Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 137(17): 2360-2372, 2021 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-33150374

RESUMO

Clonal evolution drives tumor progression, dissemination, and relapse in multiple myeloma (MM), with most patients dying of relapsed disease. This multistage process requires tumor cells to enter the circulation, extravasate, and colonize distant bone marrow (BM) sites. Here, we developed a fluorescent or DNA-barcode clone-tracking system on MM PrEDiCT (progression through evolution and dissemination of clonal tumor cells) xenograft mouse model to study clonal behavior within the BM microenvironment. We showed that only the few clones that successfully adapt to the BM microenvironment can enter the circulation and colonize distant BM sites. RNA sequencing of primary and distant-site MM tumor cells revealed a progression signature sequentially activated along human MM progression and significantly associated with overall survival when evaluated against patient data sets. A total of 28 genes were then computationally predicted to be master regulators (MRs) of MM progression. HMGA1 and PA2G4 were validated in vivo using CRISPR-Cas9 in the PrEDiCT model and were shown to be significantly depleted in distant BM sites, indicating their role in MM progression and dissemination. Loss of HMGA1 and PA2G4 also compromised the proliferation, migration, and adhesion abilities of MM cells in vitro. Overall, our model successfully recapitulates key characteristics of human MM disease progression and identified potential new therapeutic targets for MM.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/metabolismo , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Proteína HMGA1a/metabolismo , Mieloma Múltiplo/patologia , Recidiva Local de Neoplasia/patologia , Proteínas de Ligação a RNA/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Apoptose , Biomarcadores Tumorais/genética , Medula Óssea/metabolismo , Medula Óssea/patologia , Sistemas CRISPR-Cas , Adesão Celular , Movimento Celular , Proliferação de Células , Evolução Clonal , Progressão da Doença , Feminino , Proteína HMGA1a/antagonistas & inibidores , Proteína HMGA1a/genética , Humanos , Camundongos , Camundongos SCID , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Prognóstico , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Taxa de Sobrevida , Células Tumorais Cultivadas
2.
Cancer Sci ; 113(4): 1113-1124, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35122353

RESUMO

Immunotherapy is currently recognized as the fourth modality in cancer therapy. CTL can detect cancer cells via complexes involving human leukocyte antigen (HLA) class I molecules and peptides derived from tumor antigens, resulting in antigen-specific cancer rejection. The peptides may be predicted in silico using machine learning-based algorithms. Neopeptides, derived from neoantigens encoded by somatic mutations in cancer cells, are putative immunotherapy targets, as they have high tumor specificity and immunogenicity. Here, we used our pipeline to select 278 neoepitopes with high predictive "SCORE" from the tumor tissues of 46 patients with hepatocellular carcinoma or metastasis of colorectal carcinoma. We validated peptide immunogenicity and specificity by in vivo vaccination with HLA-A2, A24, B35, and B07 transgenic mice using ELISpot assay, in vitro and in vivo killing assays. We statistically evaluated the power of our prediction algorithm and demonstrated the capacity of our pipeline to predict neopeptides (area under the curve = 0.687, P < 0.0001). We also analyzed the potential of long peptides containing the predicted neoepitopes to induce CTLs. Our study indicated that the short peptides predicted using our algorithm may be intrinsically present in tumor cells as cleavage products of long peptides. Thus, we empirically demonstrated that the accuracy and specificity of our prediction tools may be potentially improved in vivo using the HLA transgenic mouse model. Our data will help to design feedback algorithms to improve in silico prediction, potentially allowing researchers to predict peptides for personalized immunotherapy.


Assuntos
Algoritmos , Antígenos de Neoplasias , Vacinas Anticâncer , Carcinoma Hepatocelular , Antígenos HLA , Neoplasias Hepáticas , Animais , Antígenos HLA/genética , Antígeno HLA-A2/genética , Antígenos de Histocompatibilidade Classe I , Antígenos de Histocompatibilidade Classe II , Humanos , Camundongos , Camundongos Transgênicos , Peptídeos , Medicina de Precisão , Linfócitos T Citotóxicos
3.
Int J Urol ; 29(1): 7-15, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34605079

RESUMO

OBJECTIVE: To confirm if mirabegron 50 mg shows efficacy in women with overactive bladder and either urgency urinary incontinence or mixed urinary incontinence versus placebo. METHODS: Post-hoc analyses were carried out using pooled data from a Japanese phase IIb and a phase III study. The primary efficacy end-point was baseline to end-of-treatment change in the mean number of micturitions/24 h. The secondary end-points were changes in the mean voided volume/micturition, mean number of urgency and incontinence episodes/24 h, and mean number of nocturia episodes/night. Other end-points were quality of life and incontinence normalization rates. RESULTS: Women with urgency urinary incontinence (placebo n = 204, mirabegron n = 214) and mixed urinary incontinence (placebo n = 122, mirabegron n = 139) were included. Change in mean micturitions/24 h at end-of-treatment for mirabegron was statistically significant versus placebo in both populations; the effect size increased over time. For all secondary end-points, median changes for mirabegron were statistically significant versus placebo at end-of-treatment, except for nocturia for the urgency urinary incontinence population and urgency for the mixed urinary incontinence population. Mirabegron showed larger improvements versus placebo in all quality-of-life domains, except for general health perception in the urgency urinary incontinence population. Incontinence normalization rates for mirabegron were 47.2% and 49.6% in the urgency urinary incontinence and mixed urinary incontinence populations, respectively, versus 42.6% and 39.3% for placebo. CONCLUSIONS: Mirabegron 50 mg significantly improved key overactive bladder symptoms versus placebo in women with urgency urinary incontinence, and it also improved most overactive bladder symptoms, including micturition frequency, in patients with mixed urinary incontinence. These findings support the benefits of using mirabegron in the female overactive bladder wet population.


Assuntos
Bexiga Urinária Hiperativa , Incontinência Urinária , Acetanilidas/uso terapêutico , Agonistas de Receptores Adrenérgicos beta 3/uso terapêutico , Método Duplo-Cego , Feminino , Humanos , Japão/epidemiologia , Antagonistas Muscarínicos , Qualidade de Vida , Tiazóis , Resultado do Tratamento , Bexiga Urinária Hiperativa/tratamento farmacológico , Bexiga Urinária Hiperativa/epidemiologia , Incontinência Urinária/tratamento farmacológico , Incontinência Urinária/epidemiologia
4.
Am J Hematol ; 94(11): 1244-1253, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31456261

RESUMO

We tested the hypothesis that using CXCR4 inhibition to target the interaction between the tumor cells and the microenvironment leads to sensitization of the tumor cells to apoptosis. Eligibility criteria included multiple myeloma (MM) patients with 1-5 prior lines of therapy. The purposes of the phase I study were to evaluate the safety and maximal-tolerated dose (MTD) of the combination. The treatment-related adverse events and response rate of the combination were assessed in the phase II study. A total of 58 patients were enrolled in the study. The median age of the patients was 63 years (range, 43-85), and 78% of them received prior bortezomib. In the phase I study, the MTD was plerixafor 0.32 mg/kg, and bortezomib 1.3 mg/m2 . The overall response rate for the phase II study was 48.5%, and the clinical benefit rate 60.6%. The median disease-free survival was 12.6 months. The CyTOF analysis demonstrated significant mobilization of plasma cells, CD34+ stem cells, and immune T cells in response to plerixafor. This is an unprecedented study that examines therapeutic targeting of the bone marrow microenvironment and its interaction with the tumor clone to overcome resistance to therapy. Our results indicate that this novel combination is safe and that the objective response rate is high even in patients with relapsed/refractory MM. ClinicalTrials.gov, NCT00903968.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Mieloma Múltiplo/tratamento farmacológico , Proteínas de Neoplasias/antagonistas & inibidores , Receptores CXCR4/antagonistas & inibidores , Terapia de Salvação , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Apoptose/efeitos dos fármacos , Benzilaminas , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Bortezomib/administração & dosagem , Bortezomib/efeitos adversos , Terapia Combinada , Ciclamos , Intervalo Livre de Doença , Relação Dose-Resposta a Droga , Feminino , Gastroenteropatias/induzido quimicamente , Doenças Hematológicas/induzido quimicamente , Transplante de Células-Tronco Hematopoéticas , Compostos Heterocíclicos/administração & dosagem , Compostos Heterocíclicos/efeitos adversos , Humanos , Estimativa de Kaplan-Meier , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Mieloma Múltiplo/genética , Mieloma Múltiplo/terapia , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Recidiva , Microambiente Tumoral/efeitos dos fármacos
5.
Blood ; 127(21): 2598-606, 2016 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-26903547

RESUMO

Familial aggregation of Waldenström macroglobulinemia (WM) cases, and the clustering of B-cell lymphoproliferative disorders among first-degree relatives of WM patients, has been reported. Nevertheless, the possible contribution of inherited susceptibility to familial WM remains unrevealed. We performed whole exome sequencing on germ line DNA obtained from 4 family members in which coinheritance for WM was documented in 3 of them, and screened additional independent 246 cases by using gene-specific mutation sequencing. Among the shared germ line variants, LAPTM5(c403t) and HCLS1(g496a) were the most recurrent, being present in 3/3 affected members of the index family, detected in 8% of the unrelated familial cases, and present in 0.5% of the nonfamilial cases and in <0.05 of a control population. LAPTM5 and HCLS1 appeared as relevant WM candidate genes that characterized familial WM individuals and were also functionally relevant to the tumor clone. These findings highlight potentially novel contributors for the genetic predisposition to familial WM and indicate that LAPTM5(c403t) and HCLS1(g496a) may represent predisposition alleles in patients with familial WM.


Assuntos
Proteínas Sanguíneas/genética , Exoma , Predisposição Genética para Doença , Mutação em Linhagem Germinativa , Proteínas de Membrana/genética , Macroglobulinemia de Waldenstrom/genética , Proteínas Adaptadoras de Transdução de Sinal , Família , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino
6.
Am J Hematol ; 92(8): E138-E145, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28474779

RESUMO

The tumor suppressors B-lymphocyte-induced maturation protein-1 (BLIMP-1) and p53 play a crucial role in B-cell lymphomas, and their inactivation contributes to the pathogenesis of a wide spectrum of lymphoid malignancies, including diffuse large B-cell lymphomas (DLBCLs). Patients with activated B-cell-like (ABC) DLBCL may present with loss of BLIMP-1, c-Myc over-expression, decreased p53, and poor prognosis. Nevertheless, there is a lack of in vivo models recapitulating the biology of high-grade ABC DLBCL. We therefore aimed to develop an in vivo model aiming to recapitulate the phenotype observed in this cohort of patients. A Cre-Lox approach was used to achieve inactivation of both p53 and BLIMP-1 in murine B-cells. Contextual ablation of BLIMP-1 and p53 led to development of IgM-positive B-cell lymphoma with an aggressive phenotype, supported by c-Myc up-regulation, and accumulation of somatic mutations, as demonstrated by whole exome sequencing. Sensitivity of B-tumor cells to BTK inhibition was demonstrated. This model mirrors what reported in patients with ABC DLBLC, and therefore represents a novel model for studying the biology of ABC-DLBCL harboring the dual loss of BLIMP-1/p53 and c-Myc over-expression.


Assuntos
Linfócitos B/metabolismo , Linfócitos B/patologia , Transformação Celular Neoplásica/genética , Proteínas Repressoras/deficiência , Proteína Supressora de Tumor p53/deficiência , Animais , Linfócitos B/efeitos dos fármacos , Biomarcadores , Evolução Clonal/genética , Modelos Animais de Doenças , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes myc , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Camundongos , Camundongos Transgênicos , Fator 1 de Ligação ao Domínio I Regulador Positivo , Inibidores de Proteínas Quinases/farmacologia
7.
Proc Natl Acad Sci U S A ; 111(28): 10287-92, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24982170

RESUMO

Bone is a favorable microenvironment for tumor growth and a frequent destination for metastatic cancer cells. Targeting cancers within the bone marrow remains a crucial oncologic challenge due to issues of drug availability and microenvironment-induced resistance. Herein, we engineered bone-homing polymeric nanoparticles (NPs) for spatiotemporally controlled delivery of therapeutics to bone, which diminish off-target effects and increase local drug concentrations. The NPs consist of poly(D,L-lactic-co-glycolic acid) (PLGA), polyethylene glycol (PEG), and bisphosphonate (or alendronate, a targeting ligand). The engineered NPs were formulated by blending varying ratios of the synthesized polymers: PLGA-b-PEG and alendronate-conjugated polymer PLGA-b-PEG-Ald, which ensured long circulation and targeting capabilities, respectively. The bone-binding ability of Ald-PEG-PLGA NPs was investigated by hydroxyapatite binding assays and ex vivo imaging of adherence to bone fragments. In vivo biodistribution of fluorescently labeled NPs showed higher retention, accumulation, and bone homing of targeted Ald-PEG-PLGA NPs, compared with nontargeted PEG-PLGA NPs. A library of bortezomib-loaded NPs (bone-targeted Ald-Bort-NPs and nontargeted Bort-NPs) were developed and screened for optimal physiochemical properties, drug loading, and release profiles. Ald-Bort-NPs were tested for efficacy in mouse models of multiple myeloma (MM). Results demonstrated significantly enhanced survival and decreased tumor burden in mice pretreated with Ald-Bort-NPs versus Ald-Empty-NPs (no drug) or the free drug. We also observed that bortezomib, as a pretreatment regimen, modified the bone microenvironment and enhanced bone strength and volume. Our findings suggest that NP-based anticancer therapies with bone-targeting specificity comprise a clinically relevant method of drug delivery that can inhibit tumor progression in MM.


Assuntos
Antineoplásicos , Neoplasias Ósseas/tratamento farmacológico , Ácidos Borônicos , Sistemas de Liberação de Medicamentos , Ácido Láctico , Mieloma Múltiplo/tratamento farmacológico , Nanopartículas , Polietilenoglicóis , Ácido Poliglicólico , Pirazinas , Microambiente Tumoral/efeitos dos fármacos , Alendronato/química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Ácidos Borônicos/química , Ácidos Borônicos/farmacologia , Bortezomib , Linhagem Celular Tumoral , Xenoenxertos , Humanos , Ácido Láctico/síntese química , Ácido Láctico/química , Ácido Láctico/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Nanopartículas/química , Nanopartículas/ultraestrutura , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/metabolismo , Neoplasias Experimentais/patologia , Polietilenoglicóis/síntese química , Polietilenoglicóis/química , Polietilenoglicóis/farmacocinética , Ácido Poliglicólico/síntese química , Ácido Poliglicólico/química , Ácido Poliglicólico/farmacologia , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Pirazinas/química , Pirazinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Blood ; 124(12): 1905-14, 2014 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-25079359

RESUMO

The CXCR4/stromal cell-derived factor-1 (SDF-1) axis is essential for cell trafficking and has been shown to regulate tumor progression and metastasis in many tumors including multiple myeloma (MM). A second chemokine receptor for SDF-1, CXCR7 was discovered recently and found on activated endothelial cells. We examined the role of CXCR7 in angiogenic mononuclear cells (AMCs) trafficking in MM. Our data demonstrate that AMCs are circulating in patients with MM and in vivo studies show that they specifically home to areas of MM tumor growth. CXCR7 expression is important for regulating trafficking and homing of AMCs into areas of MM tumor growth and neoangiogenesis. We demonstrate that the CXCR7 inhibitor, POL6926, abrogated trafficking of AMCs to areas of MM tumor progression leading to a significant inhibition of tumor progression. These effects were through regulation of endothelial cells and not through a direct tumor effect, indicating that targeting a bone marrow microenvironmental cell can lead to a delay in MM tumor progression. In conclusion, our studies demonstrate that CXCR7 may play an important role in the regulation of tumor progression in MM through an indirect effect on the recruitment of AMCs to areas of MM tumor growth in the bone marrow niche.


Assuntos
Mieloma Múltiplo/etiologia , Mieloma Múltiplo/imunologia , Receptores CXCR/metabolismo , Animais , Materiais Biomiméticos/farmacologia , Linhagem Celular Tumoral , Progressão da Doença , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Mieloma Múltiplo/patologia , Células Neoplásicas Circulantes/imunologia , Células Neoplásicas Circulantes/patologia , Neovascularização Patológica , Plasmócitos/imunologia , Plasmócitos/patologia , Receptores CXCR/antagonistas & inibidores , Nicho de Células-Tronco/imunologia , Microambiente Tumoral/imunologia
9.
Blood ; 124(22): 3250-9, 2014 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-25205118

RESUMO

Clonal proliferation of plasma cells within the bone marrow (BM) affects local cells, such as mesenchymal stromal cells (MSCs), leading to osteolysis and fatality in multiple myeloma (MM). Consequently, there is an urgent need to find better mechanisms of inhibiting myeloma growth and osteolytic lesion development. To meet this need and accelerate clinical translation, better models of myeloma within the BM are required. Herein we have developed a clinically relevant, three-dimensional (3D) myeloma BM coculture model that mimics bone cell/cancer cell interactions within the bone microenvironment. The coculture model and clinical samples were used to investigate myeloma growth, osteogenesis inhibition, and myeloma-induced abnormalities in MM-MSCs. This platform demonstrated myeloma support of capillary-like assembly of endothelial cells and cell adhesion-mediated drug resistance (CAM-DR). Also, distinct normal donor (ND)- and MM-MSC miRNA (miR) signatures were identified and used to uncover osteogenic miRs of interest for osteoblast differentiation. More broadly, our 3D platform provides a simple, clinically relevant tool to model cancer growth within the bone-useful for investigating skeletal cancer biology, screening compounds, and exploring osteogenesis. Our identification and efficacy validation of novel bone anabolic miRs in MM opens more opportunities for novel approaches to cancer therapy via stromal miR modulation.


Assuntos
Células da Medula Óssea/patologia , Mieloma Múltiplo/patologia , Osteogênese/fisiologia , Cultura Primária de Células/métodos , Nicho de Células-Tronco , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Células Endoteliais da Veia Umbilical Humana/citologia , Humanos , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/fisiologia , Modelos Biológicos , Osteoblastos/citologia , Osteoblastos/fisiologia , Nicho de Células-Tronco/fisiologia , Alicerces Teciduais
10.
Blood ; 123(26): 4120-31, 2014 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-24711662

RESUMO

The C-X-C chemokine receptor type 4 (CXCR4) plays a crucial role in modulating cell trafficking in hematopoietic stem cells and clonal B cells. We screened 418 patients with B-cell lymphoproliferative disorders and described the presence of the C1013G/CXCR4 warts, hypogammaglobulinemia, infections, and myelokathexis-associated mutation in 28.2% (37/131) of patients with lymphoplasmacytic lymphoma (Waldenström macroglobulinemia [WM]), being either absent or present in only 7% of other B-cell lymphomas. In vivo functional characterization demonstrates its activating role in WM cells, as demonstrated by significant tumor proliferation and dissemination to extramedullary organs, leading to disease progression and decreased survival. The use of a monoclonal antibody anti-CXCR4 led to significant tumor reduction in a C1013G/CXCR4 WM model, whereas drug resistance was observed in mutated WM cells exposed to Bruton's tyrosine kinase, mammalian target of rapamycin, and phosphatidylinositol 3-kinase inhibitors, but not proteasome inhibitors. These findings demonstrate that C1013G/CXCR4 is an activating mutation in WM and support its role as a critical regulator of WM molecular pathogenesis and as an important therapeutic target.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Inibidores Enzimáticos/farmacologia , Mutação de Sentido Incorreto , Receptores CXCR4/metabolismo , Macroglobulinemia de Waldenstrom/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Intervalo Livre de Doença , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos SCID , Metástase Neoplásica , Transplante de Neoplasias , Receptores CXCR4/genética , Taxa de Sobrevida , Macroglobulinemia de Waldenstrom/tratamento farmacológico , Macroglobulinemia de Waldenstrom/genética , Macroglobulinemia de Waldenstrom/mortalidade , Macroglobulinemia de Waldenstrom/patologia
11.
Blood ; 124(17): 2675-86, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25217697

RESUMO

Proline-rich tyrosine kinase 2 (Pyk2) is a member of the focal adhesion kinase family that has been recently linked to tumor development. However, its role in modulating multiple myeloma (MM) biology and disease progression remains unexplored. We first demonstrated that patients with MM present with higher expression of Pyk2 compared with healthy individuals. By using loss-of-function approaches, we found that Pyk2 inhibition led to reduction of MM tumor growth in vivo as well as decreased cell proliferation, cell-cycle progression, and adhesion ability in vitro. In turn, overexpression of Pyk2 promoted the malignant phenotype, substantiated by enhanced tumor growth and reduced survival. Mechanistically, inhibition of Pyk2 reduced activation of Wnt/ß-catenin signaling by destabilizing ß-catenin, leading to downregulation of c-Myc and Cyclin D1. Furthermore, treatment of MM cells with the FAK/Pyk2 inhibitor VS-4718 effectively inhibited MM cell growth both in vitro and in vivo. Collectively, our findings describe the tumor-promoting role of Pyk2 in MM, thus providing molecular evidence for a novel tyrosine kinase inhibitor as a new therapeutic option in MM.


Assuntos
Aminopiridinas/farmacologia , Quinase 2 de Adesão Focal/antagonistas & inibidores , Mieloma Múltiplo/prevenção & controle , Inibidores de Proteínas Quinases/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Progressão da Doença , Feminino , Quinase 2 de Adesão Focal/genética , Quinase 2 de Adesão Focal/metabolismo , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Humanos , Immunoblotting , Medições Luminescentes , Camundongos SCID , Mieloma Múltiplo/genética , Mieloma Múltiplo/metabolismo , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise de Sobrevida , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo
12.
Blood ; 124(11): 1765-76, 2014 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-25061176

RESUMO

Glycosylation is a stepwise procedure of covalent attachment of oligosaccharide chains to proteins or lipids, and alterations in this process, especially increased sialylation, have been associated with malignant transformation and metastasis. The role of altered sialylation in multiple myeloma (MM) cell trafficking has not been previously investigated. In the present study we identified high expression of ß-galactoside α-2,3-sialyltransferase, ST3GAL6, in MM cell lines and patients. This gene plays a key role in selectin ligand synthesis in humans through the generation of functional sialyl Lewis X. In MRC IX patients, high expression of this gene is associated with inferior overall survival. In this study we demonstrate that knockdown of ST3GAL6 results in a significant reduction in levels of α-2,3-linked sialic acid on the surface of MM cells with an associated significant reduction in adhesion to MM bone marrow stromal cells and fibronectin along with reduced transendothelial migration in vitro. In support of our in vitro findings, we demonstrate significantly reduced homing and engraftment of ST3GAL6 knockdown MM cells to the bone marrow niche in vivo, along with decreased tumor burden and prolonged survival. This study points to the importance of altered glycosylation, particularly sialylation, in MM cell adhesion and migration.


Assuntos
Mieloma Múltiplo/enzimologia , Proteínas de Neoplasias/metabolismo , Sialiltransferases/metabolismo , Migração Transendotelial e Transepitelial , Animais , Células da Medula Óssea/enzimologia , Células da Medula Óssea/patologia , Adesão Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/genética , Feminino , Técnicas de Silenciamento de Genes , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos SCID , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Ácido N-Acetilneuramínico/biossíntese , Ácido N-Acetilneuramínico/genética , Proteínas de Neoplasias/genética , Sialiltransferases/genética , Células Estromais/enzimologia , Células Estromais/patologia , beta-Galactosídeo alfa-2,3-Sialiltransferase
13.
Biochim Biophys Acta ; 1846(1): 26-39, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24709008

RESUMO

Tumor-associated neovasculature is a critical therapeutic target; however, despite significant progress made in the clinical efficacy of anti-vessel drugs, the effect of these agents remains transient: over time, most patients develop resistance, which inevitably leads to tumor progression. To develop more effective treatments, it is imperative that we better understand the mechanisms involved in tumor vessel formation, how they participate to the tumor progression and metastasis, and the best way to target them. Several mechanisms contribute to the formation of tumor-associated vasculature: i) neoangiogenesis; ii) vascular co-option; iii) mosaicism; iv) vasculogenic mimicry, and v) postnatal vasculogenesis. These mechanisms can also play a role in the development of resistance to anti-angiogenic drugs, and could serve as targets for designing new anti-vascular molecules to treat solid as well as hematological malignancies. Bone marrow-derived endothelial progenitor cell (EPC)-mediated vasculogenesis represents an important new target, especially at the early stage of tumor growth (when EPCs are critical for promoting the "angiogenic switch"), and during metastasis, when EPCs promote the transition from micro- to macro-metastases. In hematologic malignancies, the EPC population could be related to the neoplastic clone, and both may share a common ontogeny. Thus, characterization of tumor-associated EPCs in blood cancers may provide clues for more specific anti-vascular therapy that has both direct and indirect anti-tumor effects. Here, we review the role of vasculogenesis, mediated by bone marrow-derived EPCs, in the progression of cancer, with a particular focus on the role of these cells in promoting progression of hematological malignancies.


Assuntos
Células Endoteliais/patologia , Neoplasias/irrigação sanguínea , Neoplasias/patologia , Neovascularização Patológica/patologia , Células-Tronco/patologia , Animais , Progressão da Doença , Neoplasias Hematológicas/patologia , Humanos
14.
Am J Hematol ; 89(11): 1030-6, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25060991

RESUMO

The phosphatidylinositol-3 kinase (PI3K) pathway is activated in multiple myeloma (MM) and Waldenstrom Macroglobulenima (WM), and plays a crucial role in tumor progression and drug resistance. In this study, we characterized the role of pan-class I PI3K inhibition on cell trafficking and survival of MM and WM cells. We tested the effect of pan-class I PI3K inhibition by siRNA silencing or pharmacologic inhibition with buparlisib on MM cell survival, apoptosis and cell cycle in vitro and tumor growth and mobilization of MM cells in vivo. We then evaluated buparlisib-dependent mechanisms of induced MM cell mobilization. Moreover, the effect of buparlisib on cell survival, apoptosis, and adhesion of WM cells to bone marrow stromal cells (BMSCs) has been evaluated. We showed that buparlisib induced toxicity in MM cells, supported by induction of apoptosis and cell cycle arrest. Buparlisib was also found to reduce tumor progression in vivo. Importantly, buparlisib enhanced MM cell mobilization in vivo which was driven by decreased adhesion of MM cells to BMSCs and increased chemotaxis via up-regulation of CXCR4 expression. Similar to its effects on MM cells, buparlisib also induced cell survival and apoptosis, and decreased adhesion in WM cells. These data highlight the critical contribution of class I PI3K signaling to the regulation of survival and cell dissemination in B-cell malignancies.


Assuntos
Aminopiridinas/farmacologia , Antineoplásicos/farmacologia , Morfolinas/farmacologia , Mieloma Múltiplo/patologia , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/farmacologia , Macroglobulinemia de Waldenstrom/patologia , Aminopiridinas/uso terapêutico , Animais , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Quimiotaxia/efeitos dos fármacos , Técnicas de Cocultura , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Fibronectinas , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos SCID , Morfolinas/uso terapêutico , Mieloma Múltiplo/enzimologia , Invasividade Neoplásica , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Interferência de RNA , RNA Interferente Pequeno/farmacologia , Receptores CXCR4/biossíntese , Receptores CXCR4/genética , Macroglobulinemia de Waldenstrom/enzimologia , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Int Immunol ; 24(8): 477-83, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22438420

RESUMO

The anti-CD20 chimeric monoclonal antibody rituximab mediates cytotoxicity in malignant B cells via multiple mechanisms, including complement-dependent cytotoxicity (CDC), antibody-dependent cellular cytotoxicity (ADCC) and direct induction of apoptosis. To optimize treatment of non-Hodgkin's lymphoma, a fuller understanding of these mechanisms and their relative contributions to clinical efficacy is required. Here, we report the characteristics of the mutual impact between ADCC and CDC, the two major effector functions through the Fc receptors. To compare ADCC induced under various conditions, we developed a highly reproducible method of estimating ADCC activity using immortalized effector cells. The set of the effector cells that we established was able to calculate net ADCC with high reproducibility by comparing the cytotoxicity of effector cells expressing exogeneous FcγRIIIa to those of mock effector cells. In addition, the different property of effector cells of two FcγRIIIa single-nucleotide polymorphisms (SNP) could be also evaluated in exactly identical background. ADCC assessment in the presence of human serum directly provided the evidence of the competitive interaction of ADCC and CDC. The inhibition of ADCC of effector cells having low affinity SNP of FcγRIIIa by active complement was more potent than those having high-affinity SNP at the rituximab-concentration comparable to the serum level obtained in patients. These findings could have a profound impact on optimization of the regimen of therapeutic antibodies and on the development of antibodies that will enhance effector function.


Assuntos
Anticorpos Monoclonais Murinos/farmacologia , Citotoxicidade Celular Dependente de Anticorpos/genética , Antineoplásicos/farmacologia , Ativação do Complemento/genética , Receptores de IgG/genética , Citotoxicidade Celular Dependente de Anticorpos/efeitos dos fármacos , Citotoxicidade Celular Dependente de Anticorpos/imunologia , Linhagem Celular , Ativação do Complemento/efeitos dos fármacos , Ativação do Complemento/imunologia , Citotoxicidade Imunológica/efeitos dos fármacos , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Humanos , Células Matadoras Naturais , Microscopia Confocal , Polimorfismo de Nucleotídeo Único , Rituximab , Transfecção
17.
Cancers (Basel) ; 14(3)2022 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-35158834

RESUMO

Nucleic acid medicines have been developed as new therapeutic agents against various diseases; however, targeted delivery of these reagents into cancer cells, particularly hematologic cancer cells, via systemic administration is limited by the lack of efficient and cell-specific delivery systems. We previously demonstrated that monoclonal antibody (mAb)-oligonucleotide complexes targeting exosomal microRNAs with linear oligo-D-arginine (Arg) linkers were transferred into solid cancer cells and inhibited exosomal miRNA functions. In this study, we developed exosome-capturing anti-CD63 mAb-conjugated small interfering RNAs (siRNAs) with branched Arg linkers and investigated their effects on multiple myeloma (MM) cells. Anti-CD63 mAb-conjugated siRNAs were successfully incorporated into MM cells. The incorporation of exosomes was inhibited by endocytosis inhibitors. We also conducted a functional analysis of anti-CD63 mAb-conjugated siRNAs. Ab-conjugated luciferase+ (luc+) siRNAs significantly decreased the luminescence intensity in OPM-2-luc+ cells. Moreover, treatment with anti-CD63 mAb-conjugated with MYC and CTNNB1 siRNAs decreased the mRNA transcript levels of MYC and CTNNB1 to 52.5% and 55.3%, respectively, in OPM-2 cells. In conclusion, exosome-capturing Ab-conjugated siRNAs with branched Arg linkers can be effectively delivered into MM cells via uptake of exosomes by parental cells. This technology has the potential to lead to a breakthrough in drug delivery systems for hematologic cancers.

18.
Cancer Sci ; 102(6): 1188-92, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21401804

RESUMO

The purpose of this study was to investigate the potential of circulating tumor cells (CTC) as a surrogate marker of the clinical outcome in metastatic colorectal cancer (mCRC) patients in order to identify Japanese patients responsive to oxaliplatin-based chemotherapy. Between January 2007 and April 2008, 64 patients with mCRC were enrolled in this prospective study. The treatment regimen was oxaliplatin-based chemotherapy. Collection of CTC from whole blood was performed at baseline and at 2 and 8-12 weeks after initiation of chemotherapy. Isolation and enumeration of CTC was performed using immunomagnetics. Patients with ≥3 CTC at baseline and at 2 and 8-12 weeks had a shorter median progression-free survival (8.5, 7.3 and 1.9 months, respectively) than those with <3 CTC (9.7, 10.4 and 9.1 months, respectively) (log-rank test: P = 0.047, P < 0.001 and P < 0.001, respectively). Patients with ≥3 CTC at 2 and 8-12 weeks had a shorter median overall survival (10.2 and 4.1 months, respectively) than those with <3 CTC (29.1 and 29.1 months, respectively) (P < 0.001 and P = 0.001, respectively). A spurious early rise in carcinoembryonic antigen level was observed in 11 patients showing a partial response. In contrast, no rise in early CTC level was observed among responders. Our data support the clinical utility of CTC enumeration in improving our ability to accurately assess treatment benefit and in expediting the identification of effective treatment regimens for individual Japanese patients.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Células Neoplásicas Circulantes/efeitos dos fármacos , Compostos Organoplatínicos/uso terapêutico , Adolescente , Adulto , Idoso , Antineoplásicos/administração & dosagem , Antígeno Carcinoembrionário/sangue , Neoplasias Colorretais/patologia , Progressão da Doença , Intervalo Livre de Doença , Feminino , Humanos , Japão , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Compostos Organoplatínicos/administração & dosagem , Oxaliplatina , Estudos Prospectivos , Resultado do Tratamento
19.
Cancer ; 117(17): 4026-32, 2011 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-21858803

RESUMO

BACKGROUND: Bevacizumab plus chemotherapy is a standard option in the treatment of metastatic colorectal cancer (mCRC). The aim of this study was to investigate the potential of circulating endothelial cell progenitors (CEPs) and phenotypical circulating endothelial cells (CECs) as surrogate markers of clinical outcome in mCRC patients to identify responders to bevacizumab in combination with chemotherapy. METHODS: A total of 69 patients with measurable mCRC were enrolled in this prospective study. Whole blood samples were analyzed before initiation of treatment and on days 4 and 14. Phenotypical CECs and CEPs were then isolated and enumerated by using flow cytometry. RESULTS: CEP levels of less than 0.04% on day 4 were significantly associated with longer progression-free survival (PFS) and overall survival (OS) (P < .001, P = .002, respectively) as compared with levels of 0.04% or more. In addition, CXCR4-positive CEC levels of less than 20% at baseline were significantly associated with longer PFS and OS as compared other indicators investigated (P < .001, P = .002, respectively). CONCLUSIONS: Levels of CEPs on day 4 and proportion of CXCR4-positive CECs at baseline were correlated with the prognosis of bevacizumab combination chemotherapy, suggesting that these surrogate markers may play a core role in the selection of candidates for bevacizumab treatment.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Células Endoteliais/metabolismo , Receptores CXCR4/metabolismo , Células-Tronco , Adulto , Idoso , Inibidores da Angiogênese/uso terapêutico , Anticorpos Monoclonais Humanizados , Bevacizumab , Biomarcadores Tumorais/sangue , Neoplasias Colorretais/sangue , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Intervalo Livre de Doença , Feminino , Fluoruracila/uso terapêutico , Humanos , Leucovorina/uso terapêutico , Masculino , Pessoa de Meia-Idade , Metástase Neoplásica , Compostos Organoplatínicos/uso terapêutico , Prognóstico , Resultado do Tratamento
20.
Int J Hematol ; 113(2): 279-284, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32864713

RESUMO

Somatic gene mutations related to acceleration disease and clonal evolution in multiple myeloma strongly influence severe clinical outcomes. In this study, we traced the transition of somatic mutations during the clinical course of myeloma patients over a long-term follow-up period (8.5 year average). Seven myeloma cases treated with immuno-chemotherapy at our institution were analyzed with clinical courses and the results of FISH and G-band analyses. Furthermore, the target sequences in regard to 121 genes, related to driver mutations or acceleration of disease in myeloma, were performed using bone marrow myeloma samples by next-generation sequencing, Ion Proton™ System. We detected a relationship between an increase in the dominant mutated gene (e.g., TP53, DIS3, FAM46C, KDM6B, and EGR1) and poor prognosis. In particular, clonal escalation of the TP53 mutation could not be overcome by any treatment. The selection of a combination treatment conducted in conjunction with the monitoring of gene mutations is appropriate for long-term survival. Our data demonstrate that long-term follow-up of somatic gene mutations during the clinical course of myeloma is helpful in the development of an effective treatment strategy.


Assuntos
Evolução Clonal , Suscetibilidade a Doenças , Mieloma Múltiplo/diagnóstico , Mieloma Múltiplo/etiologia , Idoso , Biomarcadores Tumorais , Transformação Celular Neoplásica , Evolução Clonal/genética , Terapia Combinada/efeitos adversos , Terapia Combinada/métodos , Biologia Computacional/métodos , Bases de Dados Genéticas , Progressão da Doença , Feminino , Seguimentos , Predisposição Genética para Doença , Testes Genéticos , Variação Genética , Humanos , Pessoa de Meia-Idade , Mieloma Múltiplo/mortalidade , Mieloma Múltiplo/terapia , Mutação , Estadiamento de Neoplasias , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA