Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Clin Cancer Res ; 30(10): 2181-2192, 2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38437671

RESUMO

PURPOSE: FGFR2 and FGFR3 show oncogenic activation in many cancer types, often through chromosomal fusion or extracellular domain mutation. FGFR2 and FGFR3 alterations are most prevalent in intrahepatic cholangiocarcinoma (ICC) and bladder cancers, respectively, and multiple selective reversible and covalent pan-FGFR tyrosine kinase inhibitors (TKI) have been approved in these contexts. However, resistance, often due to acquired secondary mutations in the FGFR2/3 kinase domain, limits efficacy. Resistance is typically polyclonal, involving a spectrum of different mutations that most frequently affect the molecular brake and gatekeeper residues (N550 and V565 in FGFR2). EXPERIMENTAL DESIGN: Here, we characterize the activity of the next-generation covalent FGFR inhibitor, KIN-3248, in preclinical models of FGFR2 fusion+ ICC harboring a series of secondary kinase domain mutations, in vitro and in vivo. We also test select FGFR3 alleles in bladder cancer models. RESULTS: KIN-3248 exhibits potent selectivity for FGFR1-3 and retains activity against various FGFR2 kinase domain mutations, in addition to being effective against FGFR3 V555M and N540K mutations. Notably, KIN-3248 activity extends to the FGFR2 V565F gatekeeper mutation, which causes profound resistance to currently approved FGFR inhibitors. Combination treatment with EGFR or MEK inhibitors potentiates KIN-3248 efficacy in vivo, including in models harboring FGFR2 kinase domain mutations. CONCLUSIONS: Thus, KIN-3248 is a novel FGFR1-4 inhibitor whose distinct activity profile against FGFR kinase domain mutations highlights its potential for the treatment of ICC and other FGFR-driven cancers.


Assuntos
Mutação , Inibidores de Proteínas Quinases , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Neoplasias da Bexiga Urinária , Ensaios Antitumorais Modelo de Xenoenxerto , Humanos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Animais , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Camundongos , Linhagem Celular Tumoral , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/genética , Neoplasias da Bexiga Urinária/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Colangiocarcinoma/tratamento farmacológico , Colangiocarcinoma/genética , Colangiocarcinoma/patologia , Proliferação de Células/efeitos dos fármacos
2.
J Med Chem ; 67(3): 1734-1746, 2024 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-38267212

RESUMO

Fibroblast growth factor receptor (FGFR) alterations are present as oncogenic drivers and bypass mechanisms in many forms of cancer. These alterations can include fusions, amplifications, rearrangements, and mutations. Acquired drug resistance to current FGFR inhibitors often results in disease progression and unfavorable outcomes for patients. Genomic profiling of tumors refractory to current FGFR inhibitors in the clinic has revealed several acquired driver alterations that could be the target of next generation therapeutics. Herein, we describe how structure-based drug design (SBDD) was used to enable the discovery of the potent and kinome selective pan-FGFR inhibitor KIN-3248, which is active against many acquired resistance mutations. KIN-3248 is currently in phase I clinical development for the treatment of advanced tumors harboring FGFR2 and/or FGFR3 gene alterations.


Assuntos
Neoplasias , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Humanos , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Neoplasias/tratamento farmacológico , Neoplasias/genética , Mutação , Progressão da Doença , Inibidores de Proteínas Quinases/efeitos adversos , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos
3.
J Midlife Health ; 14(3): 191-195, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38312772

RESUMO

Background: Menopausal transition phase is a difficult time in a woman's life. Many factors such as age, socioeconomic status, education, ethnic cultural, and body physique determine the presence of menopausal symptoms. This study helps us to understand the severity and perseverance of menopausal symptoms in women of this locality. Aims and Objectives: The aim of the study was to find distribution and severity of menopausal symptoms by self-rated Menopause Rating Scale (MRS) through different transition phases of menopause in women aged between 40 and 60 years. Methodology: It is a hospital-based observational study. All the participants answered an 11-item MRS questionnaire. Results: Out of 300 participants, 106 belonged to premenopausal state, 111 to perimenopausal state, and 83 were postmenopause. Overall, 47% of participants had one or the other menopausal symptoms. Most of them had mild-to-moderate symptoms. Only 3% had severe symptoms. Somatic subscale was the maximum reported symptoms in our study group. The most common symptom was physical and mental exhaustion (55%) and the least common was sexual problems (8%). Difficulty in sleeping and bladder symptoms were more and statistically significant in postmenopausal group where as hot flushes and irritability were more common in perimenopausal group. Conclusion: Somatic subscale symptoms are more common than urogenital or psychosocial subscales. Postmenopause women manifested higher symptoms than premenopause or perimenopause group women and most were mild to moderate in severity in women visiting our hospital.

4.
Stem Cell Reports ; 6(5): 660-667, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-27117783

RESUMO

Contact with the extracellular matrix is essential for maintenance of epithelial cells in many tissues, while in its absence epithelial cells can detach and undergo anoikis. Here, we show that anoikis of luminal cells in the prostate epithelium is followed by a program of tissue repair that is mediated in part by differentiation of basal epithelial cells to luminal cells. We describe a mouse model in which inducible deletion of E-cadherin in prostate luminal cells results in their apoptotic cell death by anoikis, in the absence of phenotypic effects in the surrounding stroma. Quantitative assessments of proliferation and cell death in the luminal and basal compartments indicate that basal cells can rapidly generate luminal cells. Thus, our findings identify a role for basal-to-luminal differentiation in prostate epithelial repair, and provide a normal context to analogous processes that may occur during prostate cancer initiation.


Assuntos
Anoikis/genética , Caderinas/genética , Animais , Morte Celular/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Células Epiteliais/metabolismo , Humanos , Masculino , Camundongos , Próstata/crescimento & desenvolvimento , Próstata/metabolismo , Células-Tronco/metabolismo
5.
Mol Cell Biol ; 29(23): 6268-82, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19786571

RESUMO

In the 1970s, several human retinoblastoma cell lines were developed from cultures of primary tumors. As the human retinoblastoma cell lines were established in culture, growth properties and changes in cell adhesion were described. Those changes correlated with the ability of the human retinoblastoma cell lines to invade the optic nerve and metastasize in orthotopic xenograft studies. However, the mechanisms that underlie these changes were not determined. We used the recently developed knockout mouse models of retinoblastoma to begin to characterize the molecular, cellular, and genetic changes associated with retinoblastoma tumor progression and optic nerve invasion. Here we report the isolation and characterization of the first mouse retinoblastoma cell lines with targeted deletions of the Rb family. Our detailed analysis of these cells as they were propagated in culture from the primary tumor shows that changes in cadherin-mediated cell adhesion are associated with retinoblastoma invasion of the optic nerve prior to metastasis. In addition, the same changes in cadherin-mediated cell adhesion correlate with the invasive properties of the human retinoblastoma cell lines isolated decades ago, providing a molecular mechanism for these earlier observations. Most importantly, our studies are in agreement with genetic studies on human retinoblastomas, suggesting that changes in this pathway are involved in tumor progression.


Assuntos
Nervo Óptico/patologia , Neoplasias da Retina/patologia , Retinoblastoma/patologia , Animais , Caderinas/genética , Adesão Celular , Linhagem Celular , Progressão da Doença , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Invasividade Neoplásica , Nervo Óptico/metabolismo , Neoplasias da Retina/genética , Retinoblastoma/genética , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
6.
J Pediatr Hematol Oncol ; 31(1): 8-13, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19125079

RESUMO

Recent studies have shown the presence of human papillomavirus (HPV) genome in retinoblastoma (RB) tumor samples. There is no information on the HPV status in the RB tumors of Indian patients. We studied the presence of HPV genome in RB tumor samples from patients with unilateral tumor. Forty-four fresh RB tumor samples and 30 non-neoplastic donor retinas were analyzed for the presence of HPV 16 and 18 genome by nested and seminested polymerase chain reaction. Tumor tissue sections were also used to assess the expression of the retinoblastoma (Rb) protein. All 30 control tissues were negative for HPV genome. Among the 44 tumor samples, there were 23 tumors with invasion of optic nerve/choroid and 21 tumors with no invasion. HPV DNA was present in 21/44 (47%) RB tumors. Among 21 unilateral RB tumors that were positive for HPV DNA, HPV 16 was detected in 12/21 (57%) tumors. However, HPV 18 was negative in all the tumors. Rb protein was absent in 16 (71%) of 21 tumors that had HPV DNA. However, Rb was also absent in 20 (86%) of 23 tumors that were HPV negative. Children younger than 18 months old were significantly associated with the presence of HPV DNA compared with children above 24 months old (P<0.014). Our study shows the presence of HPV and HPV 16 in a subset of RB tumor samples. However, further studies are in progress to know the role played by HPV in RB.


Assuntos
DNA Viral/análise , Infecções Oculares Virais/virologia , Papillomavirus Humano 16/genética , Papillomavirus Humano 18/genética , Infecções por Papillomavirus/virologia , Neoplasias da Retina/virologia , Retinoblastoma/virologia , Criança , Pré-Escolar , Infecções Oculares Virais/diagnóstico , Feminino , Humanos , Técnicas Imunoenzimáticas , Lactente , Masculino , Infecções por Papillomavirus/diagnóstico , Reação em Cadeia da Polimerase , Neoplasias da Retina/diagnóstico , Retinoblastoma/diagnóstico , Proteína do Retinoblastoma/metabolismo , Sensibilidade e Especificidade
7.
Pediatr Blood Cancer ; 50(2): 402-6, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17091485

RESUMO

Chemotherapy is an essential modality in the treatment of retinoblastoma (RB). Mammalian serine/arginine-rich protein-specific kinase 1 (SRPK1) is a cisplatin-sensitivity-related protein and its downregulation is known to be associated with decreased response to cisplatin and carboplatin. We investigated the expression of SRPK1 in 63 archival RB and correlated its expression with pathologic staging and exposure to chemotherapy. The majority of the RB (62/63) were advanced stage (Groups D and E) with intermediate to high risk of treatment failure according to the new international classification for intraocular RB and SRPK1 was reduced in 32/62 (51%) tumors. SRPK1 protein expression was reduced in (100%) 8/8 RB that had recurred in the orbit or had metastasized. SRPK1 protein expression is reduced in RB with advanced stage of presentation and this may add to drug resistance mechanisms in RB.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Proteínas Serina-Treonina Quinases/biossíntese , Neoplasias da Retina/enzimologia , Retinoblastoma/enzimologia , Adolescente , Criança , Pré-Escolar , Feminino , Humanos , Imuno-Histoquímica , Lactente , Masculino , Estadiamento de Neoplasias , Neoplasias da Retina/tratamento farmacológico , Neoplasias da Retina/patologia , Retinoblastoma/tratamento farmacológico , Retinoblastoma/patologia
8.
Exp Eye Res ; 84(4): 781-9, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17316610

RESUMO

In our earlier study we showed that invasive retinoblastoma (RB) had down regulated tetraspanin protein KAI1/CD82, a family of cell surface glycoprotein. KAI1 may link to the cell surface molecules, such as integrins, E-cadherin, and other TM4SF members, and loss of KAI1 function may have a significant role in the progression of retinoblastoma. We also showed that epithelial cell adhesion molecule (EpCAM) is overexpressed in invasive RB. EpCAM expression decreases adhesion mediated by cadherins. Thus, we were further interested in studying the role of other adhesion molecules like cadherins and catenins in RB. We studied the expression of Motility-Related Protein 1 (MRP-1)/CD9, E-cadherin, N-cadherin, alpha-catenin and beta-catenin in RB and correlated clinicopathologically in 62 archival paraffin-embedded tumors by immunohistochemistry. There were 29 tumors with no invasion of choroids/optic nerve and 33 tumors with invasion of choroid/optic nerve/orbit. Western blotting was performed on 20 tumors using the same antibodies. We observed higher expression of CD9 (P<0.001), E-cadherin (P<0.001) and alpha-catenin (P<0.001) in the non-invasive RB and higher expression of N-cadherin (P<0.001) in invasive RB. The expression of beta-catenin was not significantly different between two groups of tumors. In Western blotting, we were able to see CD9 and E-cadherin expression in a minority of tumors while N-cadherin, alpha-catenin and beta-catenin were expressed with differing intensities in a majority of tumors. Thus, invasive tumors expressed increased N-cadherin, alpha-catenin and decreased E-cadherin and CD9. Thus, it appears that loss of E-cadherin and gain of N-cadherin expression are features of invasiveness. Further functional studies are required to evaluate the role of beta-catenin in RB.


Assuntos
Antígenos CD/análise , Caderinas/análise , Cateninas/análise , Proteínas do Olho/análise , Glicoproteínas de Membrana/análise , Neoplasias da Retina/química , Retinoblastoma/química , Diferenciação Celular , Criança , Pré-Escolar , Olho/química , Feminino , Humanos , Immunoblotting/métodos , Imuno-Histoquímica/métodos , Lactente , Recém-Nascido , Masculino , Invasividade Neoplásica , Tetraspanina 29 , alfa Catenina/análise , beta Catenina/análise
9.
Nature ; 444(7115): 61-6, 2006 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-17080083

RESUMO

Most human tumours have genetic mutations in their Rb and p53 pathways, but retinoblastoma is thought to be an exception. Studies suggest that retinoblastomas, which initiate with mutations in the gene retinoblastoma 1 (RB1), bypass the p53 pathway because they arise from intrinsically death-resistant cells during retinal development. In contrast to this prevailing theory, here we show that the tumour surveillance pathway mediated by Arf, MDM2, MDMX and p53 is activated after loss of RB1 during retinogenesis. RB1-deficient retinoblasts undergo p53-mediated apoptosis and exit the cell cycle. Subsequently, amplification of the MDMX gene and increased expression of MDMX protein are strongly selected for during tumour progression as a mechanism to suppress the p53 response in RB1-deficient retinal cells. Our data provide evidence that the p53 pathway is inactivated in retinoblastoma and that this cancer does not originate from intrinsically death-resistant cells as previously thought. In addition, they support the idea that MDMX is a specific chemotherapeutic target for treating retinoblastoma.


Assuntos
Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Retinoblastoma/metabolismo , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas de Ciclo Celular , Morte Celular , Divisão Celular , Dano ao DNA , Amplificação de Genes/genética , Humanos , Imidazóis/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/genética , Piperazinas/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Ratos , Ratos Sprague-Dawley , Retina/metabolismo , Retinoblastoma/genética , Retinoblastoma/patologia , Proteína do Retinoblastoma/deficiência , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Proteína Supressora de Tumor p14ARF/metabolismo
10.
Exp Eye Res ; 83(4): 736-40, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16716300

RESUMO

We examine the immunoreactivity of the non-receptor tyrosine kinase, c-Src kinase and its downstream molecule, signal transducer and activator of transcription 3 (pStat3) in retinoblastoma (RB), and correlation with invasiveness and differentiation. Tumor samples from 40 patients with RB were available for the study. There were 18 tumors in group 1 (non-invasive) and 22 tumors in group 2 (invasive). The immunoreactivity of c-Src kinase and pStat3 was compared in the two groups of tumors. Group 1 (non-invasive) RB showed intermediate c-Src kinase immunoreactivity (Allred score 4-5) in 14/18 tumors and low immunoreactivity (Allred score 2-3) in 4/18 tumors. pStat3 was intermediate (Allred score 4-5) in 6/18 tumors and negative (Allred score 0) in 12/18 tumors. Group 2 (invasive) RB showed high c-Src kinase immunoreactivity (Allred score 6-8) in 22/22 tumors and high pStat3 (Allred score 6-8) in 19/22 tumors. The expression of c-Src kinase (P<0.001) and pStat3 (P<0.001) was significantly higher in group 2 RB. Src kinase expression (P<0.05) and pStat3 expression (P<0.05) was higher in the poorly differentiated tumors compared to moderately- and well-differentiated tumors. The increased expression of c-Src kinase and pStat3 expression could play a role in the invasiveness of group 2 tumors. Further characterization of the pathways involved in the pathogenesis of RB will shed light on fundamental mechanisms of tumorigenesis.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Retina/metabolismo , Retinoblastoma/metabolismo , Fator de Transcrição STAT3/metabolismo , Quinases da Família src/metabolismo , Diferenciação Celular , Criança , Pré-Escolar , Feminino , Humanos , Lactente , Masculino , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Neoplasias da Retina/patologia , Retinoblastoma/patologia
11.
Invest Ophthalmol Vis Sci ; 45(12): 4247-50, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15557427

RESUMO

PURPOSE: This study was conducted to investigate the potential of targeting epithelial cell adhesion molecules (EpCAMs) in the treatment of retinoblastoma. It was first determined whether EpCAM is expressed in retinoblastoma and then whether EpCAM reactivity correlates with tumor aggressiveness. METHODS: EpCAM reactivity was evaluated by immunohistochemistry in 43 retinoblastoma specimens from 43 patients, by using the monoclonal antibody GA733.2. The tumors were divided into two groups. There were 20 tumors with no invasion of the choroid and optic nerve (group A) and 23 tumors with invasion of the choroid, optic nerve, and orbit (group B). EpCAM reactivity was correlated with invasion and differentiation of the tumors. RESULTS: Among the 43 tumors, EpCAM reactivity was observed in 100% (43/43) tumors. EpCAM reactivity was significantly higher in the invasive than the noninvasive tumors (P < 0.05) and in poorly differentiated than in well-differentiated tumors (P < 0.005). Non-neoplastic retina also expressed EpCAM. CONCLUSIONS: The results confirm that EpCAM is vastly expressed in retinoblastoma and point to its use as a target for therapy in the future.


Assuntos
Antígenos de Neoplasias/metabolismo , Moléculas de Adesão Celular/metabolismo , Neoplasias da Retina/metabolismo , Retinoblastoma/metabolismo , Adulto , Diferenciação Celular , Criança , Pré-Escolar , Molécula de Adesão da Célula Epitelial , Feminino , Humanos , Imuno-Histoquímica , Lactente , Masculino , Invasividade Neoplásica , Retina/metabolismo , Neoplasias da Retina/patologia , Neoplasias da Retina/terapia , Retinoblastoma/patologia , Retinoblastoma/terapia
12.
Cancer ; 101(7): 1672-6, 2004 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15378505

RESUMO

BACKGROUND: The importance of the Fas-Fas ligand (FasL) mechanism for the immune evasion by tumors provided a strong rationale for the examination of FasL expression in retinoblastoma. In an earlier publication, the authors reported that invasive retinoblastomas decreased Fas expression. Because to the authors' knowledge there is not much information regarding the effect of FasL expression on retinoblastoma, the authors studied the expression of FasL in retinoblastoma and correlated it with invasiveness. METHODS: Thirty-six archival retinoblastoma specimens were divided into 2 groups. Group A (n = 17) was comprised of specimens from tumors with no invasion and Group B (n = 19) was comprised of specimens from tumors with invasion of the choroid (focal, diffuse), optic nerve (laminar, postlaminar, surgical end), and orbit. Sections were immunostained with a monoclonal antibody to FasL and the immunoreactivity was assessed. RESULTS: In Group A, FasL was negative in 100% (17 of 17) of the tumor specimens. In Group B, FasL was expressed in 79% (15 of 19) of the tumor specimens (positive in 9 tumors and heterogeneous in 6 tumors). The difference in FasL expression between the two groups was significant (P < 0.001) CONCLUSIONS: Increased expression of FasL was observed in specimens taken from patients with aggressive tumors. Thus, Loss of Fas and gain of aberrant FasL expression were common features of malignant transformation. The data suggested that the Fas/FasL pathway is potentially immunosuppressive and may be involved in the escape of retinoblastoma cells from immune destruction.


Assuntos
Glicoproteínas de Membrana/análise , Neoplasias da Retina/química , Retinoblastoma/química , Adolescente , Transformação Celular Neoplásica , Criança , Pré-Escolar , Proteína Ligante Fas , Feminino , Humanos , Imuno-Histoquímica , Lactente , Masculino , Invasividade Neoplásica , Neoplasias da Retina/imunologia , Neoplasias da Retina/patologia , Retinoblastoma/imunologia , Retinoblastoma/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...