Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Crit Rev Oncol Hematol ; 198: 104355, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38621469

RESUMO

Trastuzumab deruxtecan (T-DXd) is an antibody-drug conjugate (ADC) consisting of a humanised, anti-human epidermal growth factor receptor 2 (HER2) monoclonal antibody covalently linked to a topoisomerase I inhibitor cytotoxic payload (DXd). The high drug-to-antibody ratio (8:1) ensures a high DXd concentration is delivered to target tumour cells, following internalisation of T-DXd and subsequent cleavage of its tetrapeptide-based linker. DXd's membrane-permeable nature enables it to cross cell membranes and potentially exert antitumour activity on surrounding tumour cells regardless of HER2 expression. T-DXd's unique mechanism of action is reflected in its efficacy in clinical trials in patients with HER2-positive advanced breast cancer (in heavily pretreated populations and in those previously treated with a taxane and trastuzumab), as well as HER2-low metastatic breast cancer. Thus, ADCs such as T-DXd have the potential to change the treatment paradigm of targeting HER2 in metastatic breast cancer, including eventually within the adjuvant/neoadjuvant setting.


Assuntos
Neoplasias da Mama , Camptotecina , Imunoconjugados , Trastuzumab , Humanos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Trastuzumab/uso terapêutico , Feminino , Imunoconjugados/uso terapêutico , Imunoconjugados/farmacologia , Camptotecina/análogos & derivados , Camptotecina/uso terapêutico , Receptor ErbB-2/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Antineoplásicos Imunológicos/uso terapêutico , Antineoplásicos Imunológicos/farmacologia
2.
Exp Ther Med ; 22(4): 1127, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34504579

RESUMO

Ocoxin Oral Solution (OOS) and Viusid (VS) are nutritional supplements that include several natural products which affect different cellular functions, such as proliferation or the redox status. In addition, some of their constituent components have been described to exert an antiviral effect. Considering this, it was hypothesized that treatment with OOS and VS could protect from viral infections. In order to evaluate the impact of OOS and VS on viral infection, lentivirus and retrovirus whose genomes coded for green fluorescent protein were used. In addition, and as a second approach to measure viral infection, a hemagglutinin-tagged form of the mitogen-activated protein kinase ERK5 was also inserted in the retroviral vector. Viral particles produced in 293T cells were used to infect HeLa cells in the presence or absence of OOS or VS. It was observed that VS had a minimal effect on the capacity of either lentivirus or retrovirus to infect HeLa cells. However, OOS significantly reduced the infection of HeLa cells with both of these viruses. The effect was dose-dependent, reaching a maximum at a 1:100 dilution of OOS. These results suggested that, in addition to its well-known antitumoral properties, OOS may also inhibit infection with viruses. This effect is relevant since patients receiving oncological therapies are more susceptible to viral infections, and nutritional supplements such as OOS may help in reducing the severity of these potential pathogenic infections.

3.
Cancers (Basel) ; 14(1)2021 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-35008318

RESUMO

During recent years, a number of new compounds against HER2 have reached clinics, improving the prognosis and quality of life of HER2-positive breast cancer patients. Nonetheless, resistance to standard-of-care drugs has motivated the development of novel agents, such as new antibody-drug conjugates (ADCs). The latter are a group of drugs that benefit from the potency of cytotoxic agents whose action is specifically guided to the tumor by the target-specific antibody. Two anti-HER2 ADCs have reached the clinic: trastuzumab-emtansine and, more recently, trastuzumab-deruxtecan. In addition, several other HER2-targeted ADCs are in preclinical or clinical development, some of them with promising signs of activity. In the present review, the structure, mechanism of action, and potential resistance to all these ADCs will be described. Specific attention will be given to discussing novel strategies to circumvent resistance to ADCs.

