Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
NPJ Precis Oncol ; 2: 25, 2018.
Article in English | MEDLINE | ID: mdl-30456308

ABSTRACT

Hepatocellular carcinoma (HCC) develops in the context of chronic inflammatory liver disease and has an extremely poor prognosis. An immunosuppressive tumor microenvironment may contribute to therapeutic failure in metastatic HCC. Here, we identified unique molecular signatures pertaining to HCC disease progression and tumor immunity by analyzing genome-wide RNA-Seq data derived from HCC patient tumors and non-tumor cirrhotic tissues. Unsupervised clustering of gene expression data revealed a gradual suppression of local tumor immunity that coincided with disease progression, indicating an increasingly immunosuppressive tumor environment during HCC disease advancement. IHC examination of the spatial distribution of CD8+ T cells in tumors revealed distinct intra- and peri-tumoral subsets. Differential gene expression analysis revealed an 85-gene signature that was significantly upregulated in the peri-tumoral CD8+ T cell-excluded tumors. Notably, this signature was highly enriched with components of underlying extracellular matrix, fibrosis, and epithelial-mesenchymal transition (EMT). Further analysis condensed this signature to a core set of 23 genes that are associated with CD8+ T cell localization, and were prospectively validated in an independent cohort of HCC specimens. These findings suggest a potential association between elevated fibrosis, possibly modulated by TGF-ß, PDGFR, SHH or Notch pathway, and the T cell-excluded immune phenotype. Indeed, targeting fibrosis using a TGF-ß neutralizing antibody in the STAM™ model of murine HCC, we found that ameliorating the fibrotic environment could facilitate redistribution of CD8+ lymphocytes into tumors. Our results provide a strong rationale for utilizing immunotherapies in HCC earlier during treatment, potentially in combination with anti-fibrotic therapies.

2.
Mol Cell Biol ; 9(11): 4903-13, 1989 Nov.
Article in English | MEDLINE | ID: mdl-2601700

ABSTRACT

Human neuroblastoma cells often carry amplified DNA encompassing the gene N-myc. Amplified N-myc has been found localized in "double minutes" in direct tumor cell preparations. In contrast, later passages carried amplified N-myc almost exclusively within a single homogeneously staining chromosomal region located at a chromosomal site different from the normal location of N-myc. We used pulsed field gel electrophoresis to define the structural arrangement of the amplified DNA. Long-range mapping was facilitated by the presence of several sites for rare cutting restriction endonucleases in the 5' region of N-myc. Amplified DNAs of different neuroblastoma cell lines were heterogeneous in size and had undergone recombination at various distances from N-myc. N-myc occupied a central position within the amplified DNA, and in no case was the coding region affected by recombination. Among neuroblastoma cells, varying proportions of amplified DNA (in some instances close to 100%) consisted of multiple tandem arrays of DNA segments ranging in size from 100 to 700 kilobase pairs. Tumor cells with low degrees of amplification revealed regions of amplified DNA in excess of 1,500 kilobase pairs without apparent rearrangement. Our observations, in concert with the cytogenetic findings, suggest a model of gene amplification which involves unscheduled DNA replication, recombination, and formation of extrachromosomal DNA followed by integration into a chromosome and subsequent in situ multiplication. The central position which N-myc occupies within the amplified sequences and the lack of recombination within the coding region of N-mc indicate that N-myc rather than other genetic information provides the selective advantage for retention of the amplified DNA.


Subject(s)
Gene Amplification , Multigene Family , Oncogenes , Recombination, Genetic , DNA Replication , DNA, Neoplasm/genetics , Gene Rearrangement , Humans , Models, Genetic , Neuroblastoma , Repetitive Sequences, Nucleic Acid , Restriction Mapping , Tumor Cells, Cultured
3.
Cancer Res ; 55(22): 5366-9, 1995 Nov 15.
Article in English | MEDLINE | ID: mdl-7585602

ABSTRACT

Human neuroblastoma cells often have deletions of the distal short arm of chromosome 1 (1p). Earlier studies using chromosome analysis had suggested that the 1p deletion is correlated with a poor survival chance for the patient. We have reevaluated this possibility by analyzing 51 neuroblastomas for loss of heterozygosity (LOH) at 1p. We detected LOH in 32% of the cases. LOH did not correlate with the age of the patients at diagnosis or with tumor stage but was correlated significantly with amplification of the MYCN proto-oncogene. Nine of 10 MYCN-amplified tumors had deletions in 1p (P < 0.001). Survival chances of patients with tumors carrying MYCN amplification together with the deletion at 1p were decreased significantly (eight of nine affected patients died) compared with a patient group without any of these aberrations (P < 0.001). However, the deletion of 1p alone without MYCN amplification was not associated with a poor outcome compared with patients who had neither deletion nor amplification (only two of eight affected patients died; P = 0.803). From these data we conclude that 1p deletions are not reliable markers to determine a patient's prognosis. They may, however, identify a subgroup of neuroblastomas in which MYCN is amplified readily, resulting in rapid tumor progression.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1 , Neuroblastoma/genetics , Adult , Biomarkers , Child , Child, Preschool , Female , Humans , Infant , Male , Neoplasm Staging , Neuroblastoma/mortality , Polymorphism, Restriction Fragment Length , Prognosis , Proto-Oncogene Mas , Proto-Oncogenes
4.
Cancer Res ; 55(23): 5504-6, 1995 Dec 01.
Article in English | MEDLINE | ID: mdl-7585622

ABSTRACT

Mice heterozygous for the dominant Min mutation in their Apc gene develop multiple intestinal neoplasia. Analogously, family members from familial adenomatous polyposis kindreds inheriting mutations in their human APC homologue develop a similar phenotype. Quantitative trait loci studies have identified the Mom1 locus (for modifier of Min-1), which is responsible for part of the genetic variability in polyp number found among inbred mouse strains. The secretory type II phospholipase [nonpancreatic Pla2s (type II Pla2s or Pla2s-II)] has been demonstrated to be a candidate for Mom1, and a mutation in Pla2s-II in mice carrying the Min mutation has been proposed to account for an increased polyp number compared to mice without the Pla2s-II mutation. In this study, we have mapped the chromosomal position of the human homologue of Pla2s-II. We have identified 3 mega-yeast artificial chromosomes that carry PLA2S-II and localized one of them by fluorescence in situ hybridization to the border between 1p35 and 1p36.1. The presence of the microsatellite marker D1S199 in all three clones integrates PLA2S-II into different genetic maps. This highly polymorphic CA repeat D1S199 has previously been shown by us to identify loss of heterozygosity in 48% of sporadic colorectal tumors, indicating that the human homologue of the Pla2s-II/Mom1 locus might be related to human colorectal cancer.


Subject(s)
Chromosomes, Human, Pair 1/chemistry , Phospholipases A/genetics , Base Sequence , Chromosome Mapping , DNA Primers/chemistry , Group II Phospholipases A2 , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Phospholipases A2
5.
Cancer Res ; 60(21): 6134-41, 2000 Nov 01.
Article in English | MEDLINE | ID: mdl-11085537

ABSTRACT

Treatment of metastatic prostate cancer with androgen-ablation often elicits dramatic tumor regressions, but the response is rarely complete, making clinical recurrence inevitable with time. To gain insight into therapy-related progression, changes in gene expression that occurred following androgen-deprivation of an androgen-dependent prostate tumor xenograft, CWR22, and the emergence of an androgen-independent tumor, CWR22-R, were monitored using microarray analysis. Androgen-deprivation resulted in growth arrest of CWR22 cells, as evidenced by decreased expression of genes encoding cell cycle components and basal cell metabolism, respiration and transcription, and the induced expression of putative negative regulatory genes that may act to sustain cells in a nonproliferative state. Evolution of androgen-independent growth and proliferation, represented by CWR22-R, was associated with a reentry into active cell cycle and the up-regulation of several genes that were expressed at low levels or absent in the androgen-dependent tumor. Androgen repletion to mice bearing androgen-independent CWR22-R tumors induced, augmented, or repressed the expression of a number of genes. Expression of two of these genes, the calcium-binding protein S100P and the FK-506-binding protein FKBP51, was decreased following androgen-deprivation, subsequently reexpressed in CWR22-R at levels comparable with CWR22, and elevated further upon treatment with androgens. The dysregulated behavior of these genes is analogous to other androgen-dependent genes, e.g., prostate-specific antigen and human kallikrein 2, which are commonly reexpressed in androgen-independent disease in the absence of androgens. Other androgen-responsive genes whose expression decreased during androgen-deprivation and whose expression remained decreased in CWR22 were also identified in CWR22-R. These results imply that evolution to androgen-independence is due, in part, to reactivation of the androgen-response pathway in the absence of androgens, but that this reactivation is probably incomplete.


Subject(s)
Androgens/physiology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Neoplasms, Hormone-Dependent/genetics , Prostatic Neoplasms/genetics , Androgens/deficiency , Animals , Female , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplasms, Hormone-Dependent/metabolism , Oligonucleotide Array Sequence Analysis , Prostatic Neoplasms/metabolism , Transplantation, Heterologous , Up-Regulation
6.
Leukemia ; 30(7): 1493-501, 2016 07.
Article in English | MEDLINE | ID: mdl-27044711

ABSTRACT

Chronic myeloid leukemia (CML) patients who relapse on imatinib due to acquired ABL1 kinase domain mutations are successfully treated with second-generation ABL1-tyrosine kinase inhibitors (ABL-TKIs) such as dasatinib, nilotinib or ponatinib. However, ~40% of relapsed patients have uncharacterized BCR-ABL1 kinase-independent mechanisms of resistance. To identify these mechanisms of resistance and potential treatment options, we generated ABL-TKI-resistant K562 cells through prolonged sequential exposure to imatinib and dasatinib. Dual-resistant K562 cells lacked BCR-ABL1 kinase domain mutations, but acquired other genomic aberrations that were characterized by next-generation sequencing and copy number analyses. Proteomics showed that dual-resistant cells had elevated levels of FOXO1, phospho-ERK and BCL-2, and that dasatinib no longer inhibited substrates of the PI3K/AKT pathway. In contrast to parental cells, resistant cells were sensitive to growth inhibition and apoptosis induced by the class I PI3K inhibitor, GDC-0941 (pictilisib), which also induced FOXO1 nuclear translocation. FOXO1 was elevated in a subset of primary specimens from relapsed CML patients lacking BCR-ABL1 kinase domain mutations, and these samples were responsive to GDC-0941 treatment ex vivo. We conclude that elevated FOXO1 contributes to BCR-ABL1 kinase-independent resistance experienced by these CML patients and that PI3K inhibition coupled with BCR-ABL1 inhibition may represent a novel therapeutic approach.


Subject(s)
Drug Resistance, Neoplasm , Forkhead Box Protein O1/physiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Protein Kinase Inhibitors/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Dasatinib/pharmacology , Drug Tolerance , Forkhead Box Protein O1/analysis , Fusion Proteins, bcr-abl/antagonists & inhibitors , Humans , Imatinib Mesylate/pharmacology , Indazoles/pharmacology , K562 Cells , Phosphoinositide-3 Kinase Inhibitors , Sulfonamides/pharmacology , Tumor Cells, Cultured
7.
Oncogene ; 35(50): 6403-6415, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27270421

ABSTRACT

Key molecular drivers that underlie transformation of colonic epithelium into colorectal adenocarcinoma (CRC) are well described. However, the mechanisms through which clinically targeted pathways are activated during CRC progression have yet to be elucidated. Here, we used an integrative genomics approach to examine CRC progression. We used laser capture microdissection to isolate colonic crypt cells, differentiated surface epithelium, adenomas, carcinomas and metastases, and used gene expression profiling to identify pathways that were differentially expressed between the different cell types. We identified a number of potentially important transcriptional changes in developmental and oncogenic pathways, and noted a marked upregulation of EREG in primary and metastatic cancer cells. We confirmed this pattern of gene expression by in situ hybridization and observed staining consistent with autocrine expression in the tumor cells. Upregulation of EREG during the adenoma-carcinoma transition was associated with demethylation of two key sites within its promoter, and this was accompanied by an increase in the levels of epidermal growth factor receptor (EGFR) phosphorylation, as assessed by reverse-phase protein analysis. In CRC cell lines, we demonstrated that EREG demethylation led to its transcriptional upregulation, higher levels of EGFR phosphorylation, and sensitization to EGFR inhibitors. Low levels of EREG methylation in patients who received cetuximab as part of a phase II study were associated with high expression of the ligand and a favorable response to therapy. Conversely, high levels of promoter methylation and low levels of EREG expression were observed in tumors that progressed after treatment. We also noted an inverse correlation between EREG methylation and expression levels in several other cancers, including those of the head and neck, lung and bladder. Therefore, we propose that upregulation of EREG expression through promoter demethylation might be an important means of activating the EGFR pathway during the genesis of CRC and potentially other cancers.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , Epiregulin/genetics , ErbB Receptors/physiology , Promoter Regions, Genetic , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Decitabine , Disease Progression , ErbB Receptors/antagonists & inhibitors , Humans , Phosphorylation
8.
Oncogene ; 7(4): 807-9, 1992 Apr.
Article in English | MEDLINE | ID: mdl-1565478

ABSTRACT

Human neuroblastoma cells often carry cytogenetic abnormalities signaling amplification of the gene N-myc. In most cell lines amplified N-myc is localized in homogeneously staining regions (HSRs). Varying proportions of the amplified DNA consist of multiple tandem arrays of DNA segments encompassing N-myc. Here we report the cloning and sequencing of a DNA breakpoint which represents the joint of the tandem repeats of a 280-kb amplicon of neuroblastoma line NMB. The breakpoint is located in the first intron of the N-myc gene and leads to the deletion of the 5' region of N-myc in this 20-copy amplicon. The representation of DNA derived from the non-N-myc part of the novel joint in the different amplicons suggests that an increase in N-myc copy numbers involves a multistep process proceeding from large 'precursors' to smaller multicopy amplicons.


Subject(s)
Gene Amplification , Genes, myc , Neuroblastoma/genetics , Proto-Oncogene Proteins c-myc/genetics , Base Sequence , In Vitro Techniques , Molecular Sequence Data , Recombination, Genetic , Restriction Mapping , Time Factors , Tumor Cells, Cultured
9.
Oncogene ; 11(7): 1357-62, 1995 Oct 05.
Article in English | MEDLINE | ID: mdl-7478557

ABSTRACT

Cytogenetic and molecular analyses of colorectal cancer cells have revealed deletions at 1p as prominent alterations, suggesting that genetic information on the short arm of chromosome 1 has a role in tumorigenesis. In this study we have used 33 microsatellite markers to fine map deletions at 1p in primary colorectal carcinomas. We found 1p-deletions in 84% of the cases (31/37). High frequencies of loss of heterozygosity (LOH), often the result of small independent interstitial deletions in the same tumor, defined three regions, that may harbor genetic information relevant for colorectal cancer: (i) region A between D1S243 and D1S468 (7cM; 1p36.3); (ii) region B between D1S436 and D1S199 (7cM; 1p35.1-36.31) and (iii) region C between D1S496 and D1S255 (1cM; 1p34.2-35). In addition we identified seven cell lines with LOH at 1p, all of which have deletions that span at least from the distal border of region A to the proximal border of region C.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 1 , Colorectal Neoplasms/genetics , Chromosome Mapping , Colorectal Neoplasms/pathology , Genes, Tumor Suppressor , Heterozygote , Humans , Tumor Cells, Cultured
10.
Oncogene ; 10(6): 1095-101, 1995 Mar 16.
Article in English | MEDLINE | ID: mdl-7700634

ABSTRACT

Cytogenetic analyses and molecular deletion studies of human neuroblastomas have indicated the chromosomal bands 1p36.1-1p36.2 as a location of genetic information which may be involved in tumorigenesis. To define this putative neuroblastoma locus in more detail we have analysed cell lines with alterations of distal 1p. Here we show, by fluorescence in situ hybridization (FISH), that cell line NGP has a reciprocal 1;15 translocation. Loci D1S214/D1S96 could be shown to map telomeric/distal, D1S228 centromeric/proximal to the break. We have identified yeast artificial chromosomes (YACs) that cover the break and map to D1S160 and D1S244. This chromosomal position is within the smallest region of overlap (SRO) found in neuroblastoma tumors (Weith et al., 1989; Caron et al., 1993; Schleiermacher et al., 1994) and within the region of a constitutional interstitial deletion of a neuroblastoma patient (Biegel et al., 1993). Mapping studies with FISH revealed that the translocation is associated with duplication of DNA. It appears, as if the subchromosomal region we describe here is a good candidate for harboring the postulated neuroblastoma suppressor-gene.


Subject(s)
Chromosomes, Human, Pair 15 , Chromosomes, Human, Pair 1 , DNA, Neoplasm/genetics , Genes, Retinoblastoma , Translocation, Genetic , Base Sequence , Chromosomes, Artificial, Yeast , DNA Primers , Humans , In Situ Hybridization, Fluorescence , Molecular Sequence Data , Multigene Family , Tumor Cells, Cultured
11.
Oncogene ; 17(15): 2009-12, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9788445

ABSTRACT

There is good evidence now that the secretory type II phospholipase A2 (Pla2g2a) gene represents the Mom1 locus, a genetic modifier of tumor resistance in the multiple intestinal neoplasia (Min) mouse. Previously we have mapped the human homolog PLA2G2A to 1p35-36.1 within a region that is the target of frequent deletions in sporadic colorectal tumors. Here we show 64% loss of heterozygosity (LOH) at the PLA2G2A locus in primary tumors. We studied PLA2G2A expression in both colorectal tumor cell lines and normal mucosa. Most of the lines lacked detectable PLA2G2A transcripts by Northern analysis. Large differences in expression were seen among normal mucosa of different patients with sporadic tumors. We analysed the coding region of PLA2G2A in eight colorectal cancer cell lines with hemizygous deletion at 1p35-36/PLA2G2A, in none we did detect a mutation. Biallelic expression of PLA2G2A was observed in a cell line heterozygous for an exon 3 polymorphism, rendering unlikely that imprinting is a pathway participating in the loss of PLA2G2A function. It remains uncertain if PLA2G2A, in particular its apparent lack of expression in tumor cells, might be a factor in human colorectal tumorigenesis.


Subject(s)
Colon/enzymology , Colorectal Neoplasms/enzymology , Intestinal Mucosa/enzymology , Phospholipases A/genetics , Animals , Base Sequence , Colorectal Neoplasms/pathology , DNA Primers , Group II Phospholipases A2 , Homozygote , Humans , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phospholipases A2 , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Cells, Cultured
12.
Oncogene ; 10(6): 1081-6, 1995 Mar 16.
Article in English | MEDLINE | ID: mdl-7700632

ABSTRACT

Amplification of the MYCN gene is a well documented genetic alteration of aggressively growing human neuroblastomas. Through cytogenetic studies we have identified neuroblastoma cell lines which, in addition to amplified MYCN, carry amplified DNA not harbouring MYCN. In situ hybridization of biotinylated total genomic DNA to metaphase chromosomes of normal human lymphocytes by reverse genomic hybridization revealed the amplified DNA to be derived from chromosome 12 band q13-14. Subsequent filter analyses showed a 20- to 40-fold amplification of the MDM2 gene, located at 12q13-14, both in three cell lines and in an original tumor, in addition to amplified MYCN. As the apparent consequence of amplification abundant MDM2 protein was present, a part of which was complexed with p53.


Subject(s)
Gene Amplification , Genes, myc , Neoplasm Proteins/genetics , Neuroblastoma/genetics , Nuclear Proteins , Proto-Oncogene Proteins/genetics , Chromosomes, Human, Pair 12 , Humans , Proto-Oncogene Proteins c-mdm2 , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
13.
Eur J Cancer ; 31A(4): 520-3, 1995.
Article in English | MEDLINE | ID: mdl-7576957

ABSTRACT

Amplification of the MYCN gene is frequently seen either in extrachromosomal double minutes (DMs) or in homogeneously staining regions (HSRs) of aggressively growing neuroblastomas. Total genomic DNA from cell line LS, from early passages of the same line and from original tumour material was biotinylated and hybridised to metaphase chromosomes of normal human lymphocytes. The reverse genomic hybridisation revealed the amplified DNA to be derived both from chromosome 2p23-24, which is the position of MYCN, and from chromosome 12 band q13-14. The MDM2 gene, located at 12q13-14, was found amplified both in early and late passages of LS, in addition to amplified MYCN. Amplification units of MYCN and MDM2 appear first to develop within DMs, which then integrate into different chromosomes to develop to HSRs.


Subject(s)
Chromosomes, Human, Pair 12/genetics , Genes, myc , Neuroblastoma/genetics , Nuclear Proteins , Proto-Oncogene Proteins/genetics , Chromosomes, Human, Pair 2 , Gene Amplification , Humans , In Situ Hybridization, Fluorescence , Proto-Oncogene Proteins c-mdm2 , Tumor Cells, Cultured
14.
Eur J Cancer ; 31A(4): 527-30, 1995.
Article in English | MEDLINE | ID: mdl-7576959

ABSTRACT

Band 1p36.1-1p36.2 is frequently involved in chromosomal aberrations of neuroblastoma cells, and therefore thought to harbour genetic information which may be involved in tumorigenesis. To map this putative neuroblastoma locus, we screened neuroblastoma cell lines for reciprocal translocations at 1p36.1-2 which may signal the site of an affected gene. We identified a reciprocal 1;15 translocation in cell line NGP by fluorescence in situ hybridisation (FISH). As a strategy to clone the translocation breakpoint, we isolated yeast artificial chromosomes (YACs) specific for loci at 1p36. Screening of cell line NGP by FISH identified a YAC, 1050 kbp in size, which hybridised to both derivative 1;15 and 15;1 chromosomes. We conclude that this YAC, which maps to D1S160, covers the break. This chromosomal position is within the smallest region of overlap (SRO) found in neuroblastoma tumours and within the region of a constitutional interstitial deletion of a neuroblastoma patient. The YAC we describe here should serve as a DNA source for gene cloning approaches towards the isolation of candidates for the putative neuroblastoma suppressor gene.


Subject(s)
Chromosomes, Artificial, Yeast , Chromosomes, Human, Pair 15/genetics , Chromosomes, Human, Pair 1/genetics , Neuroblastoma/genetics , Translocation, Genetic/genetics , Humans , In Situ Hybridization, Fluorescence , Tumor Cells, Cultured
15.
Int J Oncol ; 4(5): 1043-5, 1994 May.
Article in English | MEDLINE | ID: mdl-21567017

ABSTRACT

Genetic instability resulting from deficiency in replication error repair (RER) has been detected in a variety of human cancers. We have examined the stability of five 1p microsatellites in 51 human neuroblastomas including stages 1, 2, 3, 4 and 4s. None of the tumors showed an aberrant pattern indicating RER at any microsatellite. Our results do not support a major role for increased genetic instability as the result of deficient replication error repair in neuroblastoma.

16.
Oncol Res ; 9(3): 119-27, 1997.
Article in English | MEDLINE | ID: mdl-9220497

ABSTRACT

The adenomatous polyposis coli (APC) tumor suppressor gene APC is mutated in familial adenomatous polyposis and in most sporadic colorectal tumors. Through its interaction with beta-catenin the APC protein may play a role in a signal transduction pathway regulating cell proliferation. Despite the fact that APC is ubiquitously expressed, mutations leading to truncated proteins are restricted to tumors of the digestive system. To determine further alterations not resulting in protein truncation, but possibly influencing the signaling, we compared the relative expression level of the APC protein and transcripts in 24 human colorectal cancer cell lines and in additional 17 lines of noncolorectal tissue origins, which have not previously been studied. By Western analysis, the highest levels of full-length APC protein were found in a subset of neuroblastoma and retinoblastoma cell lines. In contrast, in five noncolorectal lines it was not detectable. Truncated APC was exclusively found in 18 of the 24 colorectal cancer cell lines, but was never detected in any cell line derived from other tissues. In most colorectal cancer cell lines the protein level of full-length or mutated APC was reduced. By the more sensitive immunoprecipitation analysis, weak expression of full-length APC could be shown even in those noncolorectal cancer cell lines where it was not detectable by Western blotting. In addition, APC transcript expression was found in all cell lines, the level in colorectal cancer cell lines being reduced.


Subject(s)
Adenomatous Polyposis Coli/genetics , Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Adenomatous Polyposis Coli Protein , Blotting, Western , Humans
18.
Article in English | MEDLINE | ID: mdl-16869787

ABSTRACT

Erlotinib and gefitinib are small-molecule inhibitors of the epidermal growth factor tyrosine kinase. Erlotinib is approved for the treatment of locally advanced or metastatic non-small-cell lung cancer after failure of at least one prior chemotherapy regimen. Although it is active in unselected patients, clinical characteristics and tumor molecular markers associated with enhanced benefit have been identified. Notably, never-smoker status or a positive EGFR FISH test has been consistently predictive of greater erlotinib benefit. Other markers, such as EGFR mutations and EGFR protein expression, as determined by immunohistochemistry, and KRAS mutation status have not proven to be consistently associated with differential benefit.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , ErbB Receptors/antagonists & inhibitors , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Protein Kinase Inhibitors/therapeutic use , Carcinoma, Non-Small-Cell Lung/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Erlotinib Hydrochloride , Gefitinib , Gene Dosage , Genes, ras , Humans , Lung Neoplasms/genetics , Mutation , Prognosis , Quinazolines/therapeutic use , Signal Transduction , Smoking/adverse effects , Smoking/genetics
19.
Genes Chromosomes Cancer ; 1(3): 181-93, 1990 Jan.
Article in English | MEDLINE | ID: mdl-1982063

ABSTRACT

Increased dosage of cellular oncogenes resulting from amplification of DNA is a frequent genetic abnormality of tumor cells and the study of oncogene amplification has been paradigmatic for the usefulness of molecular genetic research in clinical oncology. Certain types of human tumors carry an amplified cellular oncogene at frequencies of up to 50-60%. Human neuroblastoma has been prototypic for the importance of oncogene amplification in tumorigenesis, and evidence is emerging that amplification may be an early event involved in a more malignant form of this cancer. It is unclear at which stage amplification plays a role in other cancers. Amplification of cellular oncogenes is a good predictor of clinical outcome in some human malignancies.


Subject(s)
Gene Amplification , Neoplasms/genetics , Oncogenes , Breast Neoplasms/genetics , Carcinoma, Small Cell/genetics , Cell Transformation, Neoplastic/genetics , Chromosome Aberrations , Colorectal Neoplasms/genetics , DNA, Neoplasm/genetics , Female , Genes, myc , Humans , Lung Neoplasms/genetics , Models, Genetic , Neuroblastoma/genetics , Ovarian Neoplasms/genetics , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogenes , Receptor, ErbB-2
SELECTION OF CITATIONS
SEARCH DETAIL