Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Bioorg Med Chem Lett ; 28(20): 3404-3408, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30217415

RESUMO

Small molecule inhibitors of the p53-MDM2 protein complex are under intense investigation in clinical trials as anti-cancer agents, including our first generation inhibitor NVP-CGM097. We recently described the rational design of a novel pyrazolopyrrolidinone core as a new lead structure and now we report on the synthesis and optimization of this to provide a highly potent lead compound. This new compound displayed excellent oral efficacy in our preclinical mechanistic in vivo model and marked a significant milestone towards the identification of our second generation clinical candidate NVP-HDM201.


Assuntos
Antineoplásicos/farmacologia , Multimerização Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Pirazóis/farmacologia , Pirrolidinonas/farmacologia , Proteína Supressora de Tumor p53/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Antineoplásicos/química , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Cães , Haplorrinos , Humanos , Masculino , Camundongos , Microssomos Hepáticos/metabolismo , Pirazóis/síntese química , Pirazóis/química , Pirazóis/farmacocinética , Pirrolidinonas/síntese química , Pirrolidinonas/química , Pirrolidinonas/farmacocinética , Ratos Sprague-Dawley , Estereoisomerismo
2.
Bioorg Med Chem Lett ; 26(19): 4837-4841, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27542305

RESUMO

The p53-MDM2 interaction is an anticancer drug target under investigation in the clinic. Our compound NVP-CGM097 is one of the small molecule inhibitors of this protein-protein interaction currently evaluated in cancer patients. As part of our effort to identify new classes of p53-MDM2 inhibitors that could lead to additional clinical candidates, we report here the design of highly potent inhibitors having a pyrazolopyrrolidinone core structure. The conception of these new inhibitors originated in a consideration on the MDM2 bound conformation of the dihydroisoquinolinone class of inhibitors to which NVP-CGM097 belongs. This work forms the foundation of the discovery of HDM201, a second generation p53-MDM2 inhibitor that recently entered phase I clinical trial.


Assuntos
Descoberta de Drogas , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteína Supressora de Tumor p53/antagonistas & inibidores , Cristalografia por Raios X , Transferência Ressonante de Energia de Fluorescência , Conformação Molecular , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo
3.
Bioorg Med Chem Lett ; 25(17): 3621-5, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26141769

RESUMO

Blocking the interaction between the p53 tumor suppressor and its regulatory protein MDM2 is a promising therapeutic concept under current investigation in oncology drug research. We report here the discovery of the first representatives of a new class of small molecule inhibitors of this protein-protein interaction: the dihydroisoquinolinones. Starting from an initial hit identified by virtual screening, a derivatization program has resulted in compound 11, a low nanomolar inhibitor of the p53-MDM2 interaction showing significant cellular activity. Initially based on a binding mode hypothesis, this effort was then guided by a X-ray co-crystal structure of MDM2 in complex with one of the synthesized analogs. The X-ray structure revealed an unprecedented binding mode for p53-MDM2 inhibitors.


Assuntos
Isoquinolinas/química , Isoquinolinas/farmacologia , Mapas de Interação de Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Cristalografia por Raios X , Humanos , Simulação de Acoplamento Molecular , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Relação Estrutura-Atividade , Proteína Supressora de Tumor p53/antagonistas & inibidores
4.
Cancer Cell ; 5(3): 231-9, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15050915

RESUMO

IGF-IR-mediated signaling promotes survival, anchorage-independent growth, and oncogenic transformation, as well as tumor growth and metastasis formation in vivo. NVP-AEW541 is a pyrrolo[2,3-d]pyrimidine derivative small molecular weight kinase inhibitor of the IGF-IR, capable of distinguishing between the IGF-IR (IC50 = 0.086 microM) and the closely related InsR (IC50 = 2.3 microM) in cells. As expected for a specific IGF-IR kinase inhibitor, NVP-AEW541 abrogates IGF-I-mediated survival and colony formation in soft agar at concentrations that are consistent with inhibition of IGF-IR autophosphorylation. In vivo, this orally bioavailable compound inhibits IGF-IR signaling in tumor xenografts and significantly reduces the growth of IGF-IR-driven fibrosarcomas. Thus, NVP-AEW541 represents a class of selective, small molecule IGF-IR kinase inhibitors with proven in vivo antitumor activity and potential therapeutic application.


Assuntos
Antineoplásicos/farmacologia , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais/fisiologia , Células 3T3 , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Divisão Celular , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Humanos , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Camundongos , Fosforilação/efeitos dos fármacos , Receptor IGF Tipo 1/efeitos dos fármacos , Receptor de Insulina/efeitos dos fármacos , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/efeitos dos fármacos , Quinases da Família src/metabolismo
5.
RSC Chem Biol ; 2(6): 1661-1668, 2021 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-34977581

RESUMO

Mouse double minute 2 homolog (MDM2, Hdm2) is an important negative regulator of the tumor suppressor p53. Using a mRNA based display technique to screen a library of >1012 in vitro-translated cyclic peptides, we have identified a macrocyclic ligand that shows picomolar potency on MDM2. X-Ray crystallography reveals a novel binding mode utilizing a unique pharmacophore to occupy the Phe/Trp/Leu pockets on MDM2. Conjugation of a cyclic cell-penetrating peptide (cCPP) to the initially non cell-permeable ligand enables cellular uptake and a pharmacodynamic response in SJSA-1 cells. The demonstrated enhanced intracellular availability of cyclic peptides that are identified by a display technology exemplifies a process for the application of intracellular tools for drug discovery projects.

6.
Cancer Res ; 78(21): 6257-6267, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30135191

RESUMO

Activation of p53 by inhibitors of the p53-MDM2 interaction is being pursued as a therapeutic strategy in p53 wild-type cancers. Here, we report distinct mechanisms by which the novel, potent, and selective inhibitor of the p53-MDM2 interaction HDM201 elicits therapeutic efficacy when applied at various doses and schedules. Continuous exposure of HDM201 led to induction of p21 and delayed accumulation of apoptotic cells. By comparison, high-dose pulses of HDM201 were associated with marked induction of PUMA and a rapid onset of apoptosis. shRNA screens identified PUMA as a mediator of the p53 response specifically in the pulsed regimen. Consistent with this, the single high-dose HDM201 regimen resulted in rapid and marked induction of PUMA expression and apoptosis together with downregulation of Bcl-xL in vivo Knockdown of Bcl-xL was identified as the top sensitizer to HDM201 in vitro, and Bcl-xL was enriched in relapsing tumors from mice treated with intermittent high doses of HDM201. These findings define a regimen-dependent mechanism by which disruption of MDM2-p53 elicits therapeutic efficacy when given with infrequent dosing. In an ongoing HDM201 trial, the observed exposure-response relationship indicates that the molecular mechanism elicited by pulse dosing is likely reproducible in patients. These data support the clinical comparison of daily and intermittent regimens of p53-MDM2 inhibitors.Significance: Pulsed high doses versus sustained low doses of the p53-MDM2 inhibitor HDM201 elicit a proapoptotic response from wild-type p53 cancer cells, offering guidance to current clinical trials with this and other drugs that exploit the activity of p53. Cancer Res; 78(21); 6257-67. ©2018 AACR.


Assuntos
Antineoplásicos/administração & dosagem , Imidazóis/administração & dosagem , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Pirimidinas/administração & dosagem , Pirróis/administração & dosagem , Proteína Supressora de Tumor p53/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Apoptose , Área Sob a Curva , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imidazóis/farmacologia , Estimativa de Kaplan-Meier , Dose Máxima Tolerável , Camundongos , Transplante de Neoplasias , Pirimidinas/farmacologia , Pirróis/farmacologia , RNA Interferente Pequeno/metabolismo , Fatores de Tempo , Proteína bcl-X/metabolismo
7.
Br J Pharmacol ; 174(16): 2613-2622, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28436014

