Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Eur J Cancer ; 114: 67-75, 2019 06.
Article in English | MEDLINE | ID: mdl-31075726

ABSTRACT

BACKGROUND: Immune checkpoint inhibitors (ICIs) are being increasingly utilised in the front-line (1L) setting of metastatic clear-cell renal cell carcinoma (mccRCC). Limited data exist on responses and survival on second-line (2L) vascular endothelial growth factor-receptor tyrosine kinase inhibitor (VEGFR-TKI) therapy after 1L ICI therapy. PATIENTS AND METHODS: This is a retrospective study of mccRCC patients treated with 2L VEGFR-TKI after progressive disease (PD) with 1L ICI. Patients were treated at MD Anderson Cancer Center or Memorial Sloan Kettering Cancer Center between December 2015 and February 2018. Objective response was assessed by blinded radiologists' review using Response Evaluation Criteria in Solid Tumours v1.1. Descriptive statistics and Kaplan-Meier method were used. RESULTS: Seventy patients were included in the analysis. Median age at mccRCC diagnosis was 59 years; 8 patients (11%) had international metastatic database consortium favourable-risk disease, 48 (69%) had intermediate-risk disease and 14 (20%) had poor-risk disease. As 1L therapy, 12 patients (17%) received anti-programmed death ligand-1 (PD-(L)1) monotherapy with nivolumab or atezolizumab, 33 (47%) received nivolumab plus ipilimumab and 25 (36%) received combination anti-PD-(L)1 plus bevacizumab. 2L TKI therapies included pazopanib, sunitinib, axitinib and cabozantinib. On 2L TKI therapy, one patient (1.5%) achieved a complete response, 27 patients (39.7%) a partial response and 36 patients (52.9%) stable disease. Median progression-free survival (mPFS) was 13.2 months (95% confidence interval: 10.1, NA). Forty-five percent of subjects required a dose reduction, and twenty-seven percent of patients discontinued treatment because of toxicity. CONCLUSIONS: In this retrospective study of patients with mccRCC receiving 2L TKI monotherapy after 1L ICI, we observed 2L antitumour activity and tolerance comparable to historical data for 1L TKI.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Immunotherapy/methods , Kidney Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Female , Humans , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Male , Middle Aged , Protein Kinase Inhibitors/pharmacology , Retrospective Studies , Survival Analysis , Treatment Outcome
2.
Ann Oncol ; 28(4): 804-808, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28049139

ABSTRACT

Background: Activation of the phosphoinisitide-3 kinase (PI3K) pathway through mutation and constitutive upregulation has been described in renal cell carcinoma (RCC), making it an attractive target for therapeutic intervention. We performed a randomized phase II study in vascular endothelial growth factor (VEGF) therapy refractory patients to determine whether MK-2206, an allosteric inhibitor of AKT, was more efficacious than the mammalian target of rapamycin inhibitor everolimus. Patients and methods: A total of 43 patients were randomized in a 2:1 distribution, with 29 patients assigned to the MK-2206 arm and 14 to the everolimus arm. Progression-free survival (PFS) was the primary endpoint. Results: The trial was closed at the first futility analysis with an observed PFS of 3.68 months in the MK-2206 arm and 5.98 months in the everolimus arm. Dichotomous response rate profiles were seen in the MK-2206 arm with one complete response and three partial responses in the MK-2206 arm versus none in the everolimus arm. On the other hand, progressive disease was best response in 44.8% of MK2206 versus 14.3% of everolimus-treated patients. MK-2206 induced significantly more rash and pruritis than everolimus, and dose reduction occurred in 37.9% of MK-2206 versus 21.4% of everolimus-treated patients. Genomic analysis revealed that 57.1% of the patients in the PD group had either deleterious TP53 mutations or ATM mutations or deletions. In contrast, none of the patients in the non-PD group had TP53 or ATM defects. No predictive marker for response was observed in this small dataset. Conclusions: Dichotomous outcomes are observed when VEGF therapy refractory patients are treated with MK-2206, and MK-2206 does not demonstrate superiority to everolimus. Additionally, mutations in DNA repair genes are associated with early disease progression, indicating that dysregulation of DNA repair is associated with a more aggressive tumor phenotype in RCC.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/drug therapy , Everolimus/therapeutic use , Heterocyclic Compounds, 3-Ring/therapeutic use , Kidney Neoplasms/drug therapy , Aged , Aged, 80 and over , Carcinoma, Renal Cell/mortality , Disease-Free Survival , Female , Humans , Kidney Neoplasms/mortality , Male , Middle Aged , Treatment Outcome
3.
Oncogene ; 34(22): 2815-22, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25132269

ABSTRACT

DNA damage response (DDR) includes the activation of numerous cellular activities that prevent duplication of DNA lesions and maintain genomic integrity, which is critical for the survival of normal and cancer cells. Specific genes involved in the DDR such as BRCA1/2 and P53 are mutated during prostate cancer progression, while various oncogenic signaling such as Akt and c-Myc are activated, enhancing the replication stress and increasing the genomic instability of cancer cells. These events may render prostate cancer cells particularly sensitive to inhibition of specific DDR pathways, such as PARP in homologous recombination DNA repair and Chk1 in cell cycle checkpoint and DNA repair, creating opportunities for synthetic lethality or synergistic cytotoxicity. Recent reports highlight the critical role of androgen receptor (AR) as a regulator of DDR genes, providing a rationale for combining DNA-damaging agents or targeted DDR inhibitors with hormonal manipulation or AR inhibition as treatment for aggressive disease. The aims of this review are to discuss specific DDR defects in prostate cancer that occur during disease progression, to summarize recent advances in understanding the regulation of DDR in prostate cancer, and to present potential therapeutic opportunities through combinational targeting of the intact components of DDR signaling pathways.


Subject(s)
DNA Damage/genetics , DNA Repair/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Animals , Cell Cycle Checkpoints/genetics , DNA Replication , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Genomic Instability/genetics , Humans , Male , Receptors, Androgen/physiology
4.
Oncogene ; 32(49): 5501-11, 2013 Dec 05.
Article in English | MEDLINE | ID: mdl-23752182

ABSTRACT

Prostate cancer is the second-leading cause of cancer-related mortality in men in Western societies. Androgen receptor (AR) signaling is a critical survival pathway for prostate cancer cells, and androgen-deprivation therapy (ADT) remains the principal treatment for patients with locally advanced and metastatic disease. Although a majority of patients initially respond to ADT, most will eventually develop castrate resistance, defined as disease progression despite serum testosterone levels of <20 ng/dl. The recent discovery that AR signaling persists during systemic castration via intratumoral production of androgens led to the development of novel anti-androgen therapies including abiraterone acetate and enzalutamide. Although these agents effectively palliate symptoms and prolong life, metastatic castration-resistant prostate cancer remains incurable. An increased understanding of the mechanisms that underlie the pathogenesis of castrate resistance is therefore needed to develop novel therapeutic approaches for this disease. The aim of this review is to summarize the current literature on the biology and treatment of castrate-resistant prostate cancer.


Subject(s)
Androgen Antagonists/therapeutic use , Prostatic Neoplasms, Castration-Resistant/drug therapy , Receptors, Androgen/metabolism , Castration , Disease Progression , Drug Resistance, Neoplasm , Humans , Male , Neoplasm Metastasis/drug therapy , Receptors, Androgen/genetics
5.
Br J Cancer ; 107(9): 1547-53, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23033003

ABSTRACT

BACKGROUND: The bone-forming metastases of prostate cancer result from complex stromal-epithelial interactions within the tumour microenvironment. Autocrine-paracrine signalling pathways between prostate cancer epithelial cells, osteoblasts, and osteoclasts stimulate aberrant bone remodelling, and the activity of these three cell populations can be quantitatively measured using prostate-specific antigen (PSA), bone-specific alkaline phosphatase (BAP) and urine N-telopeptide (uNTx), respectively. The purpose of the present study was to test the hypothesis that serial measurements of BAP and uNTx during therapy would facilitate monitoring of disease activity and predict the overall survival (OS) in patients with metastatic prostate cancer receiving therapy. METHODS: Radionuclide bone scan, PSA, BAP, and uNTx data were retrospectively analysed from three clinical trials in patients with metastatic prostate cancer conducted at our institution. Qualitative changes in bone scans and quantitative changes in PSA, BAP, and uNTx concentrations during therapy were correlated with OS. RESULTS: Baseline levels of BAP, but not PSA, were prognostic for OS in both androgen-dependent and castrate-resistant disease. A reduction in PSA, BAP, uNTx, or BAP/uNTx on therapy was predictive of improved OS in both patient groups. Conversely, an increase in PSA, or BAP on therapy was predictive of worse OS in both patient groups. Baseline number of lesions and response on bone scan during therapy were neither prognostic nor predictive of OS in either patient group. CONCLUSION: These observations support the concept that serial measurements of bone turnover metabolites during therapy function as clinically informative predictive biomarkers in patients with advanced prostate cancer and skeletal metastases. PSA measurements and bone scans remain essential to monitor the overall disease activity and determine the anatomic distribution of skeletal metastases.


Subject(s)
Biomarkers, Tumor/metabolism , Bone Neoplasms/metabolism , Bone and Bones/metabolism , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Alkaline Phosphatase/analysis , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Bone Remodeling , Bone and Bones/pathology , Collagen Type I/urine , Humans , Kallikreins/analysis , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis , Peptides/urine , Prognosis , Prostate-Specific Antigen/analysis , Prostatic Neoplasms/drug therapy , Randomized Controlled Trials as Topic , Retrospective Studies , Survival Analysis
6.
Ann Oncol ; 22(5): 1048-1053, 2011 May.
Article in English | MEDLINE | ID: mdl-21115604

ABSTRACT

BACKGROUND: Cytoreductive nephrectomy (CN) became a standard procedure in metastatic renal cell carcinoma (mRCC) in the immunotherapy era. Historically, median overall survival (OS) of patients treated with interferon alpha (IFN-α) without CN was 7.8 months. Median OS in patients treated with targeted therapy (TT) without CN is unknown. PATIENTS AND METHODS: We retrospectively reviewed records of patients with mRCC who received TT without CN. Kaplan-Meier methods and Cox regression analysis were used to estimate median OS and identify poor prognostic factors. RESULTS: One hundred and eighty-eight patients were identified. Most patients had intermediate-risk (54.8%) or poor-risk (44.1%) disease. Median OS for all patients was 10.4 months [95% confidence interval (CI) 8.1-12.5]. By multivariable analysis, elevated baseline lactate dehydrogenase and corrected calcium, performance status of two or more, retroperitoneal nodal metastasis, thrombocytosis, current smoking, two or more metastatic sites, and lymphopenia were independent risk factors for inferior OS. Patients with four or more factors had increased risk of death (hazard ratio 8.83, 95% CI 5.02-15.5, P < 0.001) and 5.5-month median OS. Nineteen patients (10.0%) survived for 2+ years. CONCLUSIONS: These data highlight the improved OS of patients with mRCC treated with TT without CN, compared with historical IFN-α treatment, and may guide the design of trials investigating the role of CN in the TT era.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Molecular Targeted Therapy , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/pathology , Female , Humans , Kaplan-Meier Estimate , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Male , Middle Aged , Multivariate Analysis , Neoplasm Metastasis , Nephrectomy , Proportional Hazards Models , Retrospective Studies , Treatment Outcome , Young Adult
7.
Clin Cancer Res ; 7(9): 2765-9, 2001 Sep.
Article in English | MEDLINE | ID: mdl-11555590

ABSTRACT

PURPOSE: E-cadherin, a M(r) 120,000 transmembrane glycoprotein, mediates calcium-dependent intercellular adhesion that is essential for normal tissue homeostasis. Loss of E-cadherin occurs in a variety of epithelial tumors and is correlated with invasion and metastasis. In esophageal adenocarcinoma, reduction of E-cadherin expression has been demonstrated previously, but mutations of the gene (CDH1) are rare. EXPERIMENTAL DESIGN: In this study, we used a nested PCR approach to examine the methylation status of the 5' CpG island of E-cadherin in esophageal specimens obtained from individuals with and without a history of esophageal cancer. RESULTS: In four individuals without esophageal cancer, E-cadherin was completely unmethylated in normal squamous cell-lined esophageal mucosa. In contrast, in patients with esophageal adenocarcinoma, E-cadherin was methylated in 26 of 31 (84%) tumor specimens. In the majority of cases, matched normal tissue (esophagus or stomach) from each patient was completely unmethylated. By immunostaining, methylated tumor samples demonstrated heterogeneously decreased membranous E-cadherin staining. CONCLUSIONS: These data suggest that epigenetic silencing via aberrant methylation of the E-cadherin promoter is a common cause of inactivation of this gene in esophageal adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Cadherins/genetics , CpG Islands/genetics , DNA Methylation , Esophageal Neoplasms/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Cadherins/analysis , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Female , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Staging , Tumor Cells, Cultured
8.
Cancer Res ; 61(14): 5636-43, 2001 Jul 15.
Article in English | MEDLINE | ID: mdl-11454718

ABSTRACT

Overexpression of E2F-1 induces apoptosis by both a p14ARF-p53- and a p73-mediated pathway. p14ARF is the alternate tumor suppressor product of the INK4a/ARF locus that is inactivated frequently in lung carcinogenesis. Because p14ARF stabilizes p53, it has been proposed that the loss of p14ARF is functionally equivalent to a p53 mutation. We have tested this hypothesis by examining the genomic status of the unique exon 1beta of p14ARF in 53 human cell lines and 86 primary non-small cell lung carcinomas and correlated this with previously characterized alterations of p53. Homozygous deletions of p14ARF were detected in 12 of 53 (23%) cell lines and 16 of 86 (19%) primary tumors. A single cell line, but no primary tumors, harbored an intragenic mutation. The deletion of p14ARF was inversely correlated with the loss of p53 in the majority of cell lines (P = 0.02), but this relationship was not maintained among primary tumors (P = 0.5). E2F-1 can also induce p73 via a p53-independent apoptotic pathway. Although we did not observe inactivation of p73 by either mutation or DNA methylation, haploinsufficiency of p73 correlated positively with either p14ARF or p53 mutation or both (P = 0.01) in primary non-small cell lung carcinomas. These data are consistent with the current model of p14ARF and p53 interaction as a complex network rather than a simple linear pathway and indicate a possible role for an E2F-1-mediated failsafe, p53-independent, apoptotic pathway involving p73 in human lung carcinogenesis.


Subject(s)
Apoptosis , Carcinoma, Non-Small-Cell Lung/genetics , Carrier Proteins , Cell Cycle Proteins , Lung Neoplasms/genetics , Proteins/genetics , Transcription Factors/physiology , Base Sequence , Carcinoma, Non-Small-Cell Lung/pathology , DNA Mutational Analysis , DNA, Neoplasm/chemistry , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , E2F Transcription Factors , E2F1 Transcription Factor , Female , Gene Deletion , Genes, Tumor Suppressor , Humans , Loss of Heterozygosity , Lung Neoplasms/pathology , Male , Mutation , Nuclear Proteins/genetics , Retinoblastoma-Binding Protein 1 , Signal Transduction , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Protein p14ARF , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins
9.
Cancer Res ; 61(8): 3225-9, 2001 Apr 15.
Article in English | MEDLINE | ID: mdl-11309270

ABSTRACT

We are in an era where the potential exists for deriving comprehensive profiles of DNA alterations characterizing each form of human cancer. Such profiles would provide invaluable insight into mechanisms underlying the evolution of each tumor type and will provide molecular markers, which could radically improve cancer detection. To date, no one type of DNA change has been defined which accomplishes this purpose. Herein, by using a candidate gene approach, we show that one category of DNA alteration, aberrant methylation of gene promoter regions, can enormously contribute to the above goals. We have now analyzed a series of promoter hypermethylation changes in 12 genes (p16(INK4a), p15(INK4b), p14(ARF), p73, APC,(5) BRCA1, hMLH1, GSTP1, MGMT, CDH1, TIMP3, and DAPK), each rigorously characterized for association with abnormal gene silencing in cancer, in DNA from over 600 primary tumor samples representing 15 major tumor types. The genes play known important roles in processes encompassing tumor suppression, cell cycle regulation, apoptosis, DNA repair, and metastastic potential. A unique profile of promoter hypermethylation exists for each human cancer in which some gene changes are shared and others are cancer-type specific. The hypermethylation of the genes occurs independently to the extent that a panel of three to four markers defines an abnormality in 70-90% of each cancer type. Our results provide an unusual view of the pervasiveness of DNA alterations, in this case an epigenetic change, in human cancer and a powerful set of markers to outline the disruption of critical pathways in tumorigenesis and for derivation of sensitive molecular detection strategies for virtually every human tumor type.


Subject(s)
DNA Methylation , Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Genes, Tumor Suppressor , Genetic Markers , Humans , Promoter Regions, Genetic
10.
Cancer Res ; 61(7): 2816-21, 2001 Apr 01.
Article in English | MEDLINE | ID: mdl-11306450

ABSTRACT

The INK4a/ARF locus encodes two distinct tumor suppressors, p16INK4a and p14ARF. Although the contribution of p16INK4a to human tumorigenesis through point mutation, deletion, and hypermethylation has been widely documented, little is known about specific p14ARF lesions and their consequences. Recent data indicate that p14ARF suffers inactivation by promoter hypermethylation in colorectal cancer cells. Because it is known that p14ARF prevents MDM2 nucleocytoplasmic shuttling and thus stabilizes p53 by attenuating MDM2-mediated degradation, we studied the relationship of p14ARF epigenetic silencing to the expression and localization of MDM2 and p53. Cancer cell lines with an unmethylated p14ARF promoter showed strong nuclear expression of MDM2, whereas in a colorectal cell line with p14ARF hypermethylation-associated inactivation, MDM2 protein was also seen in the cytosol. Treatment with the demethylating agent 5-aza-2'-deoxycytidine was able to reinternalize MDM2 to the nucleus, and p53 expression was restored. No apparent changes in retinoblastoma localization were observed. We also studied the profile of p14ARF promoter hypermethylation in an extensive collection of 559 human primary tumors of different cell types, observing that in colorectal, gastric, renal, esophageal, and endometrial neoplasms and gliomas, aberrant methylation of p14ARF was a relatively common epigenetic event. MDM2 expression patterns revealed that lack of p14ARF promoter hypermethylation was associated with tumors showing exclusive nuclear MDM2 staining, whereas MDM2 cytosolic staining was frequently observed in neoplasms with aberrant p14ARF methylation. Taken together, these data support that epigenetic silencing of p14ARF by promoter hypermethylation is a key mechanism in the disturbance of the MDM2 nuclear localization in human cancer.


Subject(s)
DNA Methylation , Gene Silencing , Nuclear Proteins , Promoter Regions, Genetic , Proteins/genetics , Proto-Oncogene Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Protein Biosynthesis , Proteins/physiology , Proto-Oncogene Proteins c-mdm2 , Tumor Cells, Cultured , Tumor Suppressor Protein p14ARF
11.
Clin Cancer Res ; 6(11): 4243-8, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11106238

ABSTRACT

E-Cadherin is a transmembrane glycoprotein that mediates Ca2+-dependent intercellular adhesion in normal epithelium. In tumors of epithelial origin, E-cadherin expression frequently is reduced, an event that contributes to tumor invasion and metastasis. The role of E-cadherin in hematopoietic tissues is less clear. In normal bone marrow, E-cadherin is expressed on erythroid progenitors, CD34+ stem cells, and stromal cells, where it likely contributes to intercellular interactions during hematopoiesis. In this study, we used a nested-PCR approach to examine the methylation status of the E-cadherin 5' CpG island in blood and bone marrow samples from normal donors and in bone marrow from patients with acute leukemia. In normal peripheral blood mononuclear cells and bone marrow, E-cadherin was completely unmethylated. In peripheral blood mononuclear cells, expression was evident by reverse transcription-PCR. Immunoblotting confirmed E-cadherin protein expression in two lymphoblastoid cell lines derived from normal donors. In contrast, E-cadherin was aberrantly methylated in 4 of 4 (100%) leukemia cell lines, 14 of 44 (32%) acute myelogenous leukemias, and 18 of 33 (53%) acute lymphoblastic leukemias. Genomic bisulfite sequencing of primary leukemias confirmed dense methylation across the CpG island. Methylation was associated with loss of E-cadherin RNA and protein in leukemia cell lines and primary leukemias. Following treatment with 5-aza-2'-deoxycytidine, a methylated leukemia cell line expressed both E-cadherin transcript and protein. Our results show that methylation of E-cadherin occurs commonly in acute leukemia and suggests a hypothesis for E-cadherin down-regulation in leukemogenesis.


Subject(s)
Cadherins/genetics , CpG Islands , DNA Methylation , Gene Silencing , Leukemia, Myeloid, Acute/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Humans , Polymerase Chain Reaction
12.
Oncogene ; 19(1): 164-8, 2000 Jan 06.
Article in English | MEDLINE | ID: mdl-10644993

ABSTRACT

Germ-line mutations of the LKB1 gene cause Peutz-Jeghers syndrome (PJS) characterized by mucocutaneous pigmentation, predisposition to benign hamartomas of the gastrointestinal tract and also to several types of tumors. However, somatic mutations of this gene are very rare. To examine inactivation of LKB1 by epigenetic mechanisms, we investigated a series of primary tumors and cancer cell lines, for hypermethylation affecting the CpG island located in the 5' region of the LKB1 gene using Methylation-specific PCR (MSP). First, we screened 51 cancer cell lines. Only three colorectal and one cervical carcinoma cell lines were methylated at LKB1, and loss of the LKB1 transcript was demonstrated. Treatment with the demethylating agent 5-aza-2'-deoxycytidine restored LKB1 expression. To address the incidence of LKB1 epigenetic inactivation in primary tumors, we analysed colorectal, breast, gastric, pancreatic, thyroid, bladder and testicular carcinomas (n=195). Normal tissues from the mentioned organs were unmethylated in this region. Among the described tumors, only one colorectal carcinoma and three testicular tumors displayed LKB1 promoter hypermethylation. Further study of those histological types more commonly associated with PJS, demonstrated that LKB1 promoter hypermethylation was present in five of 11 (45%) papillary breast carcinomas. Finally, in three patients with a strong family story suggestive of PJS disease, abnormal LKB1 methylation was found in four of 22 (18%) hamartomatous polyps lesions. Our findings provide an alternative pathway for inactivation of the LKB1 tumor suppressor gene involving promoter hypermethylation.


Subject(s)
DNA Methylation , Neoplasms/genetics , Peutz-Jeghers Syndrome/genetics , Protein Serine-Threonine Kinases/genetics , AMP-Activated Protein Kinase Kinases , Genes, Tumor Suppressor , Humans , Promoter Regions, Genetic , Tumor Cells, Cultured
13.
Cancer Res ; 59(14): 3352-6, 1999 Jul 15.
Article in English | MEDLINE | ID: mdl-10416592

ABSTRACT

The p73 gene is located on 1p36.2-3, a region that is frequently deleted in human cancer. Because p73 encodes for a protein that is both structurally and functionally homologous to the p53 protein, p73 has been postulated to be a candidate tumor suppressor gene. To date, however, mutations of p73 have not been found. To study methylation of the p73 5'CpG island, a human bacterial artificial chromosome clone containing exon 1 and the 5' region of p73 was isolated. There was no evidence for p73 exon 1 methylation in normal tissues. In contrast, p73 was aberrantly methylated in approximately 30% of primary acute lymphoblastic leukemias (ALLs) and Burkitt's lymphomas. There was no evidence for methylation in any other types of hematological malignancies or solid tumors examined. In both leukemia cell lines and primary ALLs, methylation was associated with transcriptional loss of p73 by reverse transcription-PCR. We used single-strand conformational polymorphisms to screen for point mutations in a series of primary ALLs and found no mutations leading to a change in protein structure. Our results show that methylation of p73 is a frequent event in specific types of hematological malignancies and suggest that epigenetic silencing of p73 could have important consequences for cell-cycle regulation.


Subject(s)
Burkitt Lymphoma/genetics , Chromosomes, Human, Pair 1/genetics , CpG Islands , DNA Methylation , DNA, Neoplasm/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Nuclear Proteins/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Transcription, Genetic , Adult , Burkitt Lymphoma/pathology , Child , DNA Mutational Analysis , DNA, Neoplasm/chemistry , DNA-Binding Proteins/physiology , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Neoplasms/genetics , Neoplasms/pathology , Nuclear Proteins/physiology , Polymorphism, Single-Stranded Conformational , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Promoter Regions, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Protein p73 , Tumor Suppressor Proteins
14.
Cancer Res ; 59(4): 798-802, 1999 Feb 15.
Article in English | MEDLINE | ID: mdl-10029065

ABSTRACT

Tissue inhibitor of metalloproteinase-3 (TIMP-3) antagonizes matrix metalloproteinase activity and can suppress tumor growth, angiogenesis, invasion, and metastasis. Loss of TIMP-3 has been related to the acquisition of tumorigenesis. Herein, we show that TIMP-3 is silenced in association with aberrant promoter-region methylation in cell lines derived from human cancers. TIMP-3 expression was restored after 5-aza-2'deoxycytidine-mediated demethylation of the TIMP-3 proximal promoter region. Genomic bisulfite sequencing revealed that TIMP-3 silencing was related to the overall density of methylation and that discrete regions within the TIMP-3 CpG island may be important for the silencing of this gene. Aberrant methylation of TIMP-3 occurred in primary cancers of the kidney, brain, colon, breast, and lung, but not in any of 41 normal tissue samples. The most frequent TIMP-3 methylation was found in renal cancers, which originate in the tissue that normally expresses the highest TIMP-3 levels. This methylation correlated with a lack of detectable TIMP-3 protein in these tumors. Together, these data show that methylation-associated inactivation of TIMP-3 is frequent in many human tumors.


Subject(s)
Brain Neoplasms/genetics , DNA Methylation , Kidney Neoplasms/genetics , Tissue Inhibitor of Metalloproteinase-3/genetics , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , CpG Islands , Decitabine , Humans
15.
Cancer Res ; 58(20): 4515-8, 1998 Oct 15.
Article in English | MEDLINE | ID: mdl-9788592

ABSTRACT

Glutathione S-transferases (GSTs) are a family of isoenzymes that play an important role in protecting cells from cytotoxic and carcinogenic agents. The pi-class GST has been associated with preneoplastic and neoplastic changes. Recently, it has been reported that regulatory sequences near the GSTP1 gene, which encodes the human pi-class GST, are commonly hypermethylated in prostatic carcinomas. In the present study, we studied more than 300 primary human tumors originating in other organs for aberrant methylation of GSTP1 using methylation-specific PCR. GSTP1 hypermethylation was most frequent in breast and renal carcinoma, showing aberrant methylation in 30 and 20% of the cases, respectively. Other tumor types showed promoter methylation only rarely or not at all. Hypermethylation of GSTP1 was associated with loss of expression demonstrated by immunohistochemistry. Our results suggest that aberrant methylation of GSTP1 may contribute to the carcinogenetic process in breast and renal carcinomas.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Glutathione Transferase/genetics , Isoenzymes/genetics , Kidney Neoplasms/genetics , Promoter Regions, Genetic , Drug Resistance, Neoplasm , Glutathione S-Transferase pi , Glutathione Transferase/metabolism , Humans , Immunohistochemistry , Isoenzymes/metabolism
16.
J Infect Dis ; 167(2): 356-64, 1993 Feb.
Article in English | MEDLINE | ID: mdl-8421169

ABSTRACT

The genes involved in Haemophilus influenzae type b capsule expression are present as a duplication of an approximately 18-kb DNA segment (the Cap b locus). It has been shown previously that recombination occurs between the two copies of the repeat, resulting in deletion of one copy and loss of capsule expression at frequencies of 0.1%-0.5%. The present study tested the hypothesis that the duplicated arrangement could serve as a template for further amplification of capsule gene sequences. Southern hybridization analysis of 66 type b invasive isolates showed that amplifications exist and are moderately common (23/66 were amplified). In addition to three copies of the 18-kb repeat, four copies were detected in some strains, and up to five copies in 1 isolate. By ELISA, a five-copy strain made about six times more capsular polysaccharide than did an isogenic two-copy derivative. The evolutionary significance of the duplicated arrangement may be its ability to rapidly amplify under conditions where it is advantageous to produce more capsule.


Subject(s)
Bacterial Capsules/genetics , Gene Amplification , Gene Expression Regulation, Bacterial , Haemophilus Infections/microbiology , Haemophilus Vaccines , Haemophilus influenzae/genetics , Bacterial Vaccines/analysis , Bacterial Vaccines/biosynthesis , Blotting, Southern , DNA, Bacterial/analysis , District of Columbia , Enzyme-Linked Immunosorbent Assay , Finland , Haemophilus influenzae/pathogenicity , Humans , Nucleic Acid Hybridization , Polysaccharides, Bacterial/analysis , Polysaccharides, Bacterial/biosynthesis , Restriction Mapping , Virulence
18.
Pediatr Res ; 26(1): 54-7, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2771508

ABSTRACT

Recent studies have shown that immature rats display a diminished sensitivity to the phosphaturic effects of parathyroid hormone (PTH), and that the responsiveness to PTH increases with age. The attenuated phosphaturia may reflect an inability of the neonate to respond to the hormone because of functional immaturity of the developing kidney. Alternatively, PTH may actually inhibit tubular phosphate reabsorption in the neonate but, due to other phosphate conservation mechanisms, no phosphaturia occurs. Our objective was to determine whether a phosphaturic response to PTH would be elicited in immature rats during infusion of moderate amounts of phosphate (Pi). Clearance experiments were performed on 26 acutely thyroparathyroidectomized immature Wistar rats (3-5 wk of age) fed a normal Pi diet (0.63%). In response to infusion of either Pi (1 mumol/min.100 g) (group I) or PTH (8.3 ng/min.100 g) (group II) alone, the fractional excretion of phosphate rose minimally (from 0.01 +/- 0.01% to 4.9 +/- 1.9% and from 0.12 +/- 0.12% to 2.9 +/- 1.4% for groups I and II, respectively). However, when Pi and PTH were combined either Pi first followed by PTH (group III) or PTH first followed by Pi (group IV), the fractional excretion of Pi rose dramatically (from 0.01 +/- 0.01 to 21.8 +/- 3.5% and from 0.04 +/- 0.04 to 27.7 +/- 3.3% for groups III and IV, respectively). A significant increase in urinary cAMP excretion occurred during infusion of PTH even when Pi excretion was minimal, but there was no further increase in urinary cAMP during the combined infusion of Pi and PTH.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Animals, Newborn/urine , Kidney/drug effects , Parathyroid Hormone/pharmacology , Phosphates/urine , Aging/physiology , Aging/urine , Animals , Male , Rats , Rats, Inbred Strains
SELECTION OF CITATIONS
SEARCH DETAIL
...