Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Comput Biol ; 19(4): e1010995, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37068117

RESUMO

Our understanding of how speed and persistence of cell migration affects the growth rate and size of tumors remains incomplete. To address this, we developed a mathematical model wherein cells migrate in two-dimensional space, divide, die or intravasate into the vasculature. Exploring a wide range of speed and persistence combinations, we find that tumor growth positively correlates with increasing speed and higher persistence. As a biologically relevant example, we focused on Golgi fragmentation, a phenomenon often linked to alterations of cell migration. Golgi fragmentation was induced by depletion of Giantin, a Golgi matrix protein, the downregulation of which correlates with poor patient survival. Applying the experimentally obtained migration and invasion traits of Giantin depleted breast cancer cells to our mathematical model, we predict that loss of Giantin increases the number of intravasating cells. This prediction was validated, by showing that circulating tumor cells express significantly less Giantin than primary tumor cells. Altogether, our computational model identifies cell migration traits that regulate tumor progression and uncovers a role of Giantin in breast cancer progression.


Assuntos
Neoplasias da Mama , Proteínas de Membrana , Humanos , Feminino , Proteínas de Membrana/metabolismo , Proteínas da Matriz do Complexo de Golgi/metabolismo , Neoplasias da Mama/metabolismo , Complexo de Golgi/metabolismo , Complexo de Golgi/patologia
2.
Cancer Immunol Immunother ; 64(6): 769-76, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25832001

RESUMO

Malignant melanoma is highly aggressive cancer with poor prognosis and few therapeutic options. Interferon alpha (IFN-α) has been tested as adjuvant immunotherapy in high-risk melanoma patients in a number of studies, but its beneficial role is controversial. Although IFN-α treatment can prolong relapse-free survival, the effect on overall survival is not significant. However, a small subset of patients benefits from the treatment, signifying the need for biomarkers able to identify a responding subgroup. Here we evaluated whether serum osteopontin (OPN) could function as a biomarker identifying patients with poor prognosis that might benefit from IFN-α. The choice of osteopontin was based on the knowledge about the dual role of this protein in cancer and immune response, an apparent association between OPN and IFN signaling and a prognostic value of OPN in multiple other tumor types. Serum samples from 275 high-risk melanoma patients enrolled in the Nordic Adjuvant IFN Melanoma trial were analyzed for circulating OPN concentrations and OPN promoter polymorphisms in position -443. The potential relation between serum OPN levels, the genotypes and survival in non-treated patients and patients receiving adjuvant IFN-α was investigated. Although slightly better survival was observed in the treated patients that had high levels of OPN, the difference was not statistically significant. In conclusion, serum OPN (its level or the genotype) cannot distinguish melanoma patients with poor prognosis, or patients that might benefit from adjuvant treatment with IFN-α.


Assuntos
Melanoma/sangue , Melanoma/genética , Osteopontina/sangue , Osteopontina/genética , Neoplasias Cutâneas/sangue , Neoplasias Cutâneas/genética , Biomarcadores Tumorais/sangue , Biomarcadores Tumorais/genética , Feminino , Humanos , Interferon-alfa/administração & dosagem , Masculino , Melanoma/tratamento farmacológico , Melanoma/patologia , Polimorfismo de Nucleotídeo Único , Prognóstico , Ensaios Clínicos Controlados Aleatórios como Assunto , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/patologia
3.
Int J Nanomedicine ; 19: 3009-3029, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38562610