RESUMO

BACKGROUND AND PURPOSE: Helix stapling enhances the activity of peptides that interact with a target protein in a helical conformation. These staples are also supposed to change the pharmacokinetics of the molecules and promote cytoplasmic targeting. We assessed the extent to which the pharmacokinetic characteristics are a function of the staple for a peptide inhibiting the interaction of p53 with the human double minute 2 (Hdm2) protein and differ from those of the standard cationic cell-penetrating peptide nona-arginine. EXPERIMENTAL APPROACH: Stapled peptides and linear counterparts were synthesized in free and fluorescently labelled forms. Activity was determined in biochemical time-resolved Förster resonance energy transfer experiments and cellular high-content assays. Cellular uptake and intracellular trafficking were visualized by confocal microscopy. KEY RESULTS: Peptides showed sub-nanomolar potency. For short-time incubation, uptake efficiencies for the stapled and linear peptides were similar and both were taken up less efficiently than nona-arginine. Only for SJSA-1 cells expressing the Hdm2 target protein, the stapled peptides showed an enhanced cytoplasmic and nuclear accumulation after long-term incubation. This was also observed for the linear counterparts, albeit to a lesser degree. For HeLa cells, which lack target expression, no such accumulation was observed. CONCLUSION AND IMPLICATIONS: Cytosolic and nuclear accumulation was not an intrinsic property of the stapled peptide, but resulted from capture by the target Hdm2 after endo-lysosomal release. Considering the rather poor uptake of stapled peptides, further development should focus on increasing the efficiency of uptake of these peptides.


Assuntos
Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Células HEK293 , Humanos
8.
Pharmacol Ther ; 93(2-3): 79-98, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12191602

RESUMO

Many components of mitogenic signaling pathways in normal and neoplastic cells have been identified, including the large family of protein kinases, which function as components of signal transduction pathways, playing a central role in diverse biological processes, such as control of cell growth, metabolism, differentiation, and apoptosis. The development of selective protein kinase inhibitors that can block or modulate diseases caused by abnormalities in these signaling pathways is widely considered a promising approach for drug development. Because of their deregulation in human cancers, protein kinases, such as Bcr-Abl, those in the epidermal growth factor-receptor (HER) family, the cell cycle regulating kinases such as the cyclin-dependent kinases, as well as the vascular endothelial growth factor-receptor kinases involved in the neo-vascularization of tumors, are among the protein kinases considered as prime targets for the development of selective inhibitors. These drug-discovery efforts have generated inhibitors and low-molecular weight therapeutics directed against the ATP-binding site of various protein kinases that are in various stages of development (up to Phase II/III clinical trials). Three examples of inhibitors of protein kinases are reviewed, including low-molecular weight compounds targeting the cell cycle kinases; a potent and selective inhibitor of the HER1/HER2 receptor tyrosine kinase, the pyrollopyrimidine PKI166; and the 2-phenyl-aminopyrimidine STI571 (Glivec(R), Gleevec) a targeted drug therapy directed toward Bcr-Abl, the key player in chronic leukemia (CML). Some members of the HER family of receptor tyrosine kinases, in particular HER1 and HER2, have been found to be overexpressed in a variety of human tumors, suggesting that inhibition of HER signaling would be a viable antiproliferative strategy. The pyrrolo-pyrimidine PKI166 was developed as an HER1/HER2 inhibitor with potent in vitro antiproliferative and in vivo antitumor activity. Based upon its clear association with disease, the Bcr-Abl tyrosine kinase in CML represents the ideal target to validate the clinical utility of protein kinase inhibitors as therapeutic agents. In a preclinical model, STI571 (Glivec(R), Gleevec) showed potent in vitro and in vivo antitumor activity that was selective for Abl, c-Kit, and the platelet-derived growth factor-receptor. Phase I/II studies demonstrated that STI571 is well tolerated, and that it showed promising hematological and cytogenetic responses in CML and clinical responses in the c-Kit-driven gastrointestinal tumors.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/tratamento farmacológico , Proteínas Quinases , Transdução de Sinais/efeitos dos fármacos , Animais , Antineoplásicos/uso terapêutico , Benzamidas , Ciclo Celular/efeitos dos fármacos , Ensaios Clínicos como Assunto , Humanos , Mesilato de Imatinib , Camundongos , Piperazinas , Inibidores de Proteínas Quinases , Proteínas Quinases/metabolismo , Proteínas Quinases/fisiologia , Pirimidinas/uso terapêutico
9.
F1000Res ; 4: 42, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-28357033

RESUMO

We previously described a population of lymphoid progenitor cells (LPCs) in canine B-cell lymphoma defined by retention of the early progenitor markers CD34 and CD117 and "slow proliferation" molecular signatures that persist in the xenotransplantation setting. We examined whether valspodar, a selective inhibitor of the ATP binding cassette B1 transporter (ABCB1, a.k.a., p-glycoprotein/multidrug resistance protein-1) used in the neoadjuvant setting would sensitize LPCs to doxorubicin and extend the length of remission in dogs with therapy naïve large B-cell lymphoma. Twenty dogs were enrolled into a double-blinded, placebo controlled study where experimental and control groups received oral valspodar (7.5 mg/kg) or placebo, respectively, twice daily for five days followed by five treatments with doxorubicin 21 days apart with a reduction in the first dose to mitigate the potential side effects of ABCB1 inhibition. Lymph node and blood LPCs were quantified at diagnosis, on the fourth day of neoadjuvant period, and 1-week after the first chemotherapy dose. Valspodar therapy was well tolerated. There were no differences between groups in total LPCs in lymph nodes or peripheral blood, nor in event-free survival or overall survival. Overall, we conclude that valspodar can be administered safely in the neoadjuvant setting for canine B-cell lymphoma; however, its use to attenuate ABCB1 + cells does not alter the composition of lymph node or blood LPCs, and it does not appear to be sufficient to prolong doxorubicin-dependent remissions in this setting.

10.
J Med Chem ; 58(16): 6348-58, 2015 Aug 27.
Artigo em Inglês | MEDLINE | ID: mdl-26181851

RESUMO

As a result of our efforts to discover novel p53:MDM2 protein-protein interaction inhibitors useful for treating cancer, the potent and selective MDM2 inhibitor NVP-CGM097 (1) with an excellent in vivo profile was selected as a clinical candidate and is currently in phase 1 clinical development. This article provides an overview of the discovery of this new clinical p53:MDM2 inhibitor. The following aspects are addressed: mechanism of action, scientific rationale, binding mode, medicinal chemistry, pharmacokinetic and pharmacodynamic properties, and in vivo pharmacology/toxicology in preclinical species.


Assuntos
Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Isoquinolinas/síntese química , Isoquinolinas/farmacologia , Piperazinas/síntese química , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Animais , Antineoplásicos/farmacocinética , Linhagem Celular Tumoral , Ensaios Clínicos Fase I como Assunto , Descoberta de Drogas , Humanos , Isoquinolinas/farmacocinética , Piperazinas/farmacocinética , Ratos , Relação Estrutura-Atividade , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Elife ; 42015 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-25965177

RESUMO

Biomarkers for patient selection are essential for the successful and rapid development of emerging targeted anti-cancer therapeutics. In this study, we report the discovery of a novel patient selection strategy for the p53-HDM2 inhibitor NVP-CGM097, currently under evaluation in clinical trials. By intersecting high-throughput cell line sensitivity data with genomic data, we have identified a gene expression signature consisting of 13 up-regulated genes that predicts for sensitivity to NVP-CGM097 in both cell lines and in patient-derived tumor xenograft models. Interestingly, these 13 genes are known p53 downstream target genes, suggesting that the identified gene signature reflects the presence of at least a partially activated p53 pathway in NVP-CGM097-sensitive tumors. Together, our findings provide evidence for the use of this newly identified predictive gene signature to refine the selection of patients with wild-type p53 tumors and increase the likelihood of response to treatment with p53-HDM2 inhibitors, such as NVP-CGM097.


Assuntos
Biomarcadores/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Isoquinolinas/farmacologia , Neoplasias/tratamento farmacológico , Seleção de Pacientes , Piperazinas/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Linhagem Celular Tumoral , Transferência Ressonante de Energia de Fluorescência , Perfilação da Expressão Gênica , Humanos , Análise de Sequência com Séries de Oligonucleotídeos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo
12.
Curr Med Chem Anticancer Agents ; 3(1): 1-14, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12678910

RESUMO

The importance of Cdks in cell cycle regulation, their interaction with oncogenes and tumor suppressors, and their frequent deregulation in human tumors, has encouraged an active search for agents capable of perturbing the function of Cdks. In our laboratories, a variety of selective and potent low molecular weight inhibitors directed against the ATP binding sites of the Cdk1, Cdk2 have been developed. Extensive biological profiling of two distinct classes of Cdk inhibitors - the phenylamino pyrimidines (PAPs) and trisubstituted purines has revealed distinct differences in their cellular effects in normal cells compared to tumor cells. Due to their intact G1/S checkpoints, normal cells are shown to be reversibly blocked by these Cdk inhibitors in either the G1/S-phase or at the G2/M boarder. In transformed cells these control points are either absent or defective and treatment with the compounds resulted in pronounced proliferation block at the G2/M transition. Furthermore, there is strong evidence that this G2/M arrest is less well tolerated by the cells and consequently, they undergo apoptotic cell death. Finally, these dual Cdk1/ Cdk2 inhibitors are also found to be significantly more active on proliferating cells compared to quiescent cells reflecting their specific activity. Despite these encouraging results demonstrating a distinct outcome after treatment with such dual Cdk inhibitors in normal compared to de-regulated tumor cells, it remains to be determined whether a comparable therapeutic window might be observed in vivo experiments. Furthermore the intracellular kinase selectivity of inhibitors which are putatively selective in vitro remains a complicating feature that is only recently begun to be addressed by affinity chromatography and phosphoproteomics techniques. Once efficacy can be demonstrated in animal models at well-tolerated doses, there will be strong evidence for the development of cell cycle antagonists for cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Proteína Quinase CDC2/antagonistas & inibidores , Quinases relacionadas a CDC2 e CDC28 , Quinases Ciclina-Dependentes/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Animais , Antineoplásicos/química , Sítios de Ligação , Ciclo Celular/efeitos dos fármacos , Quinase 2 Dependente de Ciclina , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Purinas/química , Purinas/farmacologia , Purinas/uso terapêutico , Pirimidinas/química , Pirimidinas/farmacologia , Pirimidinas/uso terapêutico , Relação Estrutura-Atividade
14.
Clin Cancer Res ; 16(1): 212-25, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-20008843

RESUMO

PURPOSE: Identification of a generic response biomarker by comparison of chemotherapeutics with different action mechanisms on several noninvasive biomarkers in experimental tumor models. EXPERIMENTAL DESIGN: The spin-lattice relaxation time of water protons (T(1)) was quantified using an inversion recovery-TrueFISP magnetic resonance imaging method in eight different experimental tumor models before and after treatment at several different time points with five different chemotherapeutics. Effects on T(1) were compared with other minimally invasive biomarkers including vascular parameters, apparent diffusion coefficient, and interstitial fluid pressure, and were correlated with efficacy at the endpoint and histologic parameters. RESULTS: In all cases, successful chemotherapy significantly lowered tumor T(1) compared with vehicle and the fractional change in T(1) (DeltaT(1)) correlated with the eventual change in tumor size (range: r(2) = 0.21, P < 0.05 to r(2) = 0.73, P < 0.0001), except for models specifically resistant to that drug. In RIF-1 tumors, interstitial fluid pressure was decreased, but apparent diffusion coefficient and permeability increased in response to the microtubule stabilizer patupilone and 5-fluorouracil. Although DeltaT(1) was small (maximum of -20%), the variability was very low (5%) compared with other magnetic resonance imaging methods (24-48%). Analyses ex vivo showed unchanged necrosis, increased apoptosis, and decreased %Ki67 and total choline, but only Ki67 and choline correlated with DeltaT(1). Correlation of Ki67 and DeltaT(1) were observed in other models using patupilone, paclitaxel, a VEGF-R inhibitor, and the mammalian target of rapamycin inhibitor everolimus. CONCLUSIONS: These results suggest that a decrease in tumor T(1) reflects hypocellularity and is a generic marker of response. The speed and robustness of the method should facilitate its use in clinical trials.


Assuntos
Antineoplásicos/farmacologia , Biomarcadores , Imageamento por Ressonância Magnética/métodos , Neoplasias Experimentais/patologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Espectroscopia de Ressonância Magnética , Camundongos , Camundongos Endogâmicos , Transplante de Neoplasias , Neoplasias Experimentais/tratamento farmacológico , Ratos , Ratos Endogâmicos BN
15.
Mol Cancer Ther ; 9(4): 906-19, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20371713

RESUMO

Heat shock protein 90 (Hsp90) is a ubiquitously expressed molecular chaperone with ATPase activity involved in the conformational maturation and stability of key signaling molecules involved in cell proliferation, survival, and transformation. Through its ability to modulate multiple pathways involved in oncogenesis, Hsp90 has generated considerable interest as a therapeutic target. NVP-BEP800 is a novel, fully synthetic, orally bioavailable inhibitor that binds to the NH(2)-terminal ATP-binding pocket of Hsp90. NVP-BEP800 showed activity against a panel of human tumor cell lines and primary human xenografts in vitro at nanomolar concentrations. In A375 melanoma and BT-474 breast cancer cell lines, NVP-BEP800 induced client protein degradation (including ErbB2, B-Raf(V600E), Raf-1, and Akt) and Hsp70 induction. Oral administration of NVP-BEP800 was well tolerated and induced robust antitumor responses in tumor xenograft models, including regression in the BT-474 breast cancer model. In these tumor models, NVP-BEP800 modulated Hsp90 client proteins and downstream signaling pathways at doses causing antitumor activity. NVP-BEP800 showed in vivo activity in a variety of dosing regimens covering daily to weekly schedules, potentially providing a high degree of flexibility in dose and schedule within the clinical setting. Overall, given the mechanism of action, preclinical activity profile, tolerability, and pharmaceutical properties, NVP-BEP800 is an exciting new oral Hsp90 inhibitor warranting further development. Mol Cancer Ther; 9(4); 906-19. (c)2010 AACR.


Assuntos
Proteínas de Choque Térmico HSP90/antagonistas & inibidores , Pirimidinas/farmacologia , Pirimidinas/farmacocinética , Ensaios Antitumorais Modelo de Xenoenxerto , Administração Oral , Animais , Apoptose/efeitos dos fármacos , Disponibilidade Biológica , Neoplasias da Mama/patologia , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Isoxazóis/química , Isoxazóis/farmacologia , Camundongos , Camundongos Nus , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Pirimidinas/efeitos adversos , Pirimidinas/química , Resorcinóis/química , Resorcinóis/farmacologia
16.
Mol Cancer Ther ; 9(7): 1945-55, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20587663

RESUMO

The recent discovery of an acquired activating point mutation in JAK2, substituting valine at amino acid position 617 for phenylalanine, has greatly improved our understanding of the molecular mechanism underlying chronic myeloproliferative neoplasms. Strikingly, the JAK2(V617F) mutation is found in nearly all patients suffering from polycythemia vera and in roughly every second patient suffering from essential thrombocythemia and primary myelofibrosis. Thus, JAK2 represents a promising target for the treatment of myeloproliferative neoplasms and considerable efforts are ongoing to discover and develop inhibitors of the kinase. Here, we report potent inhibition of JAK2(V617F) and JAK2 wild-type enzymes by a novel substituted quinoxaline, NVP-BSK805, which acts in an ATP-competitive manner. Within the JAK family, NVP-BSK805 displays more than 20-fold selectivity towards JAK2 in vitro, as well as excellent selectivity in broader kinase profiling. The compound blunts constitutive STAT5 phosphorylation in JAK2(V617F)-bearing cells, with concomitant suppression of cell proliferation and induction of apoptosis. In vivo, NVP-BSK805 exhibited good oral bioavailability and a long half-life. The inhibitor was efficacious in suppressing leukemic cell spreading and splenomegaly in a Ba/F3 JAK2(V617F) cell-driven mouse mechanistic model. Furthermore, NVP-BSK805 potently suppressed recombinant human erythropoietin-induced polycythemia and extramedullary erythropoiesis in mice and rats.


Assuntos
Proliferação de Células/efeitos dos fármacos , Janus Quinase 2/antagonistas & inibidores , Policitemia/prevenção & controle , Quinoxalinas/farmacologia , Trifosfato de Adenosina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Eritropoese/efeitos dos fármacos , Humanos , Janus Quinase 2/química , Janus Quinase 2/genética , Células K562 , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , Modelos Moleculares , Estrutura Molecular , Mutação , Fosforilação/efeitos dos fármacos , Policitemia/metabolismo , Policitemia/patologia , Estrutura Terciária de Proteína , Quinoxalinas/química , Ratos , Fator de Transcrição STAT5/metabolismo , Esplenomegalia/metabolismo , Esplenomegalia/patologia , Esplenomegalia/prevenção & controle
17.
Cell ; 120(6): 747-59, 2005 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-15797377

RESUMO

Although DNA damaging agents have revolutionized chemotherapy against solid tumors, a narrow therapeutic window combined with severe side effects has limited their broader use. Here we show that RAD001 (everolimus), a rapamycin derivative, dramatically enhances cisplatin-induced apoptosis in wild-type p53, but not mutant p53 tumor cells. The use of isogenic tumor cell lines expressing either wild-type mTOR cDNA or a mutant that does not bind RAD001 demonstrates that the effects of RAD001 are through inhibition of mTOR function. We further show that RAD001 sensitizes cells to cisplatin by inhibiting p53-induced p21 expression. Unexpectedly, this effect is attributed to a small but significant inhibition of p21 translation combined with its short half-life. These findings provide the molecular rationale for combining DNA damaging agents with RAD001, showing that a general effect on a major anabolic process may dramatically enhance the efficacy of an established drug protocol in the treatment of cancer patients with solid tumors.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Proteínas de Ciclo Celular/antagonistas & inibidores , Cisplatino/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Quinases/metabolismo , Sirolimo/análogos & derivados , Sirolimo/farmacologia , Apoptose/genética , Proteínas de Ciclo Celular/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Inibidor de Quinase Dependente de Ciclina p21 , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/genética , Dano ao DNA/fisiologia , Sinergismo Farmacológico , Everolimo , Humanos , Mutação , Polirribossomos/efeitos dos fármacos , Polirribossomos/genética , Polirribossomos/metabolismo , Biossíntese de Proteínas/genética , Biossíntese de Proteínas/fisiologia , Proteínas Quinases/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Serina-Treonina Quinases TOR , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
18.
Exp Cell Res ; 303(2): 482-93, 2005 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-15652359

RESUMO

Ubiquitin-mediated degradation of the cyclin-dependent kinase inhibitor p27Kip1 was shown to be required for the activation of key cyclin-dependent kinases, thereby triggering the onset of DNA replication and cell cycle progression. Although the SCFSkp2 ubiquitin ligase has been reported to mediate p27Kip1 degradation, the nature of the human ubiquitin-conjugating enzyme involved in this process has not yet been determined at the cellular level. Here, we show that antisense oligonucleotides targeting the human ubiquitin-conjugating enzyme Cdc34 downregulate its expression, inhibit the degradation of p27Kip1, and prevent cellular proliferation. Elevation of p27Kip1 protein level is found to be the sole requirement for the inhibition of cellular proliferation induced upon downregulation of Cdc34. Indeed, reducing the expression of p27Kip1 with a specific antisense oligonucleotide is sufficient to reverse the anti-proliferative phenotype elicited by the Cdc34 antisense. Furthermore, downregulation of Cdc34 is found to specifically increase the abundance of the SCFSkp2) ubiquitin ligase substrate p27Kip1, but has no concomitant effect on the level of IkBalpha and beta-catenin, which are known substrates of a closely related SCF ligase.


Assuntos
Proteínas de Transporte/metabolismo , Proliferação de Células , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Complexos Ubiquitina-Proteína Ligase/metabolismo , Ciclossomo-Complexo Promotor de Anáfase , Sequência de Bases , Proteínas de Transporte/genética , Ciclo Celular , Linhagem Celular , Inibidor de Quinase Dependente de Ciclina p27 , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Oligodesoxirribonucleotídeos Antissenso/genética , Oligodesoxirribonucleotídeos Antissenso/farmacologia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Especificidade por Substrato , Enzimas de Conjugação de Ubiquitina , Complexos Ubiquitina-Proteína Ligase/genética
19.
Bioorg Med Chem Lett ; 13(2): 247-51, 2003 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-12482432

RESUMO

We describe the design, synthesis and cell-membrane translocation properties of a series of beta-peptides with the general sequence fluorescein-Adoa-(beta-homolysine)(n)-NH(2), n=5-8 and Adoa=8-amino-3,6-dioxaoctanoic acid. These beta-peptides are able to cross the cytoplasmic membrane and accumulate in the nucleus of mammalian cells.


Assuntos
Oligopeptídeos/química , Oligopeptídeos/síntese química , Oligopeptídeos/farmacologia , Animais , Linhagem Celular , Membrana Celular/metabolismo , Desenho de Fármacos , Fluoresceína/síntese química , Fluoresceína-5-Isotiocianato , Citometria por Imagem , Oligopeptídeos/metabolismo , Azida Sódica/farmacologia , Vasodilatadores/farmacologia
20.
J Pharmacol Exp Ther ; 302(3): 1055-61, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12183663

RESUMO

Bisphosphonate drugs inhibit osteoclastic bone resorption and are widely used to treat skeletal complications in patients with tumor-induced osteolysis. We now show that zoledronic acid, a new generation bisphosphonate with a heterocyclic imidazole substituent, is also a potent inhibitor of angiogenesis. In vitro, zoledronic acid inhibits proliferation of human endothelial cells stimulated with fetal calf serum, basic fibroblast growth factor (bFGF), and vascular endothelial growth factor (IC(50) values 4.1, 4.2, and 6.9 microM, respectively), and modulates endothelial cell adhesion and migration. In cultured aortic rings and in the chicken egg chorioallantoic membrane assay, zoledronic acid reduces vessel sprouting. When administered systemically to mice, zoledronic acid potently inhibits the angiogenesis induced by subcutaneous implants impregnated with bFGF [ED(50), 3 microg/kg (7.5 nmol/kg) s.c.]. These findings indicate that zoledronic acid has marked antiangiogenic properties that could augment its efficacy in the treatment of malignant bone disease and extend its potential clinical use to other diseases with an angiogenic component.


Assuntos
Inibidores da Angiogênese/farmacologia , Difosfonatos/farmacologia , Imidazóis/farmacologia , Apoptose/efeitos dos fármacos , Adesão Celular/efeitos dos fármacos , Ciclo Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Quimiotaxia/efeitos dos fármacos , Córion/química , Fatores de Crescimento Endotelial/farmacologia , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Substâncias de Crescimento/administração & dosagem , Substâncias de Crescimento/farmacologia , Humanos , Citometria por Imagem , Linfocinas/farmacologia , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Pamidronato , Fator A de Crescimento do Endotélio Vascular , Fatores de Crescimento do Endotélio Vascular , Ácido Zoledrônico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA