Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2023 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-37546807

RESUMO

The presence of cell surface protein CD47 allows cancer cells to evade innate and adaptive immune surveillance resulting in metastatic spread. CD47 binds to and activates SIRPα on the surface of myeloid cells, inhibiting their phagocytic activity. On the other hand, CD47 binds the matricellular protein Thrombospondin-1, limiting T-cell activation. Thus, blocking CD47 is a potential therapeutic strategy for preventing brain metastasis. To test this hypothesis, breast cancer patient biopsies were stained with antibodies against CD47 to determine differences in protein expression. An anti-CD47 antibody was used in a syngeneic orthotopic triple-negative breast cancer model, and CD47 null mice were used in a breast cancer brain metastasis model by intracardiac injection of the E0771-Br-Luc cell line. Immunohistochemical staining of patient biopsies revealed an 89% increase in CD47 expression in metastatic brain tumors compared to normal adjacent tissue (p ≤ 0.05). Anti-CD47 treatment in mice bearing brain metastatic 4T1br3 orthotopic tumors reduced tumor volume and tumor weight by over 50% compared to control mice (p ≤ 0.05) and increased IBA1 macrophage/microglia marker 5-fold in tumors compared to control (p ≤ 0.05). Additionally, CD47 blockade increased the M1/M2 macrophage ratio in tumors 2.5-fold (p ≤ 0.05). CD47 null mice had an 89% decrease in metastatic brain burden (p ≤ 0.05) compared to control mice in a brain metastasis model. Additionally, RNA sequencing revealed several uniquely expressed genes and significantly enriched genes related to tissue development, cell death, and cell migration tumors treated with anti-CD47 antibodies. Thus, demonstrating that CD47 blockade affects cancer cell and tumor microenvironment signaling to limit metastatic spread and may be an effective therapeutic for triple-negative breast cancer brain metastasis.

2.
Cancers (Basel) ; 15(10)2023 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-37345039

RESUMO

The purpose of this study is to further validate the utility of our previously developed CNN in an alternative small animal model of BM through transfer learning. Unlike the glioma model, the BM mouse model develops multifocal intracranial metastases, including both contrast enhancing and non-enhancing lesions on DCE MRI, thus serving as an excellent brain tumor model to study tumor vascular permeability. Here, we conducted transfer learning by transferring the previously trained GBM CNN to DCE MRI datasets of BM mice. The CNN was re-trained to learn about the relationship between BM DCE images and target permeability maps extracted from the Extended Tofts Model (ETM). The transferred network was found to accurately predict BM permeability and presented with excellent spatial correlation with the target ETM PK maps. The CNN model was further tested in another cohort of BM mice treated with WBRT to assess vascular permeability changes induced via radiotherapy. The CNN detected significantly increased permeability parameter Ktrans in WBRT-treated tumors (p < 0.01), which was in good agreement with the target ETM PK maps. In conclusion, the proposed CNN can serve as an efficient and accurate tool for characterizing vascular permeability and treatment responses in small animal brain tumor models.

3.
Cancers (Basel) ; 15(9)2023 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-37174093

RESUMO

The brain is one of the most common metastatic sites among breast cancer patients, especially in those who have Her2-positive or triple-negative tumors. The brain microenvironment has been considered immune privileged, and the exact mechanisms of how immune cells in the brain microenvironment contribute to brain metastasis remain elusive. In this study, we found that neutrophils are recruited and influenced by c-Met high brain metastatic cells in the metastatic sites, and depletion of neutrophils significantly suppressed brain metastasis in animal models. Overexpression of c-Met in tumor cells enhances the secretion of a group of cytokines, including CXCL1/2, G-CSF, and GM-CSF, which play critical roles in neutrophil attraction, granulopoiesis, and homeostasis. Meanwhile, our transcriptomic analysis demonstrated that conditioned media from c-Met high cells significantly induced the secretion of lipocalin 2 (LCN2) from neutrophils, which in turn promotes the self-renewal of cancer stem cells. Our study unveiled the molecular and pathogenic mechanisms of how crosstalk between innate immune cells and tumor cells facilitates tumor progression in the brain, which provides novel therapeutic targets for treating brain metastasis.

4.
Nano Res ; 16(4): 5300-5310, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37228440

RESUMO

Despite therapeutic advancements, the prognosis of locally advanced non-small cell lung cancer (LANSCLC), which has invaded multiple lobes or the other lung and intrapulmonary lymph nodes, remains poor. The emergence of immunotherapy with immune checkpoint blockade (ICB) is transforming cancer treatment. However, only a fraction of lung cancer patients benefit from ICB. Significant clinical evidence suggests that the proinflammatory tumor microenvironment (TME) and programmed death-ligand 1 (PD-L1) expression correlate positively with response to the PD-1/PD-L1 blockade. We report here a liposomal nanoparticle loaded with cyclic dinucleotide and aerosolized (AeroNP-CDN) for inhalation delivery to deep-seated lung tumors and target CDN to activate stimulators of interferon (IFN) genes in macrophages and dendritic cells (DCs). Using a mouse model that recapitulates the clinical LANSCLC, we show that AeroNP-CDN efficiently mitigates the immunosuppressive TME by reprogramming tumor-associated macrophage from the M2 to M1 phenotype, activating DCs for effective tumor antigen presentation and increasing tumor-infiltrating CD8+ T cells for adaptive anticancer immunity. Intriguingly, activation of interferons by AeroNP-CDN also led to increased PD-L1 expression in lung tumors, which, however, set a stage for response to anti-PD-L1 treatment. Indeed, anti-PD-L1 antibody-mediated blockade of IFNs-induced immune inhibitory PD-1/PD-L1 signaling further prolonged the survival of the LANSCLC-bearing mice. Importantly, AeroNP-CDN alone or combination immunotherapy was safe without local or systemic immunotoxicity. In conclusion, this study demonstrates a potential nano-immunotherapy strategy for LANSCLC, and mechanistic insights into the evolution of adaptive immune resistance provide a rational combination immunotherapy to overcome it.

5.
Nat Cell Biol ; 24(8): 1291-1305, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35915159

RESUMO

The epidermal growth factor receptor (EGFR) is a prime oncogene that is frequently amplified in glioblastomas. Here we demonstrate a new tumour-suppressive function of EGFR in EGFR-amplified glioblastomas regulated by EGFR ligands. Constitutive EGFR signalling promotes invasion via activation of a TAB1-TAK1-NF-κB-EMP1 pathway, resulting in large tumours and decreased survival in orthotopic models. Ligand-activated EGFR promotes proliferation and surprisingly suppresses invasion by upregulating BIN3, which inhibits a DOCK7-regulated Rho GTPase pathway, resulting in small hyperproliferating non-invasive tumours and improved survival. Data from The Cancer Genome Atlas reveal that in EGFR-amplified glioblastomas, a low level of EGFR ligands confers a worse prognosis, whereas a high level of EGFR ligands confers an improved prognosis. Thus, increased EGFR ligand levels shift the role of EGFR from oncogene to tumour suppressor in EGFR-amplified glioblastomas by suppressing invasion. The tumour-suppressive function of EGFR can be activated therapeutically using tofacitinib, which suppresses invasion by increasing EGFR ligand levels and upregulating BIN3.


Assuntos
Glioblastoma , Proteínas dos Microfilamentos/metabolismo , Linhagem Celular Tumoral , Receptores ErbB/genética , Receptores ErbB/metabolismo , Glioblastoma/metabolismo , Humanos , Ligantes , Oncogenes/genética , Regulação para Cima
6.
Clin Transl Med ; 12(3): e774, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35297203

RESUMO

Clinical evidence indicates that the microenvironment in malignant pleural effusion (MPE) is immunologically cold, which impairs tumour immunosurveillance and antitumor immune response to immune checkpoint blockade (ICB). In a recent issue of Nature Nanotechnology, Liu et al. demonstrate a new nanotechnological approach to effectively mitigate the immune cold MPE and provide insights into its mechanism of action through single-cell RNA-sequencing.


Assuntos
Neoplasias Pulmonares , Derrame Pleural Maligno , Humanos , Inibidores de Checkpoint Imunológico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Derrame Pleural Maligno/tratamento farmacológico , Derrame Pleural Maligno/genética , RNA/farmacologia , RNA/uso terapêutico , Microambiente Tumoral
7.
Front Biosci (Landmark Ed) ; 27(3): 99, 2022 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-35345331

RESUMO

BACKGROUND: Dynamic contrast-enhanced (DCE) MRI is widely used to assess vascular perfusion and permeability in cancer. In small animal applications, conventional modeling of pharmacokinetic (PK) parameters from DCE MRI images is complex and time consuming. This study is aimed at developing a deep learning approach to fully automate the generation of kinetic parameter maps, Ktrans (volume transfer coefficient) and Vp (blood plasma volume ratio), as a potential surrogate to conventional PK modeling in mouse brain tumor models based on DCE MRI. METHODS: Using a 7T MRI, DCE MRI was conducted in U87 glioma xenografts growing orthotopically in nude mice. Vascular permeability Ktrans and Vp maps were generated using the classical Tofts model as well as the extended-Tofts model. These vascular permeability maps were then processed as target images to a twenty-four layer convolutional neural network (CNN). The CNN was trained on T1-weighted DCE images as source images and designed with parallel dual pathways to capture multiscale features. Furthermore, we performed a transfer study of this glioma trained CNN on a breast cancer brain metastasis (BCBM) mouse model to assess the potential of the network for alternative brain tumors. RESULTS: Our data showed a good match for both Ktrans and Vp maps generated between the target PK parameter maps and the respective CNN maps for gliomas. Pixel-by-pixel analysis revealed intratumoral heterogeneous permeability, which was consistent between the CNN and PK models. The utility of the deep learning approach was further demonstrated in the transfer study of BCBM. CONCLUSIONS: Because of its rapid and accurate estimation of vascular PK parameters directly from the DCE dynamic images without complex mathematical modeling, the deep learning approach can serve as an efficient tool to assess tumor vascular permeability to facilitate small animal brain tumor research.


Assuntos
Neoplasias Encefálicas , Aprendizado Profundo , Animais , Neoplasias Encefálicas/patologia , Meios de Contraste , Humanos , Imageamento por Ressonância Magnética/métodos , Camundongos , Camundongos Nus
8.
Nat Nanotechnol ; 17(2): 206-216, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34916656

RESUMO

Malignant pleural effusion (MPE) is indicative of terminal malignancy with a uniformly fatal prognosis. Often, two distinct compartments of tumour microenvironment, the effusion and disseminated pleural tumours, co-exist in the pleural cavity, presenting a major challenge for therapeutic interventions and drug delivery. Clinical evidence suggests that MPE comprises abundant tumour-associated myeloid cells with the tumour-promoting phenotype, impairing antitumour immunity. Here we developed a liposomal nanoparticle loaded with cyclic dinucleotide (LNP-CDN) for targeted activation of stimulators of interferon genes signalling in macrophages and dendritic cells and showed that, on intrapleural administration, they induce drastic changes in the transcriptional landscape in MPE, mitigating the immune cold MPE in both effusion and pleural tumours. Moreover, combination immunotherapy with blockade of programmed death ligand 1 potently reduced MPE volume and inhibited tumour growth not only in the pleural cavity but also in the lung parenchyma, conferring significantly prolonged survival of MPE-bearing mice. Furthermore, the LNP-CDN-induced immunological effects were also observed with clinical MPE samples, suggesting the potential of intrapleural LNP-CDN for clinical MPE immunotherapy.


Assuntos
Antígeno B7-H1/farmacologia , Sistemas de Liberação de Medicamentos , Nanopartículas/química , Derrame Pleural Maligno/tratamento farmacológico , Imunidade Adaptativa/efeitos dos fármacos , Animais , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/química , Antígeno B7-H1/imunologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Dendríticas/efeitos dos fármacos , Humanos , Inibidores de Checkpoint Imunológico/química , Inibidores de Checkpoint Imunológico/farmacologia , Imunidade Inata/efeitos dos fármacos , Imunoterapia , Interferons/genética , Camundongos , Nanopartículas/uso terapêutico , Cavidade Pleural/efeitos dos fármacos , Cavidade Pleural/imunologia , Cavidade Pleural/patologia , Derrame Pleural Maligno/genética , Derrame Pleural Maligno/imunologia , Derrame Pleural Maligno/patologia , Microambiente Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Nat Commun ; 12(1): 7014, 2021 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-34853306

RESUMO

Inhibition of RTK pathways in cancer triggers an adaptive response that promotes therapeutic resistance. Because the adaptive response is multifaceted, the optimal approach to blunting it remains undetermined. TNF upregulation is a biologically significant response to EGFR inhibition in NSCLC. Here, we compared a specific TNF inhibitor (etanercept) to thalidomide and prednisone, two drugs that block TNF and also other inflammatory pathways. Prednisone is significantly more effective in suppressing EGFR inhibition-induced inflammatory signals. Remarkably, prednisone induces a shutdown of bypass RTK signaling and inhibits key resistance signals such as STAT3, YAP and TNF-NF-κB. Combined with EGFR inhibition, prednisone is significantly superior to etanercept or thalidomide in durably suppressing tumor growth in multiple mouse models, indicating that a broad suppression of adaptive signals is more effective than blocking a single component. We identify prednisone as a drug that can effectively inhibit adaptive resistance with acceptable toxicity in NSCLC and other cancers.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glucocorticoides/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células A549 , Animais , Carcinoma Pulmonar de Células não Pequenas , Citocinas/metabolismo , Modelos Animais de Doenças , Receptores ErbB/efeitos dos fármacos , Feminino , Humanos , Neoplasias Pulmonares , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Prednisona , Fator de Transcrição STAT3/metabolismo , Talidomida , Inibidores do Fator de Necrose Tumoral , Regulação para Cima
10.
Nat Cancer ; 1(4): 394-409, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-33269343

RESUMO

EGFR inhibition is an effective treatment in the minority of non-small cell lung cancer (NSCLC) cases harboring EGFR-activating mutations, but not in EGFR wild type (EGFRwt) tumors. Here, we demonstrate that EGFR inhibition triggers an antiviral defense pathway in NSCLC. Inhibiting mutant EGFR triggers Type I IFN-I upregulation via a RIG-I-TBK1-IRF3 pathway. The ubiquitin ligase TRIM32 associates with TBK1 upon EGFR inhibition, and is required for K63-linked ubiquitination and TBK1 activation. Inhibiting EGFRwt upregulates interferons via an NF-κB-dependent pathway. Inhibition of IFN signaling enhances EGFR-TKI sensitivity in EGFR mutant NSCLC and renders EGFRwt/KRAS mutant NSCLC sensitive to EGFR inhibition in xenograft and immunocompetent mouse models. Furthermore, NSCLC tumors with decreased IFN-I expression are more responsive to EGFR TKI treatment. We propose that IFN-I signaling is a major determinant of EGFR-TKI sensitivity in NSCLC and that a combination of EGFR TKI plus IFN-neutralizing antibody could be useful in most NSCLC patients.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Receptores ErbB , Neoplasias Pulmonares , Transdução de Sinais , Animais , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Proliferação de Células , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Inibidores de Proteínas Quinases/farmacologia
11.
Nat Commun ; 10(1): 5108, 2019 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-31704921

RESUMO

Mounting evidence suggests that the tumor microenvironment is profoundly immunosuppressive. Thus, mitigating tumor immunosuppression is crucial for inducing sustained antitumor immunity. Whereas previous studies involved intratumoral injection, we report here an inhalable nanoparticle-immunotherapy system targeting pulmonary antigen presenting cells (APCs) to enhance anticancer immunity against lung metastases. Inhalation of phosphatidylserine coated liposome loaded with STING agonist cyclic guanosine monophosphate-adenosine monophosphate (NP-cGAMP) in mouse models of lung metastases enables rapid distribution of NP-cGAMP to both lungs and subsequent uptake by APCs without causing immunopathology. NP-cGAMP designed for enhanced cytosolic release of cGAMP stimulates STING signaling and type I interferons production in APCs, resulting in the pro-inflammatory tumor microenvironment in multifocal lung metastases. Furthermore, fractionated radiation delivered to one tumor-bearing lung synergizes with inhaled NP-cGAMP, eliciting systemic anticancer immunity, controlling metastases in both lungs, and conferring long-term survival in mice with lung metastases and with repeated tumor challenge.


Assuntos
Células Apresentadoras de Antígenos/efeitos dos fármacos , Imunoterapia , Neoplasias Pulmonares/secundário , Pulmão/efeitos dos fármacos , Melanoma Experimental/secundário , Proteínas de Membrana/agonistas , Nanopartículas , Nucleotídeos Cíclicos/farmacologia , Radioterapia , Administração por Inalação , Animais , Células Apresentadoras de Antígenos/imunologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Interferon Tipo I/efeitos dos fármacos , Interferon Tipo I/imunologia , Lipossomos , Pulmão/imunologia , Pulmão/efeitos da radiação , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Camundongos , Nucleotídeos Cíclicos/administração & dosagem , Fosfatidilserinas
12.
Neuro Oncol ; 21(12): 1529-1539, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31363754

RESUMO

BACKGROUND: Glioblastoma (GBM) is the most common primary malignant adult brain tumor. Temozolomide (TMZ) is the standard of care and is most effective in GBMs that lack the DNA repair protein O6-methylguanine-DNA methyltransferase (MGMT). Moreover, even initially responsive tumors develop a secondary resistance to TMZ and become untreatable. Since aberrant epidermal growth factor receptor (EGFR) signaling is widespread in GBM, EGFR inhibition has been tried in multiple clinical trials without success. We recently reported that inhibiting EGFR leads to increased secretion of tumor necrosis factor (TNF) and activation of a survival pathway in GBM. Here, we compare the efficacy of TMZ versus EGFR plus TNF inhibition in an orthotopic mouse model of GBM. METHODS: We use an orthotopic model to examine the efficacy of TMZ versus EGFR plus TNF inhibition in multiple subsets of GBMs, including MGMT methylated and unmethylated primary GBMs, recurrent GBMs, and GBMs rendered experimentally resistant to TMZ. RESULTS: The efficacy of the 2 treatments was similar in MGMT methylated GBMs. However, in MGMT unmethylated GBMs, a combination of EGFR plus TNF inhibition was more effective. We demonstrate that the 2 treatment approaches target distinct and non-overlapping pathways. Thus, importantly, EGFR plus TNF inhibition remains effective in TMZ-resistant recurrent GBMs and in GBMs rendered experimentally resistant to TMZ. CONCLUSION: EGFR inhibition combined with a blunting of the accompanying TNF-driven adaptive response could be a viable therapeutic approach in MGMT unmethylated and recurrent EGFR-expressing GBMs.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Afatinib/administração & dosagem , Animais , Apoptose , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Proliferação de Células , Receptores ErbB/antagonistas & inibidores , Feminino , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Temozolomida/administração & dosagem , Talidomida/administração & dosagem , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Int J Radiat Biol ; 95(3): 338-346, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30499763

RESUMO

PURPOSE: To assess early changes in brain metastasis in response to whole brain radiotherapy (WBRT) by longitudinal Magnetic Resonance Imaging (MRI). MATERIALS AND METHODS: Using a 7T system, MRI examinations of brain metastases in a breast cancer MDA-MD231-Br mouse model were conducted before and 24 hours after 3 daily fractionations of 4 Gy WBRT. Besides anatomic MRI, diffusion-weighted (DW) MRI and dynamic contrast-enhanced (DCE) MRI were applied to study cytotoxic effect and blood-tumor-barrier (BTB) permeability change, respectively. RESULTS: Before treatment, high-resolution T2-weighted images revealed hyperintense multifocal lesions, many of which (∼50%) were not enhanced on T1-weighted contrast images, indicating intact BTB in the brain metastases. While no difference in the number of new lesions was observed, WBRT-treated tumors were significantly smaller than sham controls (p < .05). DW MRI detected significant increase in apparent diffusion coefficient (ADC) in WBRT tumors (p < .05), which correlated with elevated caspase 3 staining of apoptotic cells. Many lesions remained non-enhanced post WBRT. However, quantitative DCE MRI analysis showed significantly higher permeability parameter, Ktrans, in WBRT than the sham group (p < .05), despite marked spatial heterogeneity. CONCLUSIONS: MRI allowed non-invasive assessments of WBRT induced changes in BTB permeability, which may provide useful information for potential combination treatment.


Assuntos
Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/secundário , Neoplasias da Mama/patologia , Imageamento por Ressonância Magnética , Animais , Neoplasias Encefálicas/diagnóstico por imagem , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Methods Mol Biol ; 1790: 197-208, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29858793

RESUMO

Cerenkov luminescence imaging (CLI) is a relatively new imaging modality that utilizes conventional optical imaging instrumentation to detect Cerenkov radiation derived from standard and often clinically approved radiotracers. Its research versatility, low cost, and ease of use have increased its popularity within the molecular imaging community and at institutions that are interested in conducting radiotracer-based molecular imaging research, but that lack the necessary resources and infrastructure. Here, we provide a description of the materials and procedures necessary to conduct a Cerenkov luminescence imaging experiment using a variety of imaging instrumentation, radionuclides, and animal models.


Assuntos
Medições Luminescentes/métodos , Imagem Multimodal/métodos , Neoplasias/patologia , Imagens de Fantasmas , Compostos Radiofarmacêuticos/metabolismo , Animais , Humanos , Camundongos , Neoplasias/diagnóstico por imagem , Neoplasias/metabolismo , Guias de Prática Clínica como Assunto
15.
J Clin Invest ; 128(6): 2500-2518, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29613856

RESUMO

Although aberrant EGFR signaling is widespread in cancer, EGFR inhibition is effective only in a subset of non-small cell lung cancer (NSCLC) with EGFR activating mutations. A majority of NSCLCs express EGFR wild type (EGFRwt) and do not respond to EGFR inhibition. TNF is a major mediator of inflammation-induced cancer. We find that a rapid increase in TNF level is a universal adaptive response to EGFR inhibition in NSCLC, regardless of EGFR status. EGFR signaling actively suppresses TNF mRNA levels by inducing expression of miR-21, resulting in decreased TNF mRNA stability. Conversely, EGFR inhibition results in loss of miR-21 and increased TNF mRNA stability. In addition, TNF-induced NF-κB activation leads to increased TNF transcription in a feed-forward loop. Inhibition of TNF signaling renders EGFRwt-expressing NSCLC cell lines and an EGFRwt patient-derived xenograft (PDX) model highly sensitive to EGFR inhibition. In EGFR-mutant oncogene-addicted cells, blocking TNF enhances the effectiveness of EGFR inhibition. EGFR plus TNF inhibition is also effective in NSCLC with acquired resistance to EGFR inhibition. We suggest concomitant EGFR and TNF inhibition as a potentially new treatment approach that could be beneficial for a majority of lung cancer patients.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Neoplasias Pulmonares/metabolismo , Proteínas de Neoplasias , Neoplasias Experimentais/metabolismo , Fator de Necrose Tumoral alfa , Células A549 , Animais , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/terapia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Camundongos , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentais/genética , Neoplasias Experimentais/patologia , Neoplasias Experimentais/terapia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
16.
Mol Imaging ; 16: 1536012117708722, 2017 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-28654387

RESUMO

Phosphatidylserine (PS), the most abundant anionic phospholipid in cell membrane, is strictly confined to the inner leaflet in normal cells. However, this PS asymmetry is found disruptive in many tumor vascular endothelial cells. We discuss the underlying mechanisms for PS asymmetry maintenance in normal cells and its loss in tumor cells. The specificity of PS exposure in tumor vasculature but not normal blood vessels may establish it a useful biomarker for cancer molecular imaging. Indeed, utilizing PS-targeting antibodies, multiple imaging probes have been developed and multimodal imaging data have shown their high tumor-selective targeting in various cancers. There is a critical need for improved diagnosis and therapy for brain tumors. We have recently established PS-targeted nanoplatforms, aiming to enhance delivery of imaging contrast agents across the blood-brain barrier to facilitate imaging of brain tumors. Advantages of using the nanodelivery system, in particular, lipid-based nanocarriers, are discussed here. We also describe our recent research interest in developing PS-targeted nanotheranostics for potential image-guided drug delivery to treat brain tumors.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Imagem Molecular/métodos , Fosfatidilserinas/análise , Animais , Barreira Hematoencefálica/diagnóstico por imagem , Linhagem Celular Tumoral , Humanos
17.
Nat Neurosci ; 20(8): 1074-1084, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28604685

RESUMO

Aberrant epidermal growth factor receptor (EGFR) signaling is widespread in cancer, making the EGFR an important target for therapy. EGFR gene amplification and mutation are common in glioblastoma (GBM), but EGFR inhibition has not been effective in treating this tumor. Here we propose that primary resistance to EGFR inhibition in glioma cells results from a rapid compensatory response to EGFR inhibition that mediates cell survival. We show that in glioma cells expressing either EGFR wild type or the mutant EGFRvIII, EGFR inhibition triggers a rapid adaptive response driven by increased tumor necrosis factor (TNF) secretion, which leads to activation in turn of c-Jun N-terminal kinase (JNK), the Axl receptor tyrosine kinase and extracellular signal-regulated kinases (ERK). Inhibition of this adaptive axis at multiple nodes rendered glioma cells with primary resistance sensitive to EGFR inhibition. Our findings provide a possible explanation for the failures of anti-EGFR therapy in GBM and suggest a new approach to the treatment of EGFR-expressing GBM using a combination of EGFR and TNF inhibition.


Assuntos
Neoplasias Encefálicas/metabolismo , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glioblastoma/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Transdução de Sinais , Antineoplásicos/farmacologia , Neoplasias Encefálicas/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , MAP Quinases Reguladas por Sinal Extracelular/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Humanos , Transdução de Sinais/efeitos dos fármacos
18.
Transl Cancer Res ; 6(Suppl 3): S512-S514, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-31531290
19.
Oncotarget ; 7(25): 38693-38706, 2016 Jun 21.
Artigo em Inglês | MEDLINE | ID: mdl-27231848

RESUMO

Phosphatidylserine (PS), which is normally intracellular, becomes exposed on the outer surface of viable endothelial cells (ECs) of tumor vasculature. Utilizing a PS-targeting antibody, we have recently established a PS-targeted liposomal (PS-L) nanoplatform that has demonstrated to be highly tumor-selective. Because of the vascular lumen-exposed PS that is immediately accessible without a need to penetrate the intact blood brain barrier (BBB), we hypothesize that the systemically administered PS-L binds specifically to tumor vascular ECs, becomes subsequently internalized into the cells and then enables its cargos to be efficiently delivered to glioma parenchyma. To test this, we exploited the dual MRI/optical imaging contrast agents-loaded PS-L and injected it intravenously into mice bearing intracranial U87 glioma. At 24 h, both in vivo optical imaging and MRI depicted enhanced tumor contrast, distinct from the surrounding normal brain. Intriguingly, longitudinal MRI revealed temporal and spatial intratumoral distribution of the PS-L by following MRI contrast changes, which appeared punctate in tumor periphery at an earlier time point (4 h), but became clustering and disseminated throughout the tumor at 24 h post injection. Importantly, glioma-targeting specificity of the PS-L was antigen specific, since a control probe of irrelevant specificity showed minimal accumulation in the glioma. Together, these results indicate that the PS-L nanoplatform enables the enhanced, glioma-targeted delivery of imaging contrast agents by crossing the tumor BBB efficiently, which may also serve as a useful nanoplatform for anti-glioma drugs.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Glioma/diagnóstico por imagem , Lipossomos/química , Fosfatidilserinas/metabolismo , Animais , Aorta/metabolismo , Barreira Hematoencefálica , Neoplasias Encefálicas/irrigação sanguínea , Bovinos , Linhagem Celular Tumoral , Meios de Contraste/química , Células Endoteliais/metabolismo , Compostos Férricos/química , Glioma/irrigação sanguínea , Humanos , Imageamento por Ressonância Magnética , Camundongos , Camundongos Nus , Nanopartículas/química , Transplante de Neoplasias
20.
Cancer Lett ; 378(2): 97-103, 2016 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-27224892

RESUMO

OBJECTIVE: To quantitatively study the impact of intrinsic tumor characteristics and microenvironmental factors on local tumor control after irradiation with carbon ((12)C-) ions and photons in an experimental prostate tumor model. MATERIAL AND METHODS: Three sublines of a syngeneic rat prostate tumor (R3327) differing in grading (highly (-H) moderately (-HI) or anaplastic (-AT1)) were irradiated with increasing single doses of either (12)C-ions or 6 MV photons in Copenhagen rats. Primary endpoint was local tumor control within 300 days. The relative biological effectiveness (RBE) of (12)C-ions was calculated from the dose at 50% tumor control probability (TCD50) of photons and (12)C-ions and was correlated with histological, physiological and genetic tumor parameters. RESULTS: Experimental findings demonstrated that (i) TCD50-values between the three tumor sublines differed less for (12)C-ions (23.6-32.9 Gy) than for photons (38.2-75.7 Gy), (ii) the slope of the dose-response curve for each tumor line was steeper for (12)C-ions than for photons, and (iii) the RBE increased with tumor grading from 1.62 ± 0.11 (H) to 2.08 ± 0.13 (HI) to 2.30 ± 0.08 (AT1). CONCLUSION: The response to (12)C-ions is less dependent on resistance factors as well as on heterogeneity between and within tumor sublines as compared to photons. A clear correlation between decreasing differentiation status and increasing RBE was found. (12)C-ions may therefore be a therapeutic option especially in patients with undifferentiated prostate tumors, expressing high resistance against photons.


Assuntos
Adenocarcinoma/radioterapia , Radioterapia com Íons Pesados , Neoplasias Experimentais/radioterapia , Neoplasias Pancreáticas/radioterapia , Fótons , Doses de Radiação , Tolerância a Radiação , Adenocarcinoma/patologia , Animais , Diferenciação Celular , Linhagem Celular Tumoral , Relação Dose-Resposta à Radiação , Masculino , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Neoplasias Pancreáticas/patologia , Ratos , Microambiente Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...