4.
Nutrients ; 12(9)2020 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-32878230

RESUMO

Ocoxin Oral Solution (OOS) is a nutritional supplement whose formulation includes several plant extracts and natural products with demonstrated antitumoral properties. This review summarizes the antitumoral action of the different constituents of OOS. The action of this formulation on different preclinical models as well as clinical trials is reviewed, paying special attention to the mechanism of action and quality of life improvement properties of this nutritional supplement. Molecularly, its mode of action includes a double edge role on tumor biology, that involves a slowdown in cell proliferation accompanied by cell death induction. Given the safety and good tolerability of OOS, and its potentiation of the antitumoral effect of other standard of care drugs, OOS may be used in the oncology clinic in combination with conventional therapies.


Assuntos
Ácido Ascórbico/farmacologia , Suplementos Nutricionais , Ácido Fólico/farmacologia , Ácido Pantotênico/farmacologia , Extratos Vegetais/farmacologia , Vitamina B 12/farmacologia , Vitamina B 6/farmacologia , Sulfato de Zinco/farmacologia , Aminoácidos/farmacologia , Animais , Antineoplásicos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cinnamomum zeylanicum/química , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Glucosamina/farmacologia , Glycyrrhiza/química , Humanos , Neoplasias/tratamento farmacológico , Sacarose/análogos & derivados , Sacarose/farmacologia , Chá/química
5.
Angiogenesis ; 23(2): 231-247, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31897911

RESUMO

Endoglin (CD105) is an auxiliary receptor for members of the TFG-ß superfamily. Whereas it has been demonstrated that the deficiency of endoglin leads to minor and defective angiogenesis, little is known about the effect of its increased expression, characteristic of several types of cancer. Angiogenesis is essential for tumor growth, so high levels of proangiogenic molecules, such as endoglin, are supposed to be related to greater tumor growth leading to a poor cancer prognosis. However, we demonstrate here that endoglin overexpression do not stimulate sprouting or vascularization in several in vitro and in vivo models. Instead, steady endoglin overexpression keep endothelial cells in an active phenotype that results in an impairment of the correct stabilization of the endothelium and the recruitment of mural cells. In a context of continuous enhanced angiogenesis, such as in tumors, endoglin overexpression gives rise to altered vessels with an incomplete mural coverage that permit the extravasation of blood. Moreover, these alterations allow the intravasation of tumor cells, the subsequent development of metastases and, thus, a worse cancer prognosis.


Assuntos
Endoglina/genética , Metástase Neoplásica/genética , Neoplasias/irrigação sanguínea , Neoplasias/genética , Neovascularização Patológica/genética , Animais , Movimento Celular/genética , Células Cultivadas , Endoglina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Invasividade Neoplásica , Neoplasias/patologia , Neovascularização Patológica/patologia , Regulação para Cima/genética
6.
Cancer Lett ; 470: 161-169, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31765734

RESUMO

Small molecule inhibitors (TKIs) of HER2 have demonstrated clinical benefit in HER2-positive breast tumors. One of them, lapatinib, is used once advanced tumors become refractory to the HER2 antibody trastuzumab. Another one, neratinib, has shown benefit in high-risk early-stage breast cancer after trastuzumab-based therapies. A common characteristic is that patients are formerly treated with trastuzumab. We have explored whether trastuzumab previous therapy affects its antitumoral action. Long time exposure of the HER2+ cell line BT474 to trastuzumab resulted in trastuzumab-insensitive cells (BTRH cells). While treatment of wild type BT474 cells with lapatinib or neratinib resulted in decreased viability, BTRH cells were resistant to the action of these TKIs. Analogous results were obtained using trastuzumab-resistant cells derived from a PDX. Functional transcriptomic analyses and biochemical studies demonstrated that the TKIs caused DNA damage and apoptosis in wild type cells, but not in BTRH. Moreover, previous treatment with trastuzumab impairs response to small TKIs, by eliminating their proapoptotic action. Moreover, actioning on the apoptotic machinery using a chemical library of proapoptotic compounds led to the identification of clinical-stage drugs that may be used to fight trastuzumab-TKI resistance.


Assuntos
Antineoplásicos Imunológicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Trastuzumab/farmacologia , Animais , Antineoplásicos Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Neoplasias da Mama/genética , Neoplasias da Mama/imunologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Dano ao DNA/efeitos dos fármacos , Feminino , Perfilação da Expressão Gênica , Humanos , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Lapatinib/farmacologia , Lapatinib/uso terapêutico , Camundongos , Naftoquinonas/farmacologia , Naftoquinonas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Piridinas/farmacologia , Piridinas/uso terapêutico , Quinolinas/farmacologia , Quinolinas/uso terapêutico , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Receptor ErbB-2/imunologia , Trastuzumab/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Nutrients ; 11(5)2019 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-31091680

RESUMO

Nutritional supplements which include natural antitumoral compounds could represent safe and efficient additives for cancer patients. One such nutritional supplement, Ocoxin Oral solution (OOS), is a composite formulation that contains several antioxidants and exhibits antitumoral properties in several in vitro and in vivo tumor conditions. Here, we performed a functional genomic analysis to uncover the mechanism of the antitumoral action of OOS. Using in vivo models of acute myelogenous leukemia (AML, HEL cells, representative of a liquid tumor) and small-cell lung cancer (GLC-8, representative of a solid tumor), we showed that OOS treatment altered the transcriptome of xenografted tumors created by subcutaneously implanting these cells. Functional transcriptomic studies pointed to a cell cycle deregulation after OOS treatment. The main pathway responsible for this deregulation was the E2F-TFDP route, which was affected at different points. The alterations ultimately led to a decrease in pathway activation. Moreover, when OOS-deregulated genes in the AML context were analyzed in patient samples, a clear correlation with their levels and prognosis was observed. Together, these data led us to suggest that the antitumoral effect of OOS is due to blockade of cell cycle progression mainly caused by the action of OOS on the E2F-TFDP pathway.


Assuntos
Ácido Ascórbico/farmacologia , Ciclo Celular/efeitos dos fármacos , Neoplasias Experimentais/tratamento farmacológico , Extratos Vegetais/farmacologia , Vitamina B 12/farmacologia , Vitamina B 6/farmacologia , Animais , Ácido Ascórbico/administração & dosagem , Linhagem Celular Tumoral , Feminino , Ácido Fólico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos Nus , Ácido Pantotênico , Extratos Vegetais/administração & dosagem , Transcriptoma , Vitamina B 12/administração & dosagem , Vitamina B 6/administração & dosagem , Sulfato de Zinco
8.
Cancer Lett ; 453: 34-44, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-30928382

RESUMO

The appearance of resistance to the anti-HER2 targeted drug trastuzumab constitutes, nowadays, an important challenge in the oncology clinic. To fight such resistance, we searched for potential vulnerabilities in cells resistant to that drug. To that end, we used cell lines primary resistant to trastuzumab, as well as cells made secondarily resistant to the drug upon continuous exposure. Using genomic and proteomic approaches, a deregulation in cell death pathways was identified in trastuzumab-resistant cells. More precisely, an increased response to the death factor TRAIL, caused by an increase in the cellular receptors for this factor, was observed. In parallel, a decrease in inhibitory components of the pathway was detected. This combination produces a more efficient assembly of the functional complex in the trastuzumab-resistant cells that translates in the observed increased response to TRAIL. Analysis of HER2 positive patient samples confirmed deregulation of this pathway in trastuzumab-resistant patients. Taken together our data identify a vulnerability of trastuzumab-resistant cells that could be used to design new targeted therapies in that context.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Receptor ErbB-2/metabolismo , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Trastuzumab/farmacologia , Antineoplásicos Imunológicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Feminino , Células HEK293 , Humanos , Receptor ErbB-2/antagonistas & inibidores , Receptor ErbB-2/genética , Receptores do Ligante Indutor de Apoptose Relacionado a TNF/genética , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Transcriptoma
9.
Int J Oncol ; 53(1): 113-123, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29658597

