Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Breast Cancer Res ; 26(1): 69, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38650031

ABSTRACT

BACKGROUND: We previously reported our phase Ib trial, testing the safety, tolerability, and efficacy of T-DM1 + neratinib in HER2-positive metastatic breast cancer patients. Patients with ERBB2 amplification in ctDNA had deeper and more durable responses. This study extends these observations with in-depth analysis of molecular markers and mechanisms of resistance in additional patients. METHODS: Forty-nine HER2-positive patients (determined locally) who progressed on-treatment with trastuzumab + pertuzumab were enrolled in this phase Ib/II study. Mutations and HER2 amplifications were assessed in ctDNA before (C1D1) and on-treatment (C2D1) with the Guardant360 assay. Archived tissue (TP0) and study entry biopsies (TP1) were assayed for whole transcriptome, HER2 copy number, and mutations, with Ampli-Seq, and centrally for HER2 with CLIA assays. Patient responses were assessed with RECIST v1.1, and Molecular Response with the Guardant360 Response algorithm. RESULTS: The ORR in phase II was 7/22 (32%), which included all patients who had at least one dose of study therapy. In phase I, the ORR was 12/19 (63%), which included only patients who were considered evaluable, having received their first scan at 6 weeks. Central confirmation of HER2-positivity was found in 83% (30/36) of the TP0 samples. HER2-amplified ctDNA was found at C1D1 in 48% (20/42) of samples. Patients with ctHER2-amp versus non-amplified HER2 ctDNA determined in C1D1 ctDNA had a longer median progression-free survival (PFS): 480 days versus 60 days (P = 0.015). Molecular Response scores were significantly associated with both PFS (HR 0.28, 0.09-0.90, P = 0.033) and best response (P = 0.037). All five of the patients with ctHER2-amp at C1D1 who had undetectable ctDNA after study therapy had an objective response. Patients whose ctHER2-amp decreased on-treatment had better outcomes than patients whose ctHER2-amp remained unchanged. HER2 RNA levels show a correlation to HER2 CLIA IHC status and were significantly higher in patients with clinically documented responses compared to patients with progressive disease (P = 0.03). CONCLUSIONS: The following biomarkers were associated with better outcomes for patients treated with T-DM1 + neratinib: (1) ctHER2-amp (C1D1) or in TP1; (2) Molecular Response scores; (3) loss of detectable ctDNA; (4) RNA levels of HER2; and (5) on-treatment loss of detectable ctHER2-amp. HER2 transcriptional and IHC/FISH status identify HER2-low cases (IHC 1+ or IHC 2+ and FISH negative) in these heavily anti-HER2 treated patients. Due to the small number of patients and samples in this study, the associations we have shown are for hypothesis generation only and remain to be validated in future studies. Clinical Trials registration NCT02236000.


Subject(s)
Ado-Trastuzumab Emtansine , Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms , Quinolines , Receptor, ErbB-2 , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/mortality , Breast Neoplasms/metabolism , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Ado-Trastuzumab Emtansine/therapeutic use , Middle Aged , Quinolines/therapeutic use , Quinolines/administration & dosage , Aged , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Circulating Tumor DNA/genetics , Circulating Tumor DNA/blood , Biomarkers, Tumor/genetics , Mutation , Aged, 80 and over , Trastuzumab/therapeutic use , Trastuzumab/administration & dosage , Treatment Outcome , Neoplasm Metastasis
4.
Breast Cancer Res ; 21(1): 133, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31796073

ABSTRACT

PURPOSE: The primary aim of NSABP FB-7 was to determine the pathologic complete response (pCR) rate in locally advanced HER2-positive (HER2+) breast cancer patients treated with neoadjuvant trastuzumab or neratinib or the combination and weekly paclitaxel followed by standard doxorubicin plus cyclophosphamide. The secondary aims include biomarker analyses. EXPERIMENTAL DESIGN: pCR was tested for association with treatment, gene expression, and a single nucleotide polymorphism (SNP) in the Fc fragment of the IgG receptor IIIa-158V/F (FCGR3A). Pre-treatment biopsies and residual tumors were also compared to identify molecular changes. RESULTS: The numerical pCR rate in the trastuzumab plus neratinib arm (50% [95%CI 34-66%]) was greater than that for single-targeted therapies with trastuzumab (38% [95%CI 24-54]) or neratinib (33% [95%CI 20-50]) in the overall cohort but was not statistically significant. Hormone receptor-negative (HR-) tumors had a higher pCR rate than HR+ tumors in all three treatment arms, with the highest pCR rate in the combination arm. Diarrhea was the most frequent adverse event and occurred in virtually all patients who received neratinib-based therapy. Grade 3 diarrhea was reported in 31% of patients; there were no grade 4 events. Our 8-gene signature, previously validated for trastuzumab benefit in two different clinical trials in the adjuvant setting, was correlated with pCR across all arms of NSABP FB-7. Specifically, patients predicted to receive no trastuzumab benefit had a significantly lower pCR rate than did patients predicted to receive the most benefit (P = 0.03). FCGR genotyping showed that patients who were homozygous for the Fc low-binding phenylalanine (F) allele for FCGR3A-158V/F were less likely to achieve pCR. CONCLUSIONS: Combining trastuzumab plus neratinib with paclitaxel increased the absolute pCR rate in the overall cohort and in HR- patients. The 8-gene signature, which is validated for predicting trastuzumab benefit in the adjuvant setting, was associated with pCR in the neoadjuvant setting, but remains to be validated as a predictive marker in a larger neoadjuvant clinical trial. HR status, and the FCGR3A-158V/F genotype, also warrant further investigation to identify HER2+ patients who may benefit from additional anti-HER2 therapies beyond trastuzumab. All of these markers will require further validation in the neoadjuvant setting. TRIALS REGISTRATION: ClinicalTrials.gov, NCT01008150. Retrospectively registered on October 5, 2010.

5.
J Infect Dis ; 211(10): 1560-5, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25231015

ABSTRACT

Human polyomavirus 7 (HPyV7) is one of 11 HPyVs recently discovered through genomic sequencing technologies. Two lung transplant recipients receiving immunosuppressive therapy developed pruritic, brown plaques on the trunk and extremities showing a distinctive epidermal hyperplasia with virus-laden keratinocytes containing densely packed 36-45-nm icosahedral capsids. Rolling circle amplification and gradient centrifugation testing were positive for encapsidated HPyV7 DNA in skin and peripheral blood specimens from both patients, and HPyV7 early and capsid proteins were abundantly expressed in affected tissues. We describe for the first time that HPyV7 is associated with novel pathogenicity in some immunosuppressed individuals.


Subject(s)
Polyomavirus Infections/pathology , Polyomavirus Infections/virology , Polyomavirus/isolation & purification , Transplant Recipients , Tumor Virus Infections/pathology , Tumor Virus Infections/virology , Aged , Blood/virology , Exanthema/pathology , Exanthema/virology , Histocytochemistry , Humans , Immunocompromised Host , Immunohistochemistry , Male , Microscopy, Electron, Transmission , Skin/pathology , Skin/virology , Viremia
6.
Front Oncol ; 13: 1139347, 2023.
Article in English | MEDLINE | ID: mdl-37305569

ABSTRACT

Background: The cardiotoxic effects of doxorubicin, trastuzumab, and other anticancer agents are well known, but molecular genetic testing is lacking for the early identification of patients at risk for therapy-related cardiac toxicity. Methods: Using the Agena Bioscience MassARRAY system, we genotyped TRPC6 rs77679196, BRINP1 rs62568637, LDB2 rs55756123, RAB22A rs707557, intergenic rs4305714, LINC01060 rs7698718, and CBR3 rs1056892 (V244M) (previously associated with either doxorubicin or trastuzumab-related cardiotoxicity in the NCCTG N9831 trial of anthracycline-based chemotherapy ± trastuzumab) in 993 patients with HER2+ early breast cancer from the NSABP B-31 trial of adjuvant anthracycline-based chemotherapy ± trastuzumab. Association analyses were performed with outcomes of congestive heart failure (N = 29) and maximum decline in left ventricular ejection fraction (LVEF) using logistic and linear regression models, respectively, under an additive model with age, baseline LVEF, and previous use of hypertensive medications as covariates. Results: Associations of maximum decline in LVEF in the NCCTG N9831 patients did not replicate in the NSABP B-31 patients. However, TRPC6 rs77679196 and CBR3 rs1056892 were significantly associated with congestive heart failure, p < 0.05, with stronger associations observed in patients treated with chemotherapy only (no trastuzumab) or in the combined analysis of all patients relative to those patients treated with chemotherapy + trastuzumab. Conclusions: TRPC6 rs77679196 and CBR3 rs1056892 (V244M) are associated with doxorubicin-induced cardiac events in both NCCTG N9831 and NSABP B-31. Other variants previously associated with trastuzumab-related decline in LVEF failed to replicate between these studies.

7.
BMC Genomics ; 12: 625, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22185355

ABSTRACT

BACKGROUND: Kaposi's sarcoma-associated herpesvirus (KSHV) and Epstein-Barr virus (EBV) are related human tumor viruses that cause primary effusion lymphomas (PEL) and Burkitt's lymphomas (BL), respectively. Viral genes expressed in naturally-infected cancer cells contribute to disease pathogenesis; knowing which viral genes are expressed is critical in understanding how these viruses cause cancer. To evaluate the expression of viral genes, we used high-resolution separation and mass spectrometry coupled with custom tiling arrays to align the viral proteomes and transcriptomes of three PEL and two BL cell lines under latent and lytic culture conditions. RESULTS: The majority of viral genes were efficiently detected at the transcript and/or protein level on manipulating the viral life cycle. Overall the correlation of expressed viral proteins and transcripts was highly complementary in both validating and providing orthogonal data with latent/lytic viral gene expression. Our approach also identified novel viral genes in both KSHV and EBV, and extends viral genome annotation. Several previously uncharacterized genes were validated at both transcript and protein levels. CONCLUSIONS: This systems biology approach coupling proteome and transcriptome measurements provides a comprehensive view of viral gene expression that could not have been attained using each methodology independently. Detection of viral proteins in combination with viral transcripts is a potentially powerful method for establishing virus-disease relationships.


Subject(s)
Herpesvirus 4, Human/metabolism , Herpesvirus 8, Human/metabolism , Proteome , Transcriptome , Genes, Viral , Herpesvirus 4, Human/genetics , Herpesvirus 8, Human/genetics , Humans , Mass Spectrometry
8.
J Virol ; 84(14): 7064-72, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20444890

ABSTRACT

Merkel cell carcinoma (MCC) is the most aggressive skin cancer. Recently, it was demonstrated that human Merkel cell polyomavirus (MCV) is clonally integrated in approximately 80% of MCC tumors. However, direct evidence for whether oncogenic viral proteins are needed for the maintenance of MCC cells is still missing. To address this question, we knocked down MCV T-antigen (TA) expression in MCV-positive MCC cell lines using three different short hairpin RNA (shRNA)-expressing vectors targeting exon 1 of the TAs. The MCC cell lines used include three newly generated MCV-infected cell lines and one MCV-negative cell line from MCC tumors. Notably, all MCV-positive MCC cell lines underwent growth arrest and/or cell death upon TA knockdown, whereas the proliferation of MCV-negative cell lines remained unaffected. Despite an increase in the number of annexin V-positive, 7-amino-actinomycin D (7-AAD)-negative cells upon TA knockdown, activation of caspases or changes in the expression and phosphorylation of Bcl-2 family members were not consistently detected after TA suppression. Our study provides the first direct experimental evidence that TA expression is necessary for the maintenance of MCV-positive MCC and that MCV is the infectious cause of MCV-positive MCC.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Carcinoma, Merkel Cell/virology , Polyomavirus Infections/virology , Polyomavirus/immunology , Skin Neoplasms/virology , Adult , Aged , Aged, 80 and over , Antigens, Polyomavirus Transforming/genetics , Carcinoma, Merkel Cell/pathology , Cell Cycle/physiology , Cell Line, Tumor , Cell Survival , Female , Gene Knockdown Techniques , Humans , Male , Middle Aged , Polyomavirus/metabolism , Polyomavirus Infections/metabolism , Polyomavirus Infections/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology
9.
Proc Natl Acad Sci U S A ; 105(42): 16272-7, 2008 Oct 21.
Article in English | MEDLINE | ID: mdl-18812503

ABSTRACT

Merkel cell polyomavirus (MCV) is a virus discovered in our laboratory at the University of Pittsburgh that is monoclonally integrated into the genome of approximately 80% of human Merkel cell carcinomas (MCCs). Transcript mapping was performed to show that MCV expresses transcripts in MCCs similar to large T (LT), small T (ST), and 17kT transcripts of SV40. Nine MCC tumor-derived LT genomic sequences have been examined, and all were found to harbor mutations prematurely truncating the MCV LT helicase. In contrast, four presumed episomal viruses from nontumor sources did not possess this T antigen signature mutation. Using coimmunoprecipitation and origin replication assays, we show that tumor-derived virus mutations do not affect retinoblastoma tumor suppressor protein (Rb) binding by LT but do eliminate viral DNA replication capacity. Identification of an MCC cell line (MKL-1) having monoclonal MCV integration and the signature LT mutation allowed us to functionally test both tumor-derived and wild type (WT) T antigens. Only WT LT expression activates replication of integrated MCV DNA in MKL-1 cells. Our findings suggest that MCV-positive MCC tumor cells undergo selection for LT mutations to prevent autoactivation of integrated virus replication that would be detrimental to cell survival. Because these mutations render the virus replication-incompetent, MCV is not a "passenger virus" that secondarily infects MCC tumors.


Subject(s)
Antigens, Viral, Tumor/immunology , Carcinoma, Merkel Cell/metabolism , Carcinoma, Merkel Cell/virology , Polyomavirus/physiology , Amino Acid Sequence , Antigens, Viral, Tumor/chemistry , Antigens, Viral, Tumor/genetics , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/immunology , Cell Line, Tumor , Conserved Sequence , Humans , Molecular Sequence Data , Mutation/genetics , Sequence Alignment , Transcription, Genetic/genetics , Virus Replication
10.
Clin Cancer Res ; 27(6): 1612-1622, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33203645

ABSTRACT

PURPOSE: In metastatic colorectal cancer (mCRC), HER2 (ERBB2) gene amplification is implicated in anti-EGFR therapy resistance. We sought to determine the recommended phase II dose (RP2D) and efficacy of neratinib, a pan-ERBB kinase inhibitor, combined with cetuximab, in patients with progressive disease (PD) on anti-EGFR treatment. PATIENTS AND METHODS: Twenty-one patients with quadruple-wild-type, refractory mCRC enrolled in this 3+3 phase Ib study. Standard dosage cetuximab was administered with neratinib at 120 mg, 160 mg, 200 mg, and 240 mg/day orally in 28-day cycles. Samples were collected for molecular and pharmacokinetic studies. RESULTS: Sixteen patients were evaluable for dose-limiting toxicity (DLT). 240 mg was determined to be the RP2D wherein a single DLT occurred (1/7 patients). Treatment-related DLTs were not seen at lower doses. Best response was stable disease (SD) in 7 of 16 (44%) patients. HER2 amplification (chromogenic in situ IHC) was detected in 2 of 21 (9.5%) treatment-naïve tumors and 4 of 16 (25%) biopsies upon trial enrollment (post-anti-EGFR treatment and progression). Compared with matched enrollment biopsies, 6 of 8 (75%) blood samples showed concordance for HER2 CNV in circulating cell-free DNA. Five SD patients had HER2 amplification in either treatment-