Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Clin Oncol ; 39(33): 3660-3670, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34637337

ABSTRACT

PURPOSE: Malignant perivascular epithelioid cell tumor (PEComa) is a rare aggressive sarcoma, with no approved treatment. To our knowledge, this phase II, single-arm, registration trial is the first prospective clinical trial in this disease, investigating the safety and efficacy of the mammalian target of rapamycin inhibitor nab-sirolimus (AMPECT, NCT02494570). PATIENTS AND METHODS: Patients with malignant PEComa were treated with nab-sirolimus 100 mg/m2 intravenously once weekly for 2 weeks in 3-week cycles. The primary end point was objective response rate evaluated by independent radiology review. Key secondary end points included duration of response, progression-free survival, and safety. A key exploratory end point was tumor biomarker analysis. RESULTS: Thirty-four patients were treated (safety evaluable), and 31 were evaluable for efficacy. The overall response rate was 39% (12 of 31; 95% CI, 22 to 58) with one complete and 11 partial responses, 52% (16 of 31) of patients had stable disease, and 10% (3 of 31) had progressive disease. Responses were of rapid onset (67% by week 6) and durable. Median duration of response was not reached after a median follow-up for response of 2.5 years, with 7 of 12 responders with treatment ongoing (range, 5.6-47.2+ months). Twenty-five of 31 patients had tumor mutation profiling: 8 of 9 (89%) patients with a TSC2 mutation achieved a confirmed response versus 2 of 16 (13%) without TSC2 mutation (P < .001). The median progression-free survival was 10.6 months (95% CI, 5.5 months to not reached), and the median overall survival was 40.8 months (95% CI, 22.2 months to not reached). Most treatment-related adverse events were grade 1 or 2 and were manageable for long-term treatment. No grade ≥ 4 treatment-related events occurred. CONCLUSION: nab-Sirolimus is active in patients with malignant PEComa. The response rate, durability of response, disease control rate, and safety profile support that nab-sirolimus represents an important new treatment option for this disease.


Subject(s)
Albumins/metabolism , Antibiotics, Antineoplastic/therapeutic use , Nanoparticles/administration & dosage , Perivascular Epithelioid Cell Neoplasms/drug therapy , Sirolimus/therapeutic use , Adult , Aged , Albumins/chemistry , Antibiotics, Antineoplastic/chemistry , Female , Follow-Up Studies , Humans , Male , Middle Aged , Nanoparticles/chemistry , Neoplasm Metastasis , Perivascular Epithelioid Cell Neoplasms/pathology , Prognosis , Prospective Studies , Sirolimus/chemistry , Survival Rate
2.
Br J Cancer ; 115(4): 442-53, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27441498

ABSTRACT

BACKGROUND: Albumin-bound paclitaxel (nab-paclitaxel, nab-PTX) plus gemcitabine (GEM) combination has demonstrated efficient antitumour activity and statistically significant overall survival of patients with metastatic pancreatic ductal adenocarcinoma (PDAC) compared with GEM monotherapy. This regimen is currently approved as a standard of care treatment option for patients with metastatic PDAC. It is unclear whether cremophor-based PTX combined with GEM provide a similar level of therapeutic efficacy in PDAC. METHODS: We comprehensively explored the antitumour efficacy, effect on metastatic dissemination, tumour stroma and survival advantage following GEM, PTX and nab-PTX as monotherapy or in combination with GEM in a locally advanced, and a highly metastatic orthotopic model of human PDAC. RESULTS: Nab-PTX treatment resulted in significantly higher paclitaxel tumour plasma ratio (1.98-fold), robust stromal depletion, antitumour efficacy (3.79-fold) and survival benefit compared with PTX treatment. PTX plus GEM treatment showed no survival gain over GEM monotherapy. However, nab-PTX in combination with GEM decreased primary tumour burden, metastatic dissemination and significantly increased median survival of animals compared with either agents alone. These therapeutic effects were accompanied by depletion of dense fibrotic tumour stroma and decreased proliferation of carcinoma cells. Notably, nab-PTX monotherapy was equivalent to nab-PTX plus GEM in providing survival advantage to mice in a highly aggressive metastatic PDAC model, indicating that nab-PTX could potentially stop the progression of late-stage pancreatic cancer. CONCLUSIONS: Our data confirmed that therapeutic efficacy of PTX and nab-PTX vary widely, and the contention that these agents elicit similar antitumour response was not supported. The addition of PTX to GEM showed no survival advantage, concluding that a clinical combination of PTX and GEM may unlikely to provide significant survival advantage over GEM monotherapy and may not be a viable alternative to the current standard-of-care nab-PTX plus GEM regimen for the treatment of PDAC patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Kidney Neoplasms/drug therapy , Liver Neoplasms/drug therapy , Lung Neoplasms/drug therapy , Pancreatic Neoplasms/drug therapy , Splenic Neoplasms/drug therapy , Albumins/administration & dosage , Animals , Carcinoma, Pancreatic Ductal/secondary , Cell Proliferation , Deoxycytidine/administration & dosage , Deoxycytidine/analogs & derivatives , Humans , Kidney Neoplasms/secondary , Liver Neoplasms/secondary , Lung Neoplasms/secondary , Male , Mice , Mice, Nude , Neoplasm Metastasis , Neovascularization, Pathologic , Paclitaxel/administration & dosage , Pancreatic Neoplasms/pathology , Polyethylene Glycols/administration & dosage , Splenic Neoplasms/secondary , Xenograft Model Antitumor Assays , Gemcitabine
3.
Cancer Chemother Pharmacol ; 76(4): 699-712, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26231955

ABSTRACT

PURPOSE: nab-paclitaxel demonstrates improved clinical efficacy compared with conventional Cremophor EL (CrEL)-paclitaxel in multiple tumor types. This study explored the distinctions in drug distribution between nab-paclitaxel and CrEL-paclitaxel and the underlying mechanisms. METHODS: Uptake and transcytosis of paclitaxel were analyzed by vascular permeability assay across human endothelial cell monolayers. The tissue penetration of paclitaxel within tumors was evaluated by local injections into tumor xenografts and quantitative image analysis. The distribution profile of paclitaxel in solid-tumor patients was assessed using pharmacokinetic modeling and simulation. RESULTS: Live imaging demonstrated that albumin and paclitaxel were present in punctae in endothelial cells and could be observed in very close proximity, suggesting cotransport. Uptake and transport of albumin, nab-paclitaxel and paclitaxel were inhibited by clinically relevant CrEL concentrations. Further, nab-paclitaxel causes greater mitotic arrest in wider area within xenografted tumors than CrEL- or dimethyl sulfoxide-paclitaxel following local microinjection, demonstrating enhanced paclitaxel penetration and uptake by albumin within tumors. Modeling of paclitaxel distribution in patients with solid tumors indicated that nab-paclitaxel is more dependent upon transporter-mediated pathways for drug distribution into tissues than CrEL-paclitaxel. The percent dose delivered to tissue via transporter-mediated pathways is predicted to be constant with nab-paclitaxel but decrease with increasing CrEL-paclitaxel dose. CONCLUSIONS: Compared with CrEL-paclitaxel, nab-paclitaxel demonstrated more efficient transport across endothelial cells, greater penetration and cytotoxic induction in xenograft tumors, and enhanced extravascular distribution in patients that are attributed to carrier-mediated transport. These observations are consistent with the distinct clinical efficacy and toxicity profile of nab-paclitaxel.


Subject(s)
Antineoplastic Agents, Phytogenic/administration & dosage , Drug Delivery Systems , Endothelium, Vascular/metabolism , Nanoparticles/chemistry , Paclitaxel/administration & dosage , Pancreatic Neoplasms/drug therapy , Serum Albumin/chemistry , Animals , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/therapeutic use , Biological Transport/drug effects , Capillary Permeability/drug effects , Carcinoma/drug therapy , Carcinoma/metabolism , Carcinoma/pathology , Cell Line, Tumor , Cells, Cultured , Endosomes/drug effects , Endosomes/metabolism , Endosomes/pathology , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Infusions, Intravenous , Mice, Nude , Microinjections , Paclitaxel/metabolism , Paclitaxel/pharmacokinetics , Paclitaxel/therapeutic use , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Serum Albumin/metabolism , Serum Albumin, Human , Tissue Distribution , Tubulin Modulators/administration & dosage , Tubulin Modulators/metabolism , Tubulin Modulators/pharmacokinetics , Tubulin Modulators/therapeutic use , Xenograft Model Antitumor Assays , Pancreatic Neoplasms
4.
Cancer ; 121(11): 1817-26, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25649370

ABSTRACT

BACKGROUND: The optimal weekly oral dose of sirolimus and intravenous nanoparticle albumin-bound paclitaxel (nab-paclitaxel) were evaluated. METHODS: A phase 1b study was performed to evaluate escalating doses of oral sirolimus (5-60 mg) on days 2, 9, and 16 with intravenous nab-paclitaxel (100 mg/m(2) ) on days 1, 8, and 15 in a 28-day cycle. A run-in treatment of nab-paclitaxel (day -14) and sirolimus (day -7) was administered for pharmacokinetic and pharmacodynamic assessments. Clinical trial endpoints included dose-limiting toxicities (DLTs), maximum tolerated doses, and response rates. Pharmacodynamics included immunohistochemistry for phosphatase and tensin homolog, mammalian target of rapamycin (mTOR), AKT, phosphorylated AKT, S6K1, and phosphorylated S6K1; exploratory gene expression analysis; and [(18) F]fludeoxyglucose (FDG) positron emission tomography. RESULTS: Twenty-three patients with advanced solid tumors were treated. Fifteen patients had prior taxane therapy. Twenty-two patients were evaluable for responses. One patient had a complete response, and 5 patients had a partial response (3 confirmed). DLTs were seen in 1 patient each at 10 (grade 3 dyspnea/hypoxia) and 40 mg (grade 4 leukopenia/neutropenia) and in 2 patients at 60 mg (grade 3 fatigue and grade 4 pericardial effusion). Patients with higher expression of posttreatment AKT and a greater decline in FDG activity were more likely to have a treatment response or stable disease. CONCLUSIONS: Sirolimus showed an acceptable safety profile at a weekly dose of 40 mg with weekly intravenous nab-paclitaxel at 100 mg/m(2) on days 1, 8, and 15 every 28 days. The posttreatment AKT score and changes in FDG activity may have roles as early predictors of responses to mTOR inhibitors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Neoplasms/drug therapy , TOR Serine-Threonine Kinases/antagonists & inhibitors , Adult , Aged , Albumin-Bound Paclitaxel , Albumins/administration & dosage , Albumins/pharmacokinetics , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cohort Studies , Female , Fluorodeoxyglucose F18 , Humans , Male , Middle Aged , Nanoparticles/administration & dosage , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Paclitaxel/administration & dosage , Paclitaxel/pharmacokinetics , Positron-Emission Tomography/methods , Sirolimus/administration & dosage , Sirolimus/pharmacokinetics , Treatment Outcome
5.
J Biol Chem ; 282(24): 17450-9, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17438336

ABSTRACT

Focal adhesion kinase (FAK) is a cytoplasmic protein-tyrosine kinase that promotes cell migration, survival, and gene expression. Here we show that FAK signaling is important for tumor necrosis factor-alpha (TNFalpha)-induced interleukin 6 (IL-6) mRNA and protein expression in breast (4T1), lung (A549), prostate (PC-3), and neural (NB-8) tumor cells by FAK short hairpin RNA knockdown and by comparisons of FAK-null (FAK(-/-)) and FAK(+/+) mouse embryo fibroblasts. FAK promoted TNFalpha-stimulated MAPK activation needed for maximal IL-6 production. FAK was not required for TNFalpha-mediated nuclear factor-kappaB or c-Jun N-terminal kinase activation. TNFalpha-stimulated FAK catalytic activation and IL-6 production were inhibited by FAK N-terminal but not FAK C-terminal domain overexpression. Analysis of FAK(-/-) fibroblasts stably reconstituted with wild type or various FAK point mutants showed that FAK catalytic activity, Tyr-397 phosphorylation, and the Pro-712/713 proline-rich region of FAK were required for TNFalpha-stimulated MAPK activation and IL-6 production. Constitutively activated MAPK kinase-1 (MEK1) expression in FAK(-/-) and A549 FAK short hairpin RNA-expressing cells rescued TNFalpha-stimulated IL-6 production. Inhibition of Src protein-tyrosine kinase activity or mutation of Src phosphorylation sites on FAK (Tyr-861 or Tyr-925) did not affect TNFalpha-stimulated IL-6 expression. Moreover, analyses of Src(-/-), Yes(-/-), and Fyn(-/-) fibroblasts showed that Src expression was inhibitory to TNFalpha-stimulated IL-6 production. These studies provide evidence for a novel Src-independent FAK to MAPK signaling pathway regulating IL-6 expression with potential importance to inflammation and tumor progression.


Subject(s)
Focal Adhesion Protein-Tyrosine Kinases/metabolism , Interleukin-6/metabolism , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/physiology , Tumor Necrosis Factor-alpha/metabolism , Animals , Cell Line, Tumor , Enzyme Activation , Fibroblasts/cytology , Fibroblasts/metabolism , Focal Adhesion Protein-Tyrosine Kinases/genetics , Interleukin-6/genetics , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/genetics , NF-kappa B/metabolism , Proline/metabolism , RNA Interference , Tyrosine/metabolism , src-Family Kinases/metabolism
6.
Curr Protoc Cell Biol ; Chapter 14: Unit 14.7, 2006 Apr.
Article in English | MEDLINE | ID: mdl-18228476

ABSTRACT

Integrins are a family of heterodimeric alpha/beta transmembrane cell adhesion receptors that play important roles in the regulation of cell migration, proliferation, and survival. Integrins do not possess intrinsic catalytic activity, and signaling events are mediated by their lateral association with other cell surface receptors or clustering of their cytoplasmic domains with signaling proteins. Rapid activation of protein-tyrosine kinases is one of the first signaling events associated with integrin binding to the extracellular matrix protein fibronectin. The intracellular focal adhesion kinase (FAK) is recruited to sites of integrin clustering, and this unit describes the methods with which to analyze FAK phosphorylation, activity, and localization within fibroblasts. Additional methods on how to grow primary FAK+/+ and FAK-/- fibroblasts and measure integrin-stimulated cell motility are described as well as methods for evaluating the activity of the FAK-related kinase, Pyk2, which is expressed in FAK-/- cells.


Subject(s)
Cell Culture Techniques/methods , Clinical Laboratory Techniques , Focal Adhesion Kinase 2/analysis , Focal Adhesion Protein-Tyrosine Kinases/analysis , Integrins/metabolism , Signal Transduction/physiology , Animals , Cell Migration Assays/methods , Cells, Cultured , Fibroblasts/enzymology , Immunoblotting/methods , Immunohistochemistry/methods , Mice , Wound Healing/physiology
7.
J Biol Chem ; 280(11): 9957-62, 2005 Mar 18.
Article in English | MEDLINE | ID: mdl-15642694

ABSTRACT

The NF-kappaB p50/p50 homodimer is mainly associated with transcriptional repression. Previously, we demonstrated that phosphorylation of NF-kappaB p50 Ser(337) is critical for DNA binding. Here, we report that p50 Ser(337) is constitutively phosphorylated by the protein kinase A catalytic subunit (PKAc) in three different cell types, which may account for the constant binding of p50/p50 to DNA in unstimulated cells. This was demonstrated first by showing that treatment of cells with PKAc-specific inhibitors blocked p50/p50 DNA binding. Second, phosphorylation of p50 by PKAc was prevented by substitution of Ser(337) to alanine. Third, both p50 and PKAc proteins as well as kinase activity that phosphorylates p50 were found to co-fractionate following gel filtration chromatography. Finally, PKAc and p50 were shown to be able to reciprocally co-immunoprecipitate one another, and their physical association was blocked by a PKA catalytic site inhibitory peptide. This indicates that phosphorylation of p50 Ser(337) involves direct contact with the PKAc catalytic center. In contrast to the dramatic elevation of nuclear p50/p65 heterodimers induced by tumor necrosis factor alpha, DNA binding of p50/p50 homodimers was not greatly altered. Taken together, these findings reveal for the first time that there is a direct interaction between PKAc and p50 that accounts for constitutive phosphorylation of p50 Ser(337) and the existence of DNA bound p50/p50 in the nuclei of most resting cells. This mechanism of DNA binding by p50/p50 following phosphorylation of Ser(337) by PKAc may represent an important means for maintaining stable negative regulation of NF-kappaB gene expression in the absence of extracellular stimulation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/chemistry , DNA/chemistry , Transcription Factors/metabolism , Alanine/chemistry , Animals , COS Cells , Catalytic Domain , Cell Line , Cell Nucleus/metabolism , Chromatography, Gel , Cytoplasm/metabolism , Dimerization , Enzyme Inhibitors/pharmacology , HeLa Cells , Humans , Immunoblotting , Immunoprecipitation , NF-kappa B p50 Subunit , Peptides/chemistry , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Conformation , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Serine/chemistry , Transfection , Tumor Necrosis Factor-alpha/metabolism
8.
J Biol Chem ; 278(46): 45994-8, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-12947093

ABSTRACT

It has been demonstrated that phosphorylation of the p50 subunit of NF-kappaB is required for efficient DNA binding, yet the specific phospho-residues of p50 have not been determined. In this study, we substituted all of the serine and conserved threonine residues in the p50 Rel homology domain and identified three serine residues, Ser65, Ser337, and Ser342, as critical for DNA binding without affecting dimerization. Although substitution with negatively charged aspartic acid at each of these positions failed to restore DNA binding, substitution with threonine, a potential phospho-acceptor, retained DNA binding for residues 65 and 337. In particular, Ser337, in a consensus site for protein kinase A (PKA) and other kinases, was shown to be phosphorylated both in vitro and in vivo. Importantly, phosphorylation of Ser337 by PKA in vitro dramatically increased DNA binding of p50. This study shows for the first time that the DNA binding ability of NF-kappaB p50 subunit is regulated through phosphorylation of residue Ser337, which has implications for both positive and negative control of NF-kappaB transcription.


Subject(s)
DNA/metabolism , NF-kappa B/chemistry , Serine/chemistry , Animals , Binding Sites , Blotting, Western , COS Cells , Cell Line , Cyclic AMP-Dependent Protein Kinases/chemistry , Dimerization , Electrophoresis, Polyacrylamide Gel , HeLa Cells , Humans , Models, Molecular , Mutagenesis, Site-Directed , NF-kappa B/metabolism , NF-kappa B p50 Subunit , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Biosynthesis , Threonine/chemistry , Transcription Factor RelA , Transcription, Genetic , Transfection
9.
Virology ; 306(1): 68-76, 2003 Feb 01.
Article in English | MEDLINE | ID: mdl-12620799

ABSTRACT

In adenovirus type 12 transformed cells, the down-regulation of MHC class I transcription contributes to the tumorigenic phenotype and is solely mediated by Ad12 E1A. Previous in vitro studies with class I enhancer sequences have indicated that there is an increased binding of repressor COUP-TFII and its associated HDAC and a decreased binding of activator NF-kappaB. In this study, we used chromatin immunoprecipitation (ChIP) assay in order to determine in vivo whether these proteins regulate class I transcription by affecting chromatin. The ChIP assay revealed that there is lack of chromatin histone acetylation in the region of the class I enhancer in Ad12-transformed cells. This is regulated by histone deacetylation as it was further demonstrated in vivo that COUP-TFII and HDAC are associated with the class I enhancer chromatin. In agreement with in vitro studies, NF-kappaB could be recruited to the class I enhancer following induction by TNF-alpha. However, this enhancer-bound NF-kappaB failed to up-regulate class I expression because the class I enhancer chromatin remained repressed as a result of histone deacetylation by HDAC in association with COUP-TFII. Thus, we have demonstrated for the first time that repression of chromatin through histone deacetylation is a major mechanism in down-regulating class I transcription in Ad12-transformed cells. Finally, Ad12 E1A, a non-DNA binding protein, was shown to be present in the natural protein complex bound to the class I enhancer.


Subject(s)
Chromatin/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Histocompatibility Antigens Class I/metabolism , Histone Deacetylases/metabolism , NF-kappa B/metabolism , Receptors, Steroid , Transcription Factors/metabolism , Transcription, Genetic , Adenoviridae , Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Animals , Base Sequence , COUP Transcription Factor II , COUP Transcription Factors , Cell Transformation, Viral , Enhancer Elements, Genetic/genetics , Histocompatibility Antigens Class I/genetics , Mice , Molecular Sequence Data , Rats , Transcriptional Activation , Tumor Cells, Cultured
10.
J Virol ; 76(7): 3212-20, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11884545

ABSTRACT

The surface levels of major histocompatibility complex class I antigens are diminished on tumorigenic adenovirus type 12 (Ad12)-transformed cells, enabling them to escape from immunosurveillant cytotoxic T lymphocytes (CTLs). This is due to the down-regulation of the class I transcriptional enhancer, in which there is strong binding of the repressor COUP-TFII and lack of binding of the activator NF-kappaB. Even though NF-kappaB (p65/p50) translocates to the nuclei of Ad12-transformed cells, it fails to bind to DNA efficiently due to the hypophosphorylation of the p50 subunit. In this study, tumor necrosis factor alpha (TNF-alpha) and interleukin 1beta (IL-1beta) were shown to promote degradation of the NF-kappaB cytoplasmic inhibitor IkappaBalpha and permit the nuclear translocation of a phosphorylated form of NF-kappaB that is capable of binding DNA. Interestingly, when Ad12-transformed cells were treated with TNF-alpha or IL-1beta, class I gene transcription substantially increased when transcriptional repression by COUP-TFII was blocked. This indicates that in cytokine-treated Ad12-transformed cells, COUP-TFII is able to repress activation of class I transcription by newly nucleus-localized NF-kappaB. Our results suggest that Ad12 likely employs a "fail-safe" mechanism to ensure that the transcription of class I genes remains tightly repressed under various physiological conditions, thus providing tumorigenic Ad12-transformed cells with a means of escaping CTL recognition and lysis.


Subject(s)
DNA-Binding Proteins/metabolism , Genes, MHC Class I , I-kappa B Proteins , NF-kappa B/metabolism , Receptors, Steroid , Repressor Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Adenoviridae , Animals , COUP Transcription Factor II , COUP Transcription Factors , Cell Line, Transformed , Cell Nucleus/metabolism , Cell Transformation, Viral , DNA-Binding Proteins/pharmacology , Down-Regulation , Interleukin-1/pharmacology , NF-KappaB Inhibitor alpha , NF-kappa B/antagonists & inhibitors , NF-kappa B p50 Subunit , Protein Transport/drug effects , Rats , Transcription Factor RelA , Tumor Necrosis Factor-alpha/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...