RESUMO

Lung cancer is the most frequently diagnosed neoplasia and represents the leading cause of cancer-related deaths worldwide. Due to this fact, efforts to improve patient survival through the introduction of novel therapies, as well as preventive actions, are urgently required. Considering this scenario, the antitumoral action of the composite formulation Ocoxin® oral solution (OOS), that contains several antitumoral compounds including antioxidants, was tested in small cell lung cancer (SCLC) in vitro and in vivo preclinical models. OOS exhibited anti-SCLC action that was both time and dose dependent. In vivo OOS decreased the growth of tumors implanted in mice without showing signs of toxicity. The antitumoral effect was due to inhibition of cell proliferation and increased cell death. Genomic and biochemical analyses indicated that OOS augmented p27 and decreased the functioning of several routes involved in cell proliferation. In addition, OOS caused cell death by activation of caspases. Importantly, OOS favored the action of several standard of care drugs used in the SCLC clinic. Our results suggest that OOS has antitumoral action on SCLC, and could be used to supplement the action of drugs commonly used to treat this type of tumor.


Assuntos
Antioxidantes/administração & dosagem , Ácido Ascórbico/administração & dosagem , Suplementos Nutricionais , Extratos Vegetais/administração & dosagem , Carcinoma de Pequenas Células do Pulmão/dietoterapia , Vitamina B 12/administração & dosagem , Vitamina B 6/administração & dosagem , Administração Oral , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Composição de Medicamentos , Ácido Fólico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Camundongos , Ácido Pantotênico , Antígeno Nuclear de Célula em Proliferação/genética , Carcinoma de Pequenas Células do Pulmão/patologia , Ensaios Antitumorais Modelo de Xenoenxerto , Sulfato de Zinco
10.
Haematologica ; 102(12): 2113-2124, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28860344

RESUMO

Kinesin spindle protein inhibition is known to be an effective therapeutic approach in several malignancies. Filanesib (ARRY-520), an inhibitor of this protein, has demonstrated activity in heavily pre-treated multiple myeloma patients. The aim of the work herein was to investigate the activity of filanesib in combination with pomalidomide plus dexamethasone backbone, and the mechanisms underlying the potential synergistic effect. The ability of filanesib to enhance the activity of pomalidomide plus dexamethasone was studied in several in vitro and in vivo models. Mechanisms of this synergistic combination were dissected by gene expression profiling, immunostaining, cell cycle and short interfering ribonucleic acid studies. Filanesib showed in vitro, ex vivo, and in vivo synergy with pomalidomide plus dexamethasone treatment. Importantly, the in vivo synergy observed in this combination was more evident in large, highly proliferative tumors, and was shown to be mediated by the impairment of mitosis transcriptional control, an increase in monopolar spindles, cell cycle arrest and the induction of apoptosis in cells in proliferative phases. In addition, the triple combination increased the activation of the proapoptotic protein BAX, which has previously been associated with sensitivity to filanesib, and could potentially be used as a predictive biomarker of response to this combination. Our results provide preclinical evidence for the potential benefit of the combination of filanesib with pomalidomide and dexamethasone, and supported the initiation of a recently activated trial being conducted by the Spanish Myeloma group which is investigating this combination in relapsed myeloma patients.


Assuntos
Dexametasona/uso terapêutico , Cinesinas/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Talidomida/análogos & derivados , Tiadiazóis/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Células Cultivadas , Sinergismo Farmacológico , Humanos , Camundongos , Talidomida/uso terapêutico , Resultado do Tratamento
11.
Oncol Lett ; 14(2): 1950-1958, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28781639

RESUMO

Hepatocellular carcinoma (HCC) is becoming one of the most prevalent types of cancer worldwide. The most efficient types of treatment at present include surgical resection and liver transplantation, but these treatments may only be used in a small percentage of patients. In order to identify novel therapeutic strategies for this disease, the present study explored the potential antitumoral effect of Ocoxin® oral solution (OOS) in HCC. OOS inhibited the proliferation of HCC cell lines in a time- and dose-dependent manner, being more efficient when used in combination with sorafenib, a standard of care treatment for patients diagnosed with advanced-stage disease. Mechanistic studies indicated that the effect of OOS was due to the induction of cell cycle arrest rather than the stimulation of apoptotic cell death. The cell cycle was slowed down in all phases in the HCC cell lines treated with OOS. Finally, when tested in animal models of HCC, OOS reduced tumor progression through the induction of necrosis in xenograft tumor models. Considering the poor prognosis and high resistance to antitumor treatments of HCC, the antiproliferative action of OOS, particularly in combination with sorafenib, provides the opportunity to investigate the effect of combined therapy in a clinical setting.

12.
Cancer Res ; 77(17): 4639-4651, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28687619

RESUMO

Trastuzumab-emtansine (T-DM1) is an antibody-drug conjugate (ADC) that was approved recently to treat HER2+ breast cancers. Despite its impressive clinical efficacy in many patients, intrinsic and acquired resistance to T-DM1 has emerged as a challenge. To identify mechanisms of T-DM1 resistance, we isolated several resistant HER2+ clones exhibiting stable drug refractoriness in vitro and in vivo Genomic comparisons showed substantial differences among three of the isolated clones, indicating several potential mechanisms of resistance to T-DM1. However, we observed no differences in HER2 levels and signaling among the resistant models and parental HER2+ cells. Bioinformatics studies suggested that intracellular trafficking of T-DM1 could underlie resistance to T-DM1, and systematic analysis of the path followed by T-DM1 showed that the early steps in the internalization of the drug were unaltered. However, in some of the resistant clones, T-DM1 accumulated in lysosomes. In these clones, lysosomal pH was increased and the proteolytic activity of these organelles was deranged. These results were confirmed in T-DM1-resistant cells from patient-derived HER2+ samples. We postulate that resistance to T-DM1 occurs through multiple mechanisms, one of which is impaired lysosomal proteolytic activity. Because other ADC may use the same internalization-degradation pathway to deliver active payloads, strategies aimed at restoring lysosomal functionality might overcome resistance to ADC-based therapies and improve their effectiveness. Cancer Res; 77(17); 4639-51. ©2017 AACR.


Assuntos
Anticorpos Monoclonais Humanizados/farmacologia , Neoplasias da Mama/patologia , Resistencia a Medicamentos Antineoplásicos , Imunoconjugados/farmacologia , Lisossomos/metabolismo , Maitansina/análogos & derivados , Proteólise/efeitos dos fármacos , Receptor ErbB-2/antagonistas & inibidores , Ado-Trastuzumab Emtansina , Animais , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/enzimologia , Feminino , Perfilação da Expressão Gênica , Humanos , Lisossomos/efeitos dos fármacos , Maitansina/farmacologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Trastuzumab , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Oncotarget ; 7(5): 6231-42, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26756220

RESUMO

Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy whose incidence is growing in developed countries. In the relapse setting, very limited therapeutic options are available and in most cases only palliative care can be offered to patients. The effect of a composite formulation that contains several antioxidants, Ocoxin Oral solution (OOS), was tested in this condition. When analyzed in vitro, OOS exhibited anti-AML action that was both time and dose dependent. In vivo OOS induced a ralentization of tumor growth that was due to a decrease in cell proliferation. Such effect could, at least partially, be due to an increase in the cell cycle inhibitor p27, although other cell cycle proteins seemed to be altered. Besides, OOS induced an immunomodulatory effect through the induction of IL6. When tested in combination with other therapeutic agents normally used in the treatment of AML patients, OOS demonstrated a higher antiproliferative action, suggesting that it may be used in combination with those standard of care treatments to potentiate their antiproliferative action in the AML clinic.


Assuntos
Ácido Ascórbico/farmacologia , Leucemia Mieloide Aguda/tratamento farmacológico , Extratos Vegetais/farmacologia , Vitamina B 12/farmacologia , Vitamina B 6/farmacologia , Animais , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Ácido Fólico , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Camundongos Nus , Ácido Pantotênico , Distribuição Aleatória , Ensaios Antitumorais Modelo de Xenoenxerto , Sulfato de Zinco
14.
Leuk Lymphoma ; 57(1): 167-76, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-25860244

RESUMO

The use of thalidomide derivatives (IMIDs) has improved multiple myeloma prognosis, through an unknown mechanism of action. Recently one molecular target, the cereblon (CRBN) protein, has been identified. CRBN acts by binding to DDB1-CUL4-ROC1 forming a ubiquitin ligase multiprotein complex. We have generated antibodies to different regions of CRBN protein, and analyzed the biological consequences of augmenting or decreasing CRBN levels. CRBN was expressed in all the myeloma cell lines tested, independently of their sensitivity to IMIDs, and the CRBN-DDB1-CUL4 complex was efficiently formed. At the molecular level, long-term treatment with IMIDs induced a slight decrease in CRBN levels and a reduction in the CRBN-DDB1-CUL4 complex. Interestingly, treatment with other anti-myeloma drugs downregulated cellular contents of CRBN, and in a much faster fashion. These results suggest that CRBN is an important mediator of the cellular response to IMIDs, but also critical in the maintenance of cell viability and/or proliferation.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Peptídeo Hidrolases/genética , Peptídeo Hidrolases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Apoptose/efeitos dos fármacos , Apoptose/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células , Citosol/metabolismo , Proteínas de Ligação a DNA/metabolismo , Humanos , Mieloma Múltiplo/patologia , Complexos Multiproteicos/metabolismo , Ligação Proteica , Talidomida/análogos & derivados , Talidomida/farmacologia , Ubiquitina-Proteína Ligases
15.
Oncotarget ; 5(18): 8284-305, 2014 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-25268740

RESUMO

Despite evidence about the implication of the bone marrow (BM) stromal microenvironment in multiple myeloma (MM) cell growth and survival, little is known about the effects of myelomatous cells on BM stromal cells. Mesenchymal stromal cells (MSCs) from healthy donors (dMSCs) or myeloma patients (pMSCs) were co-cultured with the myeloma cell line MM.1S, and the transcriptomic profile of MSCs induced by this interaction was analyzed. Deregulated genes after co-culture common to both d/pMSCs revealed functional involvement in tumor microenvironment cross-talk, myeloma growth induction and drug resistance, angiogenesis and signals for osteoclast activation and osteoblast inhibition. Additional genes induced by co-culture were exclusively deregulated in pMSCs and predominantly associated to RNA processing, the ubiquitine-proteasome pathway, cell cycle regulation, cellular stress and non-canonical Wnt signaling. The upregulated expression of five genes after co-culture (CXCL1, CXCL5 and CXCL6 in d/pMSCs, and Neuregulin 3 and Norrie disease protein exclusively in pMSCs) was confirmed, and functional in vitro assays revealed putative roles in MM pathophysiology. The transcriptomic profile of pMSCs co-cultured with myeloma cells may better reflect that of MSCs in the BM of myeloma patients, and provides new molecular insights to the contribution of these cells to MM pathophysiology and to myeloma bone disease.


Assuntos
Doenças Ósseas/genética , Células da Medula Óssea/metabolismo , Comunicação Celular , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Mieloma Múltiplo/genética , Doenças Ósseas/metabolismo , Doenças Ósseas/patologia , Células da Medula Óssea/patologia , Linhagem Celular Tumoral , Análise por Conglomerados , Técnicas de Cocultura , Progressão da Doença , Perfilação da Expressão Gênica/métodos , Redes Reguladoras de Genes , Humanos , Células-Tronco Mesenquimais/patologia , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , RNA Mensageiro/metabolismo , Transdução de Sinais , Nicho de Células-Tronco , Microambiente Tumoral
16.
Mol Cancer Ther ; 13(2): 504-16, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24431075

RESUMO

The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that regulates cell growth, proliferation, metabolism, and cell survival, and plays those roles by forming two functionally distinct multiprotein complexes: mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2). Deregulation of the mTOR pathway has been found in different cancers, including multiple myeloma. Agents acting on mTORC1, such as rapamycin and derivatives, are being explored as antitumoral strategies. However, whether targeting mTOR would be a more effective antimyeloma strategy than exclusively acting on the mTORC1 branch remains to be established. In this report, we explored the activation status of mTOR routes in malignant plasma cells, and analyzed the contribution of mTOR and its two signaling branches to the proliferation of myeloma cells. Gene expression profiling demonstrated deregulation of mTOR pathway-related genes in myeloma plasma cells from patients. Activation of the mTOR pathway in myelomatous plasma cells was corroborated by flow cytometric analyses. RNA interference (RNAi) experiments indicated that mTORC1 predominated over mTORC2 in the control of myeloma cell proliferation. However, mTOR knockdown had a superior antiproliferative effect than acting only on mTORC1 or mTORC2. Pharmacologic studies corroborated that the neutralization of mTOR has a stronger antimyeloma effect than the individual inhibition of mTORC1 or mTORC2. Together, our data support the clinical development of agents that widely target mTOR, instead of agents, such as rapamycin or its derivatives, that solely act on mTORC1.


Assuntos
Mieloma Múltiplo/metabolismo , Complexos Multiproteicos/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Antibióticos Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Western Blotting , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Cocultura , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Imidazóis/farmacologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Alvo Mecanístico do Complexo 2 de Rapamicina , Mieloma Múltiplo/tratamento farmacológico , Mieloma Múltiplo/genética , Complexos Multiproteicos/antagonistas & inibidores , Complexos Multiproteicos/genética , Pirazinas/farmacologia , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Sirolimo/farmacologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Células Tumorais Cultivadas
17.
PLoS One ; 6(11): e27583, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22132115

RESUMO

Multiple myeloma (MM) is a hematological disease characterized by an abnormal accumulation of plasma cells in the bone marrow. These cells have frequent cytogenetic abnormalities including translocations of the immunoglobulin heavy chain gene and chromosomal gains and losses. In fact, a singular characteristic differentiating MM from other hematological malignancies is the presence of a high degree of aneuploidies. As chromosomal abnormalities can be generated by alterations in the spindle assembly checkpoint (SAC), the functionality of such checkpoint was tested in MM. When SAC components were analyzed in MM cell lines, the RNA levels of most of them were conserved. Nevertheless, the protein content of some key constituents was very low in several cell lines, as was the case of MAD2 or CDC20 in RPMI-8226 or RPMI-LR5 cells. The recovery of their cellular content did not substantially affect cell growth, but improved their ability to segregate chromosomes. Finally, SAC functionality was tested by challenging cells with agents disrupting microtubule dynamics. Most of the cell lines analyzed exhibited functional defects in this checkpoint. Based on the data obtained, alterations both in SAC components and their functionality have been detected in MM, pointing to this pathway as a potential target in MM treatment.


Assuntos
Pontos de Checagem da Fase M do Ciclo Celular , Mieloma Múltiplo/patologia , Aneuploidia , Proteínas de Ligação ao Cálcio/metabolismo , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Instabilidade Genômica/efeitos dos fármacos , Humanos , Pontos de Checagem da Fase M do Ciclo Celular/genética , Proteínas Mad2 , Mieloma Múltiplo/genética , Nocodazol/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/metabolismo , Retroviridae/efeitos dos fármacos , Retroviridae/genética , Transdução Genética
18.
Bioorg Med Chem Lett ; 21(22): 6641-5, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21986585

RESUMO

A Phenotypic Drug Discovery strategy was applied to study a set of pyrimidine analogs prepared by means of intramolecular oxidation-reduction reactions of N-substituted-N-(2,6-disubstituted-5-nitro-4-pyrimidinyl)aminoacetic acid methyl esters in basic media. The combined and rational use of specific assays allowed in short time reducing from all possible cellular targets to those involved in metaphase to anaphase transition.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Ciclo Celular/efeitos dos fármacos , Descoberta de Drogas , Pirimidinas/química , Pirimidinas/farmacologia , Anáfase/efeitos dos fármacos , Antineoplásicos/síntese química , Linhagem Celular Tumoral , Humanos , Metáfase/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Oxirredução , Pirimidinas/síntese química , Relação Estrutura-Atividade
19.
J Cell Sci ; 123(Pt 18): 3146-56, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20736311

RESUMO

The MAP kinase Erk5 plays important roles in cellular proliferation, and has recently been implicated in the regulation of mitosis. The classic pathway of Erk5 activation involves dual phosphorylation at its TEY microdomain by the upstream regulating kinase MEK5. Here we describe a second pathway that controls Erk5 phosphorylation. This pathway is activated in mitotic cells and involves kinase activities distinct from MEK5. Studies aimed at identifying these kinases suggested that CDK1 activity is required to sustain Erk5 phosphorylation in mitosis, as treatment with RO3306, a CDK1 inhibitor, reversed mitotic phosphorylation of Erk5. Moreover, CDK1 co-precipitated with Erk5 in mitotic cells. The mitotic phosphorylation of Erk5 occurs at multiple sites located at its unique C-terminal region, within an Erk5 subdomain that has formerly been implicated in the control of the subcellular location of Erk5. Furthermore, molecular studies indicated that phosphorylation at these sites may participate in the control of the transit of Erk5 between the cytosol and the nucleus, in addition to regulating its transcriptional activity. Together, our results demonstrate the existence of a second Erk5 phosphorylation pathway, that is activated in mitosis, and that may participate in the regulation of Erk5 functions.


Assuntos
Proteína Quinase 7 Ativada por Mitógeno/metabolismo , Mitose , Motivos de Aminoácidos , Sequência de Aminoácidos , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Linhagem Celular Tumoral , Núcleo Celular/química , Núcleo Celular/genética , Núcleo Celular/metabolismo , Citosol/química , Citosol/metabolismo , Humanos , MAP Quinase Quinase 5/genética , MAP Quinase Quinase 5/metabolismo , Proteína Quinase 7 Ativada por Mitógeno/química , Proteína Quinase 7 Ativada por Mitógeno/genética , Dados de Sequência Molecular , Fosforilação , Transporte Proteico
20.
Proc Natl Acad Sci U S A ; 105(43): 16719-24, 2008 Oct 28.
Artigo em Inglês | MEDLINE | ID: mdl-18940925

RESUMO

Hec1 (Highly Expressed in Cancer 1) is one of four proteins of the outer kinetochore Ndc80 complex involved in the dynamic interface between centromeres and spindle microtubules. Its overexpression is seen in a variety of human tumors and correlates with tumor grade and prognosis. We show here that the overexpression of Hec1 in an inducible mouse model results in mitotic checkpoint hyperactivation. As previously observed with overexpression of the Mad2 gene, hyperactivation of the mitotic checkpoint leads to aneuploidy in vitro and is sufficient to generate tumors in vivo that harbor significant levels of aneuploidy. These results underscore the role of chromosomal instability as a result of mitotic checkpoint hyperactivation in the initiation of tumorigenesis.


Assuntos
Proteínas de Ciclo Celular/farmacologia , Mitose , Neoplasias/etiologia , Proteínas Nucleares/genética , Proteínas Nucleares/farmacologia , Aneuploidia , Animais , Proteínas de Ciclo Celular/administração & dosagem , Proteínas de Ciclo Celular/genética , Instabilidade Cromossômica , Doxiciclina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Cinetocoros , Camundongos , Camundongos Transgênicos , Proteínas Associadas aos Microtúbulos , Neoplasias/genética , Proteínas Nucleares/administração & dosagem , Distribuição Tecidual
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...