RESUMO

Background: Biodegradable poly(alkyl cyanoacrylate) (PACA) nanoparticles (NPs) are receiving increasing attention in anti-cancer nanomedicine development not only for targeted cancer chemotherapy, but also for modulation of the tumor microenvironment. We previously reported promising results with cabazitaxel (CBZ) loaded poly(2-ethylbutyl cyanoacrylate) NPs (PEBCA-CBZ NPs) in a patient derived xenograft (PDX) model of triple-negative breast cancer, and this was associated with a decrease in M2 macrophages. The present study aims at comparing two endotoxin-free PACA NP variants (PEBCA and poly(2-ethylhexyl cyanoacrylate); PEHCA), loaded with CBZ and test whether conjugation with folate would improve their effect. Methods: Cytotoxicity assays and cellular uptake of NPs by flow cytometry were performed in different breast cancer cells. Biodistribution and efficacy studies were performed in PDX models of breast cancer. Tumor associated immune cells were analyzed by multiparametric flow cytometry. Results: In vitro studies showed similar NP-induced cytotoxicity patterns despite difference in early NP internalization. On intravenous injection, the liver cleared the majority of NPs. Efficacy studies in the HBCx39 PDX model demonstrated an enhanced effect of drug-loaded PEBCA variants compared with free drug and PEHCA NPs. Furthermore, the folate conjugated PEBCA variant did not show any enhanced effects compared with the unconjugated counterpart which might be due to unfavorable orientation of folate on the NPs. Finally, analyses of the immune cell populations in tumors revealed that treatment with drug loaded PEBCA variants affected the myeloid cells, especially macrophages, contributing to an inflammatory, immune activated tumor microenvironment. Conclusion: We report for the first time, comparative efficacy of PEBCA and PEHCA NP variants in triple negative breast cancer models and show that CBZ-loaded PEBCA NPs exhibit a combined effect on tumor cells and on the tumor associated myeloid compartment, which may boost the anti-tumor response.


Assuntos
Neoplasias da Mama , Nanopartículas , Taxoides , Humanos , Feminino , Portadores de Fármacos , Distribuição Tecidual , Cianoacrilatos , Neoplasias da Mama/tratamento farmacológico , Ácido Fólico , Linhagem Celular Tumoral , Microambiente Tumoral
4.
Front Oncol ; 13: 1040665, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36910663

RESUMO

Assessment of drug sensitivity in tumor tissue ex vivo may significantly contribute to functional diagnostics to guide personalized treatment of cancer. Tumor organoid- and explant-cultures have become attractive tools towards this goal, although culturing conditions for breast cancer (BC) tissue have been among the most challenging to develop. Validation of possibilities to detect concordant responses in individual tumors and their respective cultures ex vivo is still needed. Here we employed BC patient-derived xenografts (PDXs) with distinct drug sensitivity, to evaluate different conditions for tissue dissociation, culturing and monitoring of treatment efficacy ex vivo, aiming to recapitulate the in vivo drug responses. The common challenge of discriminating between tumor and normal cells in the cultured tissue was also addressed. Following conventional enzymatic dissociation of BC tissue, the tumor cells stayed within the non-disrupted tissue fragments, while the single cells represented mostly normal host cells. By culturing such fragments as explants, viable tumor tissue could be maintained and treated ex vivo, providing representative indications on efficacy of the tested treatment. Thus, drug sensitivity profiles, including acquired chemoresistance seen in the PDXs, were recapitulated in the respective explants. To detect the concordant responses, however, the effect monitoring had to be harmonized with the characteristics of the cultured tissue. In conclusion, we present the feasibility of BC explants ex vivo to capture differences in drug sensitivity of individual tumors. The established protocols will aid in setting up an analogous platform for BC patient biopsies with the aim to facilitate functional precision medicine.

5.
Sci Rep ; 12(1): 5076, 2022 03 24.
Artigo em Inglês | MEDLINE | ID: mdl-35332208

RESUMO

More than half of metastatic melanoma patients receiving standard therapy fail to achieve a long-term survival due to primary and/or acquired resistance. Tumor cell ability to switch from epithelial to a more aggressive mesenchymal phenotype, attributed with AXLhigh molecular profile in melanoma, has been recently linked to such event, limiting treatment efficacy. In the current study, we investigated the therapeutic potential of the AXL inhibitor (AXLi) BGB324 alone or in combination with the clinically relevant BRAF inhibitor (BRAFi) vemurafenib. Firstly, AXL was shown to be expressed in majority of melanoma lymph node metastases. When treated ex vivo, the largest reduction in cell viability was observed when the two drugs were combined. In addition, a therapeutic benefit of adding AXLi to the BRAF-targeted therapy was observed in pre-clinical AXLhigh melanoma models in vitro and in vivo. When searching for mechanistic insights, AXLi was found to potentiate BRAFi-induced apoptosis, stimulate ferroptosis and inhibit autophagy. Altogether, our findings propose AXLi as a promising treatment in combination with standard therapy to improve therapeutic outcome in metastatic melanoma.


Assuntos
Melanoma , Neoplasias Cutâneas , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Humanos , Melanoma/patologia , Mutação , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/genética , Neoplasias Cutâneas/patologia , Vemurafenib/farmacologia
6.
Mol Oncol ; 15(8): 2026-2045, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33759347

RESUMO

Cellular phenotype plasticity between the epithelial and mesenchymal states has been linked to metastasis and heterogeneous responses to cancer therapy, and remains a challenge for the treatment of triple-negative breast cancer (TNBC). Here, we used isogenic human breast epithelial cell lines, D492 and D492M, representing the epithelial and mesenchymal phenotypes, respectively. We employed a CRISPR-Cas9 loss-of-function screen targeting a 2240-gene 'druggable genome' to identify phenotype-specific vulnerabilities. Cells with the epithelial phenotype were more vulnerable to the loss of genes related to EGFR-RAS-MAPK signaling, while the mesenchymal-like cells had increased sensitivity to knockout of G2 -M cell cycle regulators. Furthermore, we discovered knockouts that sensitize to the mTOR inhibitor everolimus and the chemotherapeutic drug fluorouracil in a phenotype-specific manner. Specifically, loss of EGFR and fatty acid synthase (FASN) increased the effectiveness of the drugs in the epithelial and mesenchymal phenotypes, respectively. These phenotype-associated genetic vulnerabilities were confirmed using targeted inhibitors of EGFR (gefitinib), G2 -M transition (STLC), and FASN (Fasnall). In conclusion, a CRISPR-Cas9 loss-of-function screen enables the identification of phenotype-specific genetic vulnerabilities that can pinpoint actionable targets and promising therapeutic combinations.


Assuntos
Sistemas CRISPR-Cas , Mutação com Perda de Função , Fenótipo , Neoplasias de Mama Triplo Negativas/patologia , Antineoplásicos/uso terapêutico , Proliferação de Células , Ensaios de Seleção de Medicamentos Antitumorais , Transição Epitelial-Mesenquimal , Everolimo/uso terapêutico , Feminino , Fluoruracila/uso terapêutico , Humanos , Transdução de Sinais/genética , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética
7.
Top Curr Chem ; 296: 251-81, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21504105

RESUMO

Photochemical internalization (PCI) is a novel technology for release of endocytosed macromolecules into the cytosol. The technology is based on the use of photosensitizers located in endocytic vesicles. Upon activation by light such photosensitizers induce a release of macromolecules from their compartmentalization in endocytic vesicles. PCI has been shown to increase the biological activity of a large variety of macromolecules and other molecules that do not readily penetrate the plasma membrane, including type I ribosome-inactivating proteins, immunotoxins, plasmids, adenovirus, various oligonucleotides, dendrimer-based delivery of chemotherapeutica and unconjugated chemotherapeutica such as bleomycin and doxorubicin. This review will present the basis for the PCI concept and the most recent significant developments.


Assuntos
Oligonucleotídeos/genética , Processos Fotoquímicos , Fármacos Fotossensibilizantes/metabolismo , Transfecção/métodos , Citosol/efeitos dos fármacos , Citosol/metabolismo , Endocitose/efeitos dos fármacos , Oligonucleotídeos/metabolismo , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia
8.
Sci Rep ; 10(1): 16992, 2020 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-33046784

RESUMO

In this study, we probed the importance of O-GlcNAc transferase (OGT) activity for the survival of tamoxifen-sensitive (TamS) and tamoxifen-resistant (TamR) breast cancer cells. Tamoxifen is an antagonist of estrogen receptor (ERα), a transcription factor expressed in over 50% of breast cancers. ERα-positive breast cancers are successfully treated with tamoxifen; however, a significant number of patients develop tamoxifen-resistant disease. We show that in vitro development of tamoxifen-resistance is associated with increased sensitivity to the OGT small molecule inhibitor OSMI-1. Global transcriptome profiling revealed that TamS cells adapt to OSMI-1 treatment by increasing the expression of histone genes. This is known to mediate chromatin compaction. In contrast, TamR cells respond to OGT inhibition by activating the unfolded protein response and by significantly increasing ERRFI1 expression. ERRFI1 is an endogenous inhibitor of ERBB-signaling, which is a known driver of tamoxifen-resistance. We show that ERRFI1 is selectively downregulated in ERα-positive breast cancers and breast cancers driven by ERBB2. This likely occurs via promoter methylation. Finally, we show that increased ERRFI1 expression is associated with extended survival in patients with ERα-positive tumors (p = 9.2e-8). In summary, we show that tamoxifen-resistance is associated with sensitivity to OSMI-1, and propose that this is explained in part through an epigenetic activation of the tumor-suppressor ERRFI1 in response to OSMI-1 treatment.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , N-Acetilglucosaminiltransferases/metabolismo , Tamoxifeno/uso terapêutico , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Neoplasias da Mama/mortalidade , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/metabolismo , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Células MCF-7 , N-Acetilglucosaminiltransferases/genética , RNA Interferente Pequeno/genética , Transdução de Sinais , Análise de Sobrevida , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Resposta a Proteínas não Dobradas
9.
Exp Cell Res ; 314(10): 2110-22, 2008 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-18423605

RESUMO

Malignant tumors comprise a small proportion of cancer-initiating cells (CIC), capable of sustaining tumor formation and growth. CIC are the main potential target for anticancer therapy. However, the identification of molecular therapeutic targets in CIC isolated from primary tumors is an extremely difficult task. Here, we show that after years of passaging under differentiating conditions, glioblastoma, mammary carcinoma, and melanoma cell lines contained a fraction of cells capable of forming spheroids upon in vitro growth under stem cell-like conditions. We found an increased expression of surface markers associated with the stem cell phenotype and of oncogenes in cell lines and clones cultured as spheroids vs. adherent cultures. Also, spheroid-forming cells displayed increased tumorigenicity and an altered pattern of chemosensitivity. Interestingly, also from single retrovirally marked clones, it was possible to isolate cells able to grow as spheroids and associated with increased tumorigenicity. Our findings indicate that short-term selection and propagation of CIC as spheroid cultures from established cancer cell lines, coupled with gene expression profiling, represents a suitable tool to study and therapeutically target CIC: the notion of which genes have been down-regulated during growth under differentiating conditions will help find CIC-associated therapeutic targets.


Assuntos
Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Neoplasias/terapia , Esferoides Celulares , Células-Tronco/fisiologia , Animais , Biomarcadores Tumorais/metabolismo , Técnicas de Cultura de Células , Transformação Celular Neoplásica , Feminino , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transplante de Neoplasias , Neoplasias/patologia , Análise de Sequência com Séries de Oligonucleotídeos
10.
Methods Mol Biol ; 434: 171-81, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18470645

RESUMO

Photochemical internalization (PCI) is a physico-chemical targeting method that enables light directed delivery of nucleic acids into cells. The technology is based on photosensitizers that localize in the membranes of endocytic vesicles. A light activation of the photosensitizers induces photochemical reactions that lead to rupture of the vesicular membranes. This results in the release of endocytosed compounds (e.g., nucleic acids) into the cell cytosol. Physico-chemical and biological targeting techniques can be combined to promote efficient and specific gene delivery to target cells. The present protocol describes PCI of epidermal growth factor receptor (EGFR)-targeted DNA polyplexes. The DNA polyplexes made are small (50-100 nm in diameter), and they contain polyethylenimine (PEI) conjugated with the EGF protein as a cell-binding ligand for EGFR-mediated endocytosis and polyethylene glycol (PEG) for masking the polyplex surface charge. PCI of such targeted PEG-PEI/DNA polyplexes enables high and EGFR-specific gene transfer activity in cells. Although describing in detail PCI of DNA polyplexes, the methodology presented in this protocol is also applicable for PCI of other gene therapy vectors (e.g. viral vectors), peptide nucleic acids (PNA), small interfering RNA (siRNA), and for vectors targeted to alternate cell surface receptors. Generally, PCI can be applied whenever 100% survival of the treated cell population is not required.


Assuntos
DNA/administração & dosagem , Receptores ErbB/metabolismo , Técnicas de Transferência de Genes , Fármacos Fotossensibilizantes/química , Polietilenoglicóis/química , Polietilenoimina/química , DNA/química , DNA/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Vetores Genéticos , Luz , Transfecção
11.
Cancer Lett ; 439: 1-13, 2018 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-30240588

RESUMO

Cancer cells' phenotypic plasticity, promoted by stromal cells, contributes to intra-tumoral heterogeneity and affects response to therapy. We have disclosed an association between fibroblast-stimulated phenotype switching and resistance to the clinically used BRAF inhibitor (BRAFi) vemurafenib in malignant melanoma, revealing a challenge in targeting the fibroblast-induced phenotype. Here we compared molecular features and drug sensitivity in melanoma cells grown as co-cultures with fibroblasts versus mono-cultures. In the presence of fibroblasts, melanoma cells switched to the dedifferentiated, mesenchymal-like, inflammatory phenotype that showed reduced sensitivity to the most of 275 tested cancer drugs. Fibroblasts, however, sensitized melanoma cells to PI3K inhibitors (PI3Ki) and particularly the inhibitor of GSK3, AR-A014418 (GSK3i), that showed superior efficacy in co-cultures. The proteome changes induced by the BRAFi + GSK3i combination mimicked changes induced by BRAFi in mono-cultures, and GSK3i in co-cultures. This suggests that the single drug drives the response to the combination treatment, depending on fibroblast presence or absence, consequently, phenotype. We propose that the BRAFi and GSK3i (or PI3Ki) combination exemplifies phenotype-specific combinatorial treatment that should be beneficial in phenotypically heterogeneous tumors rich in stromal interactions.


Assuntos
Adaptação Fisiológica/efeitos dos fármacos , Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Fibroblastos/efeitos dos fármacos , Melanoma/metabolismo , Apoptose/efeitos dos fármacos , Fibroblastos Associados a Câncer/efeitos dos fármacos , Fibroblastos Associados a Câncer/metabolismo , Fibroblastos Associados a Câncer/patologia , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Técnicas de Cocultura , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Melanoma/genética , Melanoma/patologia , Fenótipo , Inibidores de Proteínas Quinases/farmacologia
12.
Mol Oncol ; 12(9): 1540-1558, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29741811

RESUMO

The tumor microenvironment (TME) may influence both cancer progression and therapeutic response. In breast cancer, particularly in the aggressive triple-negative/basal-like subgroup, patient outcome is strongly associated with the tumor's inflammatory profile. Tumor-associated macrophages (TAMs) are among the most abundant immune cells in the TME, shown to be linked to poor prognosis and therapeutic resistance. In this study, we investigated the effect of the metastasis- and inflammation-associated microenvironmental factor S100A4 on breast cancer cells (BCCs) of different subtypes and explored their further interactions with myeloid cells. We demonstrated that extracellular S100A4 activates BCCs, particularly the basal-like subtype, to elevate secretion of pro-inflammatory cytokines. The secreted factors promoted conversion of monocytes to TAM-like cells that exhibited protumorigenic activities: stimulated epithelial-mesenchymal transition, proliferation, chemoresistance, and motility in cancer cells. In conclusion, we have shown that extracellular S100A4 instigates a tumor-supportive microenvironment, involving a network of cytokines and TAM-like cells, which was particularly characteristic for basal-like BCCs and potentiated their aggressive properties. The S100A4-BCC-TAM interaction cascade could be an important contributor to the aggressive behavior of this subtype and should be further explored for therapeutic targeting.


Assuntos
Citocinas/metabolismo , Macrófagos/metabolismo , Macrófagos/patologia , Proteína A4 de Ligação a Cálcio da Família S100/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Neoplasias de Mama Triplo Negativas/patologia , Microambiente Tumoral , Biópsia , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Resistencia a Medicamentos Antineoplásicos , Transição Epitelial-Mesenquimal , Feminino , Xenoenxertos , Humanos , Inflamação/metabolismo , Células MCF-7 , Monócitos/patologia , Esferoides Celulares
13.
FEBS Lett ; 580(24): 5739-46, 2006 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-17007842

RESUMO

The photochemical internalisation (PCI) technology liberates endocytosed macromolecules like transgenes from endocytic vesicles in response to photochemical treatment. Thereby PCI improves gene transfection and is suggested for use in gene therapy. It has been proposed that PCI might also stimulate transcription of internalised transgenes, especially if they are controlled by photochemically inducible promoters (transcriptional targeting). In order to identify inducible promoters, and to evaluate the treatments influence on cellular transcriptional activity, the effect of the photochemical treatment as used in PCI (with the photosensitizer disulfonated meso-tetraphenylporphin followed by illumination) on gene transcription in WiDr adenocarcinoma cells was evaluated using microarrays. The expression of 390 genes were identified significantly changed (89% were up-regulated), of which genes associated with DNA binding and transcriptional functions were the most represented. This may be important for the expression of a photochemically internalised transgene under a specific promoter control. Real-time PCR verified photochemical up-regulation of the HSP family genes, as well as down-regulation of EGR-1 at 2-10h post-treatment, suggesting that the HSP (particularly HSP70), in addition to the microarray-identified metallothioneins, but not the EGR-1 promoters, could be relevant promoter candidates for transcriptional targeting via PCI. The resulting overview of gene expression changes in WiDr cells exposed to the PCI-relevant photochemical treatment also provide a basis for the design of new PCI-based strategies with respect of transcriptional targeting.


Assuntos
Adenocarcinoma/genética , Neoplasias do Colo/genética , Fotoquímica , Fármacos Fotossensibilizantes/farmacologia , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/efeitos da radiação , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Proteínas de Choque Térmico/genética , Humanos , RNA Mensageiro/genética , Fatores de Tempo , Transcrição Gênica/genética
14.
Photochem Photobiol ; 82(3): 809-16, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16420102

RESUMO

Photochemical internalization (PCI) is a targeting technique that facilitates endosomal escape of macromolecules, such as transgenes, in response to photochemical treatment with endosome/lysosome-localized photosensitizers, such as disulfonated meso-tetraphenylporphine (TPPS(2a)). In gene therapy this leads to enhanced transgene expression. Moreover, photochemical treatment generally activates transcription of stress-response genes, such as heat-shock proteins (HSPs), via stimulation of corresponding promoters. Therefore, we used HSP70 (HSPp; a promoter from the HSP family gene) and investigated whether the PCI stimulus could also activate HSPp and thereby stimulate transcription (expression) of the HSPp-controlled transgene internalized via PCI. Using human colorectal carcinoma and hepatoma cell lines in vitro, we showed that TPPS(2a)-based photochemical treatment enhances expression of cellular HSP70, which correlated with a photochemically enhanced expression (approximately 2-fold, at PCI-optimal doses) of the HSPp-controlled transgene integrated in the genome. Furthermore, PCI enhanced expression of the HSPp-controlled episomal transgene delivered as a plasmid. However, in plasmid-based transfection, PCI-mediated enhancement with HSPp did not exceed the enhancement achieved with the constitutive active CMV promoter. In conclusion, we demonstrated that the PCI-relevant treatment initiates HSP70 response and that the HSP70 promoter can be used in combination with PCI, leading to PCI-enhanced expression of the HSPp-controlled transgene.


Assuntos
Proteínas de Choque Térmico HSP70/genética , Fotoquímica , Regiões Promotoras Genéticas , Transfecção/métodos , Linhagem Celular Tumoral , Endocitose , Terapia Genética/métodos , Humanos , Plasmídeos/administração & dosagem , Plasmídeos/farmacocinética , Porfirinas/farmacologia , Transgenes , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética
15.
J Environ Pathol Toxicol Oncol ; 25(1-2): 521-36, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16566739

RESUMO

Photochemical internalization (PCI) is a new technology, where certain photosensitizing substances (photosensitizers) are used to improve the utilization of macromolecules for cancer therapy, in a site-specific manner. Degradation of macromolecules in endocytic vesicles after uptake by endocytosis is a major intracellular barrier for the therapeutic application of molecules having intracellular targets of action. PCI is based on the light activation of photosensitizers specifically located in the membrane of endocytic vesicles inducing the rupture of this membrane upon illumination. Thereby endocytosed molecules can be released to reach their target of action before being degraded in lysosomes. This has been shown to enhance the biological activity of a large variety of macromolecules and other molecules that do not readily penetrate the plasma membrane, including type I ribosome-inactivating proteins (RIPs), immunotoxins, gene-encoding plasmids, adenovirus, peptidenucleic acids, and the chemotherapeuticum bleomycin. In several cases up to a 100-fold increase in biological activity has been observed. This article reviews the background and present status of PCI.


Assuntos
Sistemas de Liberação de Medicamentos , Endocitose , Fotoquimioterapia , Animais , Terapia Genética/métodos , Humanos , Luz , Preparações Farmacêuticas/administração & dosagem , Proteínas/administração & dosagem
16.
Cancer Res ; 63(13): 3490-4, 2003 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-12839932

RESUMO

Because peptide nucleic acids (PNAs) are poorly taken up by mammalian cells, strategies need to be developed for their intracellular delivery. In the present study, we demonstrated the possibility to efficiently release a naked PNA targeting the catalytic component of human telomerase reverse transcriptase (hTERT-PNA) into the cytoplasm of DU145 prostate cancer cells through the photochemical internalization approach. After light exposure, cells treated with the hTERT-PNA and photosensitizer TPPS(2a) showed a marked inhibition of telomerase activity and a reduced cell survival, which was not observed after treatment with hTERT-PNA alone. Moreover, in a direct comparison, photochemical internalization technology proved to be more efficient to internalize the hTERT-PNA than an HIV-Tat protein-based approach.


Assuntos
Ácidos Nucleicos Peptídicos/farmacocinética , Fotoquímica/métodos , Telomerase/metabolismo , Adenocarcinoma , Neoplasias Ósseas , Domínio Catalítico , Linhagem Celular , Citoplasma/metabolismo , Proteínas de Ligação a DNA , Humanos , Masculino , Osteossarcoma , Neoplasias da Próstata , Subunidades Proteicas/farmacocinética , Telomerase/química , Células Tumorais Cultivadas
17.
Cell Death Discov ; 2: 16081, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28028438

RESUMO

In recent years, new treatment options for malignant melanoma patients have enhanced the overall survival for selected patients. Despite new hope, most melanoma patients still relapse with drug-resistant tumors or experience intrinsic resistance to the therapy. Therefore, novel treatment modalities beneficial for subgroups of patients are needed. TRAIL receptor agonists have been suggested as promising candidates for use in cancer treatment as they preferentially induce apoptosis in cancer cells. Unfortunately, the first generation of TRAIL receptor agonists showed poor clinical efficacy. hvTRA is a second-generation TRAIL receptor agonist with improved composition giving increased potency, and in the present study, we showed hvTRA-induced activation of apoptosis leading to an efficient and sustained reduction in melanoma cell growth in cell lines and xenograft models. Furthermore, the potential of hvTRA in a clinical setting was demonstrated by showing efficacy on tumor cells harvested from melanoma patients with lymph node metastasis in an ex vivo drug sensitivity assay. Inhibition of mutated BRAF has been shown to regulate proteins in the intrinsic apoptotic pathway, making the cells more susceptible for apoptosis induction. In an attempt to increase the efficacy of hvTRA, combination treatment with the mutated BRAF inhibitor vemurafenib was investigated. A synergistic effect by the combination was observed for several cell lines in vitro, and an initial cytotoxic effect was observed in vivo. Unfortunately, the initial increased reduction in tumor growth compared with hvTRA mono treatment was not sustained, and this was related to downregulation of the DR5 level by vemurafenib. Altogether, the presented data imply that hvTRA efficiently induce apoptosis and growth delay in melanoma models and patient material, and the potential of this TRAIL receptor agonist should be further evaluated for treatment of subgroups of melanoma patients.

18.
Oncotarget ; 7(15): 19997-20015, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26918352

RESUMO

The knowledge on how tumor-associated stroma influences efficacy of anti-cancer therapy just started to emerge. Here we show that lung fibroblasts reduce melanoma sensitivity to the BRAF inhibitor (BRAFi) vemurafenib only if the two cell types are in close proximity. In the presence of fibroblasts, the adjacent melanoma cells acquire de-differentiated mesenchymal-like phenotype. Upon treatment with BRAFi, such melanoma cells maintain high levels of phospho ribosomal protein S6 (pS6), i.e. active mTOR signaling, which is suppressed in the BRAFi sensitive cells without stromal contacts. Inhibitors of PI3K/mTOR in combination with BRAFi eradicate pS6high cell subpopulations and potentiate anti-cancer effects in melanoma protected by the fibroblasts. mTOR and BRAF co-inhibition also delayed the development of early-stage lung metastases in vivo. In conclusion, we demonstrate that upon influence from fibroblasts, melanoma cells undergo a phenotype switch to the mesenchymal state, which can support PI3K/mTOR signaling. The lost sensitivity to BRAFi in such cells can be overcome by co-targeting PI3K/mTOR. This knowledge could be explored for designing BRAFi combination therapies aiming to eliminate both stroma-protected and non-protected counterparts of metastases.


Assuntos
Fibroblastos/patologia , Melanoma/patologia , Mesoderma/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Animais , Apoptose , Proliferação de Células , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Melanoma/tratamento farmacológico , Melanoma/metabolismo , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Camundongos Nus , Transdução de Sinais/efeitos dos fármacos , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Células Estromais/patologia , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Nanoscale ; 8(2): 862-77, 2016 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-26648525

RESUMO

Therapeutic nanoparticles (NPs) have great potential to deliver drugs against human diseases. Encapsulation of drugs in NPs protects them from being metabolized, while they are delivered specifically to a target site, thereby reducing toxicity and other side-effects. However, non-specific tissue accumulation of NPs, for example in macrophages, especially in the spleen and liver is a general problem with many NPs being developed for cancer therapy. To address the problem of non-specific tissue accumulation of NPs we describe the development of the zebrafish embryo as a transparent vertebrate system for characterization of NPs against cancer. We show that injection of human cancer cells results in tumor-like structures, and that subsequently injected fluorescent NPs, either made of polystyrene or liposomes can be imaged in real-time. NP biodistribution and general in vivo properties can be easily monitored in embryos having selective fluorescent labeling of specific tissues. We demonstrate in vitro, by using optical tweezer micromanipulation, microscopy and flow cytometry that polyethylene glycol (PEG) coating of NPs decreases the level of adhesion of NPs to macrophages, and also to cancer cells. In vivo in zebrafish embryos, PEG coating resulted in longer NP circulation times, decreased macrophage uptake, and reduced adhesion to the endothelium. Importantly, liposomes were observed to accumulate passively and selectively in tumor-like structures comprised of human cancer cells. These results show that zebrafish embryo is a powerful system for microscopy-based screening of NPs on the route to preclinical testing.


Assuntos
Micromanipulação/métodos , Nanopartículas/química , Neoplasias/tratamento farmacológico , Peixe-Zebra/embriologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Citometria de Fluxo , Corantes Fluorescentes/química , Células HEK293 , Humanos , Lipossomos/química , Macrófagos/metabolismo , Nanopartículas Metálicas/química , Microscopia , Microscopia Eletrônica de Varredura , Microscopia Eletrônica de Transmissão , Nanomedicina/métodos , Neoplasias/metabolismo , Neoplasias/terapia , Pinças Ópticas , Polietilenoglicóis/química , Polímeros/química , Poliestirenos/química , Distribuição Tecidual
20.
Biochim Biophys Acta ; 1570(3): 210-8, 2002 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-12020812

RESUMO

The efficiency of gene transfection mediated by nonviral vectors is limited because of nonoptimal intracellular trafficking of transfecting DNA. Most nonviral vectors deliver transfecting DNA into a cell through endocytosis. However, poor escape from endocytic vesicles and inefficient transport of DNA into the nucleus often limits a success of gene transfection. Photochemical transfection is a new method, based on light-induced permeabilisation of endocytic vesicles, liberating transfecting DNA into the cytosol, concurrently increasing the chances for DNA to enter the nucleus. The aim of this study was to investigate the role of the cell cycle for the efficiency of photochemical transfection. It was demonstrated that in asynchronous human colon carcinoma HCT 116 cells photochemical treatment increased the transfection mediated by the nonviral vectors, the cationic polypeptide polylysine and the cationic lipid N-(2-aminoethyl)-N,N-dimethyl-2,3-bis(tetradecyloxy)-1-propanaminium bromide/dioleoylphosphatidylethanolamine (beta AE-DMRIE/DOPE), by 30- and 2.5-fold, respectively. In aphidicolin-synchronised cells, photochemical transfection mediated by polylysine was dependent on the cell cycle: transfection level was 4-fold higher when illumination, inducing photochemical reactions, was performed during the G2/M phase as compared to the G1/early-S phase. The cell cycle influence on photochemical transfection mediated by beta AE-DMRIE/DOPE was very low: only 20% difference between G2/M and the G1/S phase was observed. We suggest that transgenes, photochemically liberated close/during mitosis, perhaps have the highest opportunity to enter the nucleus and be expressed. However, the dependence of photochemical transfection on the cell cycle might be partially disguised by various factors induced by photochemical treatment.


Assuntos
Ciclo Celular/fisiologia , Fotoquímica/métodos , Transfecção/métodos , Afidicolina/farmacologia , Citometria de Fluxo , Fluoresceína-5-Isotiocianato , Humanos , Lipídeos , Oligonucleotídeos/metabolismo , Fosfatidiletanolaminas , Biossíntese de Proteínas , Compostos de Amônio Quaternário , Transgenes , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA