluN3A subunits were shown to interact intimately within transfected tsA 201 cells. Isolated GluN1/GluN3A receptors integrated into lipid bilayers responded to addition of either glycine or d-serine, but not glutamate, with a ∼1 nm reduction in height of the extracellular domain. The height reduction in response to glycine was abolished by the glycine antagonist 5,7-dichlorokynurenic acid. Our results represent the first demonstration of the effect of activation on the conformation of this receptor.


Assuntos
Isoformas de Proteínas/metabolismo , Receptores de Glutamato/metabolismo , Células HEK293 , Humanos , Conformação Proteica , Isoformas de Proteínas/química , Receptores de Glutamato/química
12.
Adv Cancer Res ; 117: 143-200, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23290780

RESUMO

Sphingosine kinase 1 (SK1) is a lipid enzyme with oncogenic properties that converts the proapoptotic lipids ceramide and sphingosine into the antiapoptotic lipid sphingosine-1-phosphate and activates the signal transduction pathways that lead to cell proliferation, migration, the activation of the inflammatory response, and the impairment of apoptosis. There is compelling evidence that SK1 activation contributes to cancer progression leading to increased oncogenic transformation, tumor growth, resistance to therapies, tumor neovascularization, and metastatic spread. High levels of SK1 expression or activity have been associated with a poor prognosis in several human cancers. Recent studies using cancer cell and mouse models demonstrate a significant potential for SK1-targeting therapies to synergize with the effects of chemotherapy and radiotherapy; however, until recently the absence of clinically applicable SK1 inhibitors has limited the translation of these findings into patients. With the recent discovery of SK1 inhibiting properties of a clinically approved drug FTY720 (Fingolimod), SK1 has gained significant attention from both clinicians and the pharmaceutical industry and it is hoped that trials of newly developed SK1 inhibitors may follow soon. This review provides an overview of the SK1 signaling, its relevance to cancer progression, and the potential clinical significance of targeting SK1 for improved local or systemic control of human cancers.


Assuntos
Antineoplásicos/uso terapêutico , Lisofosfolipídeos/metabolismo , Neoplasias/tratamento farmacológico , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Esfingosina/análogos & derivados , Animais , Humanos , Camundongos , Neoplasias/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Transdução de Sinais , Esfingosina/metabolismo
13.
Pak J Pharm Sci ; 25(1): 27-34, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22186306

RESUMO

In the present study, the effect of DG chloroform root extract was assessed on isolated rat heart and in-vitro antioxidant models. Ischemia reperfusion injury was experimentally induced by using Langendroff apparatus. The free radical scavenging potential was studied in vitro by using different antioxidant models such as DPPH, super oxide scavenging activity, hydroxide scavenging activity and nitric oxide scavenging activity. Both experimental approaches not only substantiate its antioxidant potential but also the cardio-protection imparted by the extract. The cardio-stimulatory effects were investigated for the extract by treating it as a pre-conditioning agent against myocardial ischemia reperfusion injury. The improved antioxidant status of the myocardium indirectly predicts reduced oxidative stress mediated by ischemic reperfusion with evident reduction of infarct size determined by cardiac marker protein. These findings indicate that DG chloroform root extract may possess therapeutic potential against ischemia reperfusion injury.


Assuntos
Clorofórmio/química , Sequestradores de Radicais Livres/farmacologia , Cromatografia Gasosa-Espectrometria de Massas/métodos , Traumatismo por Reperfusão Miocárdica/tratamento farmacológico , Extratos Vegetais/farmacologia , Raízes de Plantas/química , Animais , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Fabaceae , Sequestradores de Radicais Livres/química , Humanos , Técnicas In Vitro , Masculino , Traumatismo por Reperfusão Miocárdica/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/química , Ratos , Ratos Wistar
14.
Nanotechnology ; 22(26): 265101, 2011 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-21576779

RESUMO

The use of cisplatin, a first line chemotherapy for most cancers, is dose-limited due to nephrotoxicity. While this toxicity can be addressed through nanotechnology, previous attempts at engineering cisplatin nanoparticles have been limited by the impact on the potency of cisplatin. Here we report the rational engineering of a novel cisplatin nanoparticle by harnessing a novel polyethylene glycol-functionalized poly-isobutylene-maleic acid (PEG-PIMA) copolymer, which can complex with cis-platinum (II) through a monocarboxylato and a coordinate bond. We show that this complex self-assembles into a nanoparticle, and exhibits an IC(50) = 0.77 ± 0.11 µM comparable to that of free cisplatin (IC(50) = 0.44 ± 0.09 µM). The nanoparticles are internalized into the endolysosomal compartment of cancer cells, and release cisplatin in a pH-dependent manner. Furthermore, the nanoparticles exhibit significantly improved antitumor efficacy in a 4T1 breast cancer model in vivo, with limited nephrotoxicity, which can be explained by preferential biodistribution in the tumor with reduced kidney concentrations. Our results suggest that the PEG-PIMA-cisplatin nanoparticle can emerge as an attractive solution to the challenges in cisplatin chemotherapy.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Nanopartículas/uso terapêutico , Nanotecnologia/métodos , Neoplasias/tratamento farmacológico , Polietilenoglicóis/química , Polímeros/química , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Cisplatino/química , Cisplatino/farmacologia , Citometria de Fluxo , Humanos , Rim/efeitos dos fármacos , Rim/patologia , Camundongos , Nanopartículas/ultraestrutura , Tamanho do Órgão/efeitos dos fármacos , Baço/efeitos dos fármacos , Baço/patologia , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA