Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
FASEB J ; 37(3): e22804, 2023 03.
Article in English | MEDLINE | ID: mdl-36753402

ABSTRACT

Normal human tissue is a critical reference control in biomedical research. However, the type of tissue donor can significantly affect the underlying biology of the samples. We investigated the impact of tissue donor source type by performing transcriptomic analysis on healthy kidney tissue from three donor source types: cadavers, organ donors, and normal-adjacent tissue from surgical resections of clear cell renal cell carcinomas, and we compared the gene expression profiles to those of clear cell renal cell carcinoma samples. Comparisons among the normal samples revealed general similarity, with notable differences in gene expression pathways involving immune system and inflammatory processes, response to extracellular stimuli, ion transport, and metabolism. When compared to tumors, the transcriptomic profiles of the normal adjacent tissue were highly similar to the profiles from cadaveric and organ donor tissue samples, arguing against the presence of a field cancerization effect in clear cell renal cell carcinoma. We conclude that all three normal source types are suitable for reference kidney control samples, but important differences must be noted for particular research areas and tissue banking strategies.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Transcriptome , Kidney , Kidney Neoplasms/genetics , Tissue Donors
2.
Mol Cancer Ther ; 11(7): 1565-75, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22504949

ABSTRACT

T-cell acute lymphoblastic leukemias (T-ALL) and lymphomas are aggressive hematologic cancers frequently associated with activating mutations in NOTCH1. Early studies identified NOTCH1 as an attractive therapeutic target for the treatment of T-ALL through the use of γ-secretase inhibitors (GSI). Here, we characterized the interaction between PF-03084014, a clinically relevant GSI, and dexamethasone in preclinical models of glucocorticoid-resistant T-ALL. Combination treatment of the GSI PF-03084014 with glucocorticoids induced a synergistic antileukemic effect in human T-ALL cell lines and primary human T-ALL patient samples. Mechanistically PF-03084014 plus glucocorticoid treatment induced increased transcriptional upregulation of the glucocorticoid receptor and glucocorticoid target genes. Treatment with PF-03084014 and glucocorticoids in combination was highly efficacious in vivo, with enhanced reduction of tumor burden in a xenograft model of T-ALL. Finally, glucocorticoid treatment effectively reversed PF-03084014-induced gastrointestinal toxicity via inhibition of goblet cell metaplasia. These results warrant the analysis of PF-03084014 and glucocorticoids in combination for the treatment of glucocorticoid-resistant T-ALL.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Glucocorticoids/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Tetrahydronaphthalenes/pharmacology , Valine/analogs & derivatives , Amyloid Precursor Protein Secretases/metabolism , Animals , Cell Line, Tumor , Cluster Analysis , Dexamethasone/administration & dosage , Drug Resistance, Neoplasm/drug effects , Female , Gene Expression Profiling , Gene Expression Regulation, Leukemic/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/metabolism , Tetrahydronaphthalenes/chemistry , Tetrahydronaphthalenes/toxicity , Valine/chemistry , Valine/pharmacology , Valine/toxicity
3.
Eur J Cancer ; 47(15): 2256-64, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21852114

ABSTRACT

This phase I study (ClinicalTrials.gov ID: NCT00424632) evaluated the safe dose, pharmacokinetics, and pharmacodynamics of the aurora kinase A and B inhibitor, PF-03814735. Patients with advanced solid tumours received oral, once-daily (QD) PF-03814735 on Schedule A: days 1-5 (5-100mg); or Schedule B: days 1-10 (40-60mg) of 21-day cycles. Fifty-seven patients were treated: 32 and 25 on Schedules A and B, respectively. Dose-limiting toxicities were: febrile neutropenia (Schedule A); and increased levels of aspartate amino transferase, left ventricular dysfunction, and prolonged low-grade neutropenia (Schedule B). Maximum tolerated doses were 80mg QD (Schedule A) and 50mg QD (Schedule B). Common treatment-related adverse events were mainly mild to moderate and included diarrhoea, fatigue, nausea, and vomiting. Nineteen patients achieved stable disease, which was prolonged in four cases. PF-03814735 was rapidly absorbed and demonstrated linear pharmacokinetics up to 100mg QD; mean terminal half-life ranged from 14.4 to 23.6h. Aurora B activity, assessed by histone H3 phosphorylation in mitotic cells, decreased in tumour tissue from 10/12 patients evaluated (range: -70% to -3%). (18)F-fluorodeoxyglucose positron emission tomography demonstrated metabolic responses in only 1/21 patients. PF-03814735 was generally well tolerated with manageable toxicities, and a recommended phase II dose could be established for both schedules. Aurora B activity was inhibited in tumour tissue, but clinical or metabolic antitumour activity was limited.


Subject(s)
Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Neoplasms/drug therapy , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Aged , Aurora Kinase A , Aurora Kinase B , Aurora Kinases , Belgium , Biopsy , Dose-Response Relationship, Drug , Female , Fluorodeoxyglucose F18 , Heterocyclic Compounds, 3-Ring/adverse effects , Histones/metabolism , Humans , Immunohistochemistry , Linear Models , Male , Maximum Tolerated Dose , Middle Aged , Mitosis/drug effects , Models, Biological , Molecular Targeted Therapy , Neoplasms/diagnostic imaging , Neoplasms/enzymology , Phosphorylation , Positron-Emission Tomography , Protein Kinase Inhibitors/adverse effects , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/adverse effects , Radiopharmaceuticals , Tennessee , Treatment Outcome
4.
J Med Chem ; 53(12): 4615-22, 2010 Jun 24.
Article in English | MEDLINE | ID: mdl-20481595

ABSTRACT

This paper describes the design and synthesis of novel, ATP-competitive Akt inhibitors from an elaborated 3-aminopyrrolidine scaffold. Key findings include the discovery of an initial lead that was modestly selective and medicinal chemistry optimization of that lead to provide more selective analogues. Analysis of the data suggested that highly lipophilic analogues would likely suffer from poor overall properties. Central to the discussion is the concept of optimization of lipophilic efficiency and the ability to balance overall druglike propeties with the careful control of lipophilicity in the lead series. Discovery of the nonracemic amide series and subsequent modification produced an advanced analogue that performed well in advanced preclinical assays, including xenograft tumor growth inhibition studies, and this analogue was nominated for clinical development.


Subject(s)
Adenosine Triphosphate/physiology , Amides/chemical synthesis , Aminoquinolines/chemical synthesis , Antineoplastic Agents/chemical synthesis , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Amides/pharmacokinetics , Amides/pharmacology , Aminoquinolines/pharmacokinetics , Aminoquinolines/pharmacology , Animals , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Crystallography, X-Ray , Dogs , Mice , Models, Molecular , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/pharmacology , Rats , Stereoisomerism , Structure-Activity Relationship , Xenograft Model Antitumor Assays
5.
Mol Cancer Ther ; 9(4): 883-94, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20354118

ABSTRACT

The Aurora family of highly related serine/threonine kinases plays a key role in the regulation of mitosis. Aurora1 and Aurora2 play important but distinct roles in the G(2) and M phases of the cell cycle and are essential for proper chromosome segregation and cell division. Overexpression and amplification of Aurora2 have been reported in different tumor types, including breast, colon, pancreatic, ovarian, and gastric cancer. PF-03814735 is a novel, potent, orally bioavailable, reversible inhibitor of both Aurora1 and Aurora2 kinases that is currently in phase I clinical trials for the treatment of advanced solid tumors. In intact cells, the inhibitory activity of PF-03814735 on the Aurora1 and Aurora2 kinases reduces levels of phospho-Aurora1, phosphohistone H3, and phospho-Aurora2. PF-03814735 produces a block in cytokinesis, resulting in inhibition of cell proliferation and the formation of polyploid multinucleated cells. Although PF-03814735 produces significant inhibition of several other protein kinases, the predominant biochemical effects in cellular assays are consistent with inhibition of Aurora kinases. Once-daily oral administration of PF-03814735 to mice bearing human xenograft tumors produces a reduction in phosphohistone H3 in tumors at doses that are tolerable and that result in significant inhibition of tumor growth. The combination of PF-03814735 and docetaxel in xenograft mouse tumor models shows additive tumor growth inhibition. These results support the clinical evaluation of PF-03814735 in cancer patients. Mol Cancer Ther; 9(4); 883-94. (c)2010 AACR.


Subject(s)
Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/therapeutic use , Neoplasms/drug therapy , Neoplasms/enzymology , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/pharmacokinetics , Pyrimidines/therapeutic use , Administration, Oral , Animals , Aurora Kinases , Biological Availability , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Heterocyclic Compounds, 3-Ring/administration & dosage , Heterocyclic Compounds, 3-Ring/pharmacology , Histones/metabolism , Humans , Mice , Mice, Nude , Neoplasms/pathology , Phosphorylation/drug effects , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Substrate Specificity/drug effects , Xenograft Model Antitumor Assays
6.
Cancer Gene Ther ; 11(8): 555-69, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15232601

ABSTRACT

A potentially promising treatment of metastatic cancer is the systemic delivery of oncolytic adenoviruses. This requires engineering viruses which selectively replicate in tumors. We have constructed such an oncolytic adenovirus, OAS403, in which two early region genes are under the control of tumor-selective promoters that play a role in two key pathways involved in tumorigenesis. The early region E1A is controlled by the promoter for the E2F-1 gene, a transcription factor that primarily upregulates genes for cell growth. The E4 region is under control of the promoter for human telomerase reverse transcriptase, a gene upregulated in most cancer cells. OAS403 was evaluated in vitro on a panel of human cells and found to elicit tumor-selective cell killing. Also, OAS403 was less toxic in human hepatocyte cultures, as well as in vivo when compared to an oncolytic virus that lacked selective E4 control. A single intravenous injection of 3 x 10(12) vp/kg in a Hep3B xenograft mouse tumor model led to significant antitumor efficacy. Additionally, systemic administration in a pre-established LNCaP prostate tumor model resulted in over 80% complete tumor regressions at a tolerable dose. Vector genome copy number was measured in tumors and livers at various times following tail vein injection and showed a selective time-dependent increase in tumors but not livers over 29 days. Furthermore, efficacy was significantly improved when OAS403 treatment was combined with doxorubicin. This virus holds promise for the treatment of a broad range of human cancers including metastatic disease.


Subject(s)
Adenoviridae/genetics , Neoplasms/therapy , Adenoviridae/metabolism , Animals , DNA-Binding Proteins , Doxorubicin/therapeutic use , Genetic Vectors/administration & dosage , Hepatocytes/metabolism , Humans , Inhibitory Concentration 50 , Injections , Mice , Mice, SCID , Neoplasm Metastasis , Neoplasms/genetics , Neoplasms/metabolism , Promoter Regions, Genetic , Telomerase/genetics , Telomerase/metabolism , Virus Replication/genetics , Xenograft Model Antitumor Assays
7.
Cancer Gene Ther ; 11(2): 92-102, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14730331

ABSTRACT

Oncolytic adenoviral vectors selectively replicate in and lyse human tumor cells, providing a promising means for targeted tumor destruction. However, oncolytic vectors have limited capacity for incorporation of additional genetic material that could encode therapeutic transgenes and/or transcriptional regulatory control elements to augment the efficacy and/or safety of the vector. Therefore, we hypothesized that coadministration of an oncolytic vector with a replication-defective, gutless adenoviral vector encoding a therapeutic transgene would result in replication of both vectors within a tumor and potentiate antitumor efficacy relative to the use of either vector alone. We constructed gutless vectors encoding the murine granulocyte-macrophage colony-stimulating factor (AGVmGMF) or human tumor necrosis factor alpha-related apoptosis-inducing ligand (AGVhTRAIL) gene and tested the ability of these vectors to augment the efficacy of an oncolytic vector (Ar6pAE2fE3F) in a potentiating vector strategy. In Hep3B cells in vitro, cotreatment with Ar6pAE2fE3F increased transgene expression from AGVhTRAIL and permitted replication of AGVhTRAIL, suggesting that an oncolytic vector can propagate gutless vector spread in vivo. In pre-established Hep3B xenograft tumors, neither gutless vector alone inhibited tumor growth; however, coadministration of AGVmGMF or AGVhTRAIL with Ar6pAE2fE3F significantly reduced tumor growth relative to Ar6pAE2fE3F alone. Additionally, use of AGVhTRAIL with Ar6pAE2fE3F increased the number of complete or partial tumor regressions observed at study end. These data provide evidence that coadministration of an oncolytic vector with a gutless vector holds promise for potentiating tumor ablation efficacy.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Membrane Glycoproteins/genetics , Neoplasms, Experimental/therapy , Tumor Necrosis Factor-alpha/genetics , Animals , Antineoplastic Agents/therapeutic use , Apoptosis Regulatory Proteins , Cell Line, Tumor , Female , Genetic Vectors/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Helper Viruses/genetics , Humans , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/pharmacology , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
8.
Cancer Res ; 63(7): 1490-9, 2003 Apr 01.
Article in English | MEDLINE | ID: mdl-12670895

ABSTRACT

The use of oncolytic adenoviruses as a cancer therapeutic is dependent on the lytic properties of the viral life cycle, and the molecular differences between tumor cells and nontumor cells. One strategy for achieving safe and efficacious adenoviral therapies is to control expression of viral early gene(s) required for replication with tumor-selective promoter(s), particularly those active in a broad range of cancer cells. The retinoblastoma tumor suppressor protein (Rb) pathway is dysregulated in a majority of human cancers. The human E2F-1 promoter has been shown to be selectively activated/derepressed in tumor cells with a defect in the Rb pathway. Ar6pAE2fE3F and Ar6pAE2fF are oncolytic adenoviral vectors (with and without the viral E3 region, respectively) that use the tumor-selective E2F-1 promoter to limit expression of the viral E1A transcription unit, and, thus, replication, to tumor cells. We demonstrate that the antitumor activity of Ar6pAE2fF in vitro and in vivo is dependent on the E2F-1 promoter driving E1A expression in Rb pathway-defective cells, and furthermore, that its oncolytic activity is enhanced by viral replication. Selective oncolysis by Ar6pAE2fF was dependent on the presence of functional E2F binding sites in the E2F-1 promoter, thus linking antitumor viral activity to the Rb pathway. Potent antitumor efficacy was demonstrated with Ar6pAE2fF and Ar6pAE2fE3F in a xenograft model following intratumoral administration. Ar6pAE2fF and Ar6pAE2fE3F were compared with Addl1520, which is reported to be molecularly identical to an E1B-55K deleted vector currently in clinical trials. These vectors were compared in in vitro cytotoxicity and virus production assays, after systemic delivery in an in vivo E1A-related hepatotoxicity model, and in a mouse xenograft tumor model after intratumoral administration. Our results support the use of oncolytic adenoviruses using tumor-selective promoter(s) that are activated or derepressed in tumor cells by virtue of a particular defective pathway, such as the Rb pathway.


Subject(s)
Adenoviridae/physiology , Adenovirus E1A Proteins/biosynthesis , Cell Cycle Proteins , DNA-Binding Proteins , Neoplasms/virology , Retinoblastoma Protein/physiology , Transcription Factors/genetics , Adenoviridae/genetics , Adenoviridae/metabolism , Adenovirus E1A Proteins/genetics , Animals , Binding Sites , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Carcinoma, Hepatocellular/virology , Cytopathogenic Effect, Viral/physiology , E2F Transcription Factors , E2F1 Transcription Factor , Genetic Vectors/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/therapy , Liver Neoplasms/virology , Male , Mice , Mice, SCID , Neoplasms/genetics , Neoplasms/therapy , Promoter Regions, Genetic , Retinoblastoma Protein/metabolism , Signal Transduction/physiology , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
9.
Proc Natl Acad Sci U S A ; 99(6): 3770-5, 2002 Mar 19.
Article in English | MEDLINE | ID: mdl-11867754

ABSTRACT

It has been argued that genetic instability is required to generate the myriad mutations that fuel tumor initiation and progression and, in fact, patients with heritable cancer susceptibility syndromes harbor defects in specific genes that normally maintain DNA integrity. However, the vast majority of human cancers arise sporadically, in the absence of deficiencies in known "mutator" genes. We used a cII-based mutation detection assay to show that the mean frequency of forward mutations in primary mammary adenocarcinomas arising in mouse mammary tumor virus-c-erbB2 transgenic mice harboring multiple copies of the lambda bacteriophage genome was significantly higher than in aged-matched, wild-type mammary tissue. Analysis of the cII mutational spectrum within the mammary tumor genomic DNA demonstrated a >6-fold elevation in transversion mutation frequency, resulting in a highly unusual inversion of the transition/transversion ratio characteristic of normal epithelium; frameshift mutation frequencies were unaltered. Arising oncogenic point mutations within the c-erbB2 transgene of such tumors were predominantly transversions as well. Data from this model system support the notion that elaboration of a mutator phenotype is a consequential event in breast cancer and suggest that a novel DNA replication/repair gene is a relatively early mutational target in c-erbB2-induced mammary tumorigenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , DNA Damage/genetics , Mammary Neoplasms, Animal/genetics , Mutagenesis/genetics , Receptor, ErbB-2/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/virology , Aging , Amino Acid Sequence , Animals , Bacteriophage lambda/genetics , DNA Mutational Analysis , Female , Fibroblasts , Gene Frequency/genetics , Kinetics , Male , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Animal/virology , Mammary Tumor Virus, Mouse/physiology , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation, Missense/genetics , Receptor, ErbB-2/genetics , Transgenes/genetics
10.
Evolution ; 42(3): 626-630, 1988 May.
Article in English | MEDLINE | ID: mdl-28564009
SELECTION OF CITATIONS
SEARCH DETAIL
...