Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 120(14): e2220413120, 2023 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-36972439

RESUMO

Human epidermal growth factor receptor 2 (HER2) is overexpressed in various cancer types. HER2-targeting trastuzumab plus chemotherapy is used as first-line therapy for HER2-positive recurrent or primary metastatic gastric cancer, but intrinsic and acquired trastuzumab resistance inevitably develop over time. To overcome gastric cancer resistance to HER2-targeted therapies, we have conjugated trastuzumab with a beta-emitting therapeutic isotope, lutetium-177, to deliver radiation locally to gastric tumors with minimal toxicity. Because trastuzumab-based targeted radioligand therapy (RLT) requires only the extramembrane domain binding of membrane-bound HER2 receptors, HER2-targeting RLT can bypass any resistance mechanisms that occur downstream of HER2 binding. Leveraging our previous discoveries that statins, a class of cholesterol-lowering drugs, can enhance the cell surface-bound HER2 to achieve effective drug delivery in tumors, we proposed that the combination of statins and [177Lu]Lu-trastuzumab-based RLT can enhance the therapeutic efficacy of HER2-targeted RLT in drug-resistant gastric cancers. We demonstrate that lovastatin elevates cell surface HER2 levels and increases the tumor-absorbed radiation dose of [177Lu]Lu-DOTA-trastuzumab. Furthermore, lovastatin-modulated [177Lu]Lu-DOTA-trastuzumab RLT durably inhibits tumor growth and prolongs overall survival in mice bearing NCI-N87 gastric tumors and HER2-positive patient-derived xenografts (PDXs) of known clinical resistance to trastuzumab therapy. Statins also exhibit a radioprotective effect, reducing radiotoxicity in a mice cohort given the combination of statins and [177Lu]Lu-DOTA-trastuzumab. Since statins are commonly prescribed to patients, our results strongly support the feasibility of clinical studies that combine lovastatin with HER2-targeted RLT in HER2-postive patients and trastuzumab-resistant HER2-positive patients.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Neoplasias Gástricas , Humanos , Animais , Camundongos , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/radioterapia , Neoplasias Gástricas/metabolismo , Inibidores de Hidroximetilglutaril-CoA Redutases/uso terapêutico , Preparações Farmacêuticas , Receptor ErbB-2/metabolismo , Trastuzumab/farmacologia , Trastuzumab/uso terapêutico , Lovastatina/farmacologia , Lovastatina/uso terapêutico , Linhagem Celular Tumoral
2.
Mol Pharm ; 17(1): 327-337, 2020 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-31804840

RESUMO

Human epidermal growth factor receptor 2 (HER2) is a biomarker in breast cancer, and its overexpression is required to initiate therapies using HER2-targeted antibodies. Although trastuzumab is one of the most effective therapeutic antibodies in HER2-overexpressing breast cancer, a significant number of patients do not benefit from this therapy due to inherent or acquired resistance mechanisms. One reported mechanism of resistance is the steric hindering effect caused by the polymeric complex formed between hyaluronan and CD44, thus preventing trastuzumab from binding to HER2. Hyaluronan/CD44 contributes as an obstacle for trastuzumab to bind HER2, but it is also involved in HER2 internalization. In this study, we used zirconium-89 (89Zr)-labeled trastuzumab immunoPET to investigate whether degradation of hyaluronan can resensitize HER2-overexpressing breast cancer cells to trastuzumab. Targeted degradation of endogenously produced hyaluronan and inhibition of its synthesis were achieved by treating trastuzumab-resistant JIMT1 breast cancer cells with hyaluronidase (HLX) and 4-methylumbelliferone (4MU). The 4MU/HLX treatment reduced HER2 internalization by depleting hyaluronan/CD44 and the caveolin-1 (CAV1) endocytic protein, resulting in enhanced membrane-bound 89Zr-labeled trastuzumab. 4MU/HLX enhanced trastuzumab tumor uptake, as evidenced by increased tumor binding of the 89Zr-labeled trastuzumab in JIMT1 tumor xenografts. In vitro mechanistic studies demonstrated a decrease in HER2-mediated oncogenic signaling upon cell treatment with 4MU/HLX. Importantly, 4MU/HLX enhanced trastuzumab efficacy in JIMT1 xenografts. These data showed the utility of 89Zr-labeled trastuzumab as a PET imaging agent to monitor the affinity of the antibody to HER2 during CD44/hyaluronan-specific inhibition with the overall goal of improving trastuzumab therapy.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Neoplasias da Mama/diagnóstico por imagem , Neoplasias da Mama/tratamento farmacológico , Ácido Hialurônico/metabolismo , Tomografia por Emissão de Pósitrons , Receptor ErbB-2/metabolismo , Trastuzumab/uso terapêutico , Animais , Anticorpos Monoclonais Humanizados/farmacocinética , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Antineoplásicos Imunológicos/farmacocinética , Antineoplásicos Imunológicos/farmacologia , Neoplasias da Mama/enzimologia , Neoplasias da Mama/metabolismo , Caveolina 1/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Receptores de Hialuronatos/metabolismo , Hialuronoglucosaminidase , Himecromona/farmacologia , Camundongos , Camundongos Nus , Radioisótopos , Receptor ErbB-2/genética , Distribuição Tecidual , Trastuzumab/farmacocinética , Trastuzumab/farmacologia , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto , Zircônio
3.
Mol Pharm ; 17(6): 2145-2154, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32339462

RESUMO

Photodynamic therapy (PDT) has demonstrated encouraging anticancer therapeutic results, but the current clinically approved photosensitizers (PSs) are not ideal in the treatment of bladder cancer. Conventional PSs have low selectivity to the bladder tumor tissue and induce toxicity or bystander effects on nontumor urothelium. Previous studies demonstrated that the use of galactose-photosensitizer (PS) conjugates is a more selective method of delivering PDT-mediated toxicity due to their ability to recognize carbohydrate-binding domains overexpressed in bladder tumors. Using patient-derived bladder tumor specimens cultured ex vivo and bladder cancer cell lines with different PDT sensitivity, we find that a galactose-phthalocyanine (PcGal16) accumulates in bladder tumors expressing galactose-binding proteins and internalizes through an endocytic process. The endocytosis mechanism is cell line-dependent. In HT-1376 bladder cancer lines resistant to PDT, depletion of caveolin-1-the main structural protein of caveolae structures-increased the amount of sugar-binding proteins, i.e. GLUT1, at the cell membrane resulting in an improved PcGal16 uptake and PDT efficacy. These data show the potential of ex vivo cultures of bladder cancer, that ideally could mimic the original microenvironment, in screening galacto-PDT agents. Additionally, our studies demonstrate that PDT efficacy in bladder cancer depends on the endocytic mechanisms that regulate PS accumulation and internalization in cancer cells.


Assuntos
Caveolina 1/metabolismo , Indóis/química , Indóis/uso terapêutico , Fotoquimioterapia/métodos , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/terapia , Idoso , Western Blotting , Caveolina 1/genética , Linhagem Celular Tumoral , Endocitose/efeitos dos fármacos , Feminino , Galectina 1/genética , Galectina 1/metabolismo , Galectina 3/genética , Galectina 3/metabolismo , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Humanos , Técnicas In Vitro , Isoindóis , Masculino , Microscopia de Fluorescência
4.
Molecules ; 25(20)2020 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-33050602

RESUMO

The specificity of antibodies for antigens overexpressed or uniquely expressed in tumor cells makes them ideal candidates in the development of bioconjugates for tumor imaging. Molecular imaging can aid clinicians in the diagnosis of gastric tumors and in selecting patients for therapies targeting receptors with a heterogeneous intratumoral or intertumoral expression. Antibodies labeled with an imaging radiometal can be used to detect primary tumors and metastases using whole-body positron emission tomography (PET) or single photon emission computed tomography (SPECT), both during diagnosis and monitoring disease response. Conjugated with fluorescent dyes, antibodies can image tumors by targeted optical imaging. This review provides an overview of the most recent advances in the use of antibodies labeled with radiometals or conjugated with fluorescent dyes for gastric cancer imaging.


Assuntos
Tomografia por Emissão de Pósitrons/métodos , Neoplasias Gástricas/diagnóstico por imagem , Tomografia Computadorizada de Emissão de Fóton Único/métodos , Animais , Humanos , Imagem Óptica/métodos
5.
Org Biomol Chem ; 17(10): 2579-2593, 2019 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-30648722

RESUMO

Photodynamic therapy (PDT) combines a photosensitizer (PS) with the physical energy of non-ionizing light to trigger cell death pathways. PDT has potential as a therapeutic modality to be used in alternative or in combination with other conventional cancer treatment protocols (e.g. surgery, chemotherapy and radiotherapy). Still, due to the lack of specificity of the current PSs to target the tumor cells, several studies have exploited their conjugation with targeting moieties. PSs conjugated with antibodies (Abs) or their fragments, able to bind antigens overexpressed in the tumors, have demonstrated potential in PDT of tumors. This review provides an overview of the most recent advances on photoimmunoconjugates (PICs) for cancer PDT, which involve the first and second-generation PSs conjugated to Abs. This is an update of our previous review "Antibodies armed with photosensitizers: from chemical synthesis to photobiological applications", published in 2015 in Org. Biomol. Chem.


Assuntos
Imunoconjugados/uso terapêutico , Neoplasias/terapia , Fármacos Fotossensibilizantes/uso terapêutico , Animais , Anticorpos Monoclonais/imunologia , Receptores ErbB/imunologia , Humanos , Imunoconjugados/imunologia , Luz , Nanopartículas Metálicas/química , Nanopartículas Metálicas/efeitos da radiação , Neoplasias/imunologia , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/efeitos da radiação
6.
Molecules ; 24(4)2019 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-30781675

RESUMO

The stabilization of G-Quadruplex DNA structures by ligands is a promising strategy for telomerase inhibition in cancer therapy since this enzyme is responsible for the unlimited proliferation of cancer cells. To assess the potential of a compound as a telomerase inhibitor, selectivity for quadruplex over duplex DNA is a fundamental attribute, as the drug must be able to recognize quadruplex DNA in the presence of a large amount of duplex DNA, in the cellular nucleus. By using different spectroscopic techniques, such as ultraviolet-visible, fluorescence and circular dichroism, this work evaluates the potential of a series of multicharged phthalocyanines, bearing four or eight positive charges, as G-Quadruplex stabilizing ligands. This work led us to conclude that the existence of a balance between the number and position of the positive charges in the phthalocyanine structure is a fundamental attribute for its selectivity for G-Quadruplex structures over duplex DNA structures. Two of the studied phthalocyanines, one with four peripheral positive charges (ZnPc1) and the other with less exposed eight positive charges (ZnPc4) showed high selectivity and affinity for G-Quadruplex over duplex DNA structures and were able to accumulate in the nucleus of UM-UC-3 bladder cancer cells.


Assuntos
DNA/química , Inibidores Enzimáticos/química , Corantes Fluorescentes/química , Quadruplex G/efeitos dos fármacos , Indóis/química , Linhagem Celular Tumoral , Complexos de Coordenação/química , Humanos , Isoindóis , Ligantes , Espectrometria de Fluorescência/métodos , Relação Estrutura-Atividade , Telomerase/antagonistas & inibidores , Zinco/química
7.
Bioconjug Chem ; 29(2): 306-315, 2018 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-29313666

RESUMO

The use of glycosylated compounds is actively pursued as a therapeutic strategy for cancer due to the overexpression of various types of sugar receptors and transporters on most cancer cells. Conjugation of saccharides to photosensitizers such as porphyrins provides a promising strategy to improve the selectivity and cell uptake of the photosensitizers, enhancing the overall photosensitizing efficacy. Most porphyrin-carbohydrate conjugates are linked via the carbon-1 position of the carbohydrate because this is the most synthetically accessible approach. Previous studies suggest that carbon-1 galactose derivatives show diminished binding since the hydroxyl group in the carbon-1 position of the sugar is a hydrogen bond acceptor in the galectin-1 sugar binding site. We therefore synthesized two isomeric porphyrin-galactose conjugates using click chemistry: one linked via the carbon-1 of the galactose and one linked via carbon-3. Free base and zinc analogs of both conjugates were synthesized. We assessed the uptake and photodynamic therapeutic (PDT) activity of the two conjugates in both monolayer and spheroidal cell cultures of four different cell lines. For both the monolayer and spheroid models, we observe that the uptake of both conjugates is proportional to the protein levels of galectin-1 and the uptake is suppressed after preincubation with an excess of thiogalactose, as measured by fluorescence spectroscopy. Compared to that of the carbon-1 conjugate, the uptake of the carbon-3 conjugate was greater in cell lines containing high expression levels of galectin-1. After photodynamic activation, MTT and lactate dehydrogenase assays demonstrated that the conjugates induce phototoxicity in both monolayers and spheroids of cancer cells.


Assuntos
Galactose/análogos & derivados , Galactose/farmacologia , Neoplasias/tratamento farmacológico , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/farmacologia , Porfirinas/química , Porfirinas/farmacologia , Carbono/química , Linhagem Celular Tumoral , Galactose/síntese química , Galactose/farmacocinética , Humanos , Fotoquimioterapia , Fármacos Fotossensibilizantes/síntese química , Fármacos Fotossensibilizantes/farmacocinética , Porfirinas/síntese química , Porfirinas/farmacocinética
8.
Int J Cancer ; 141(7): 1478-1489, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28639285

RESUMO

Gastric cancer (GC) is the 3rd deadliest cancer worldwide, due to limited treatment options and late diagnosis. Human epidermal growth factor receptor-2 (HER2) is overexpressed in ∼20% of GC cases and anti-HER2 antibody trastuzumab in combination with conventional chemotherapy, is recognized as standard therapy for HER2-positive metastatic GC. This strategy improves GC patients' survival by 2-3 months, however its optimal results in breast cancer indicate that GC survival may be improved. A new photoimmunoconjugate was developed by conjugating a porphyrin with trastuzumab (Trast:Porph) for targeted photodynamic therapy in HER2-positive GC. Using mass spectrometry analysis, the lysine residues in the trastuzumab structure most prone for porphyrin conjugation were mapped. The in vitro data demonstrates that Trast:Porph specifically binds to HER2-positive cells, accumulates intracellularly, co-localizes with lysosomal marker LAMP1, and induces massive HER2-positive cell death upon cellular irradiation. The high selectivity and cytotoxicity of Trast:Porph based photoimmunotherapy is confirmed in vivo in comparison with trastuzumab alone, using nude mice xenografted with a HER2-positive GC cell line. In the setting of human disease, these data suggest that repetitive cycles of Trast:Porph photoimmunotherapy may be used as an improved treatment strategy in HER2-positive GC patients.


Assuntos
Antineoplásicos/uso terapêutico , Morte Celular , Imunoterapia/métodos , Fotoquimioterapia/métodos , Porfirinas/uso terapêutico , Receptor ErbB-2 , Neoplasias Gástricas/tratamento farmacológico , Trastuzumab/uso terapêutico , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Humanos , Lisina/química , Proteínas de Membrana Lisossomal/farmacocinética , Masculino , Espectrometria de Massas , Camundongos Nus , Porfirinas/química , Porfirinas/farmacocinética , Distribuição Aleatória , Neoplasias Gástricas/metabolismo , Trastuzumab/química , Trastuzumab/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Bioconjug Chem ; 27(11): 2762-2769, 2016 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-27750007

RESUMO

Here, we report the rational design of a new third-generation photosensitizer (PS), a chlorin conjugated with galactodendritic units, ChlGal8, to improve the effectiveness of bladder cancer treatment. ChlGal8 shows better photochemical and photophysical properties than a recently reported homologous porphyrin, PorGal8. In addition to inheriting excellent photostability, the ability to generate singlet oxygen, and the ability to interact with the proteins galectin-1 and human serum albumin (HSA), ChlGal8 exhibits high absorption in the red region of the electromagnetic spectrum. In vitro studies of anticancer activity of ChlGal8 revealed that once this PS is taken up by UM-UC-3 bladder cancer cells, it induces high cytotoxicity after a single dose of light irradiation. In HT-1376 bladder cancer cells resistant to therapy, a second light irradiation treatment enhanced in vitro and in vivo photodynamic efficacy. The enhanced phototoxicity in HT-1376 cancer cells seems to be due to the ability of ChlGal8 to accumulate in the mitochondria, via facilitative glucose transporter 1 (GLUT1), in the period between single and repeated irradiation. A photodynamic therapy (PDT) regimen using an extra dose of light irradiation and ChlGal8 as PS represents a promising strategy in treating resistant cancers in a clinical setting.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Galactose/química , Mitocôndrias/efeitos dos fármacos , Fotoquimioterapia , Porfirinas/química , Porfirinas/farmacologia , Neoplasias da Bexiga Urinária/patologia , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/efeitos da radiação , Linhagem Celular Tumoral , Desenho de Fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Galectina 1/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Humanos , Luz , Masculino , Camundongos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos da radiação , Fármacos Fotossensibilizantes/química , Fármacos Fotossensibilizantes/metabolismo , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Porfirinas/metabolismo , Porfirinas/uso terapêutico , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/efeitos da radiação , Albumina Sérica/metabolismo , Neoplasias da Bexiga Urinária/tratamento farmacológico
10.
Inorg Chem ; 54(9): 4382-93, 2015 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-25897563

RESUMO

This work employed [5,10,15,20-tetrakis(pentafluorophenyl)porphyrin] ([H2(TPPF20)], H2P1) as the platform to prepare a tetrasubstituted galactodendritic conjugate porphyrin (H2P3). After metalation with excess copper(II) acetate, H2P3 afforded a new solid porphyrin material, Cu4CuP3S. This work also assessed the ability of the copper(II) complex (CuP3) of H2P3 to coordinate with zinc(II) acetate, to yield the new material Zn4CuP3S. UV-visible, Fourier transform infrared, and electron paramagnetic resonance spectroscopies aided full characterization of the synthesized solids. (Z)-Cyclooctene epoxidation under heterogeneous conditions helped to evaluate the catalytic activity of Cu4CuP3S and Zn4CuP3S. The efficiency of Cu4CuP3S in the oxidation of another organic substrate, catechol, was also investigated. According to the results obtained in the heterogeneous process, Cu4CuP3S mimicked the activity of cytochrome P-450 and catecholase. In addition, Cu4CuP3S was reusable after recovery and reactivation. The data obtained herein were compared with the results achieved for the copper complex (CuP1) of [H2(TPPF20)] and for CuP3 under homogeneous conditions.


Assuntos
Materiais Biomiméticos/química , Catecóis/química , Metaloporfirinas/química , Compostos Organometálicos/química , Acetato de Zinco/química , Catálise , Catecol Oxidase/química , Sistema Enzimático do Citocromo P-450/química , Espectroscopia de Ressonância de Spin Eletrônica , Estrutura Molecular , Oxirredução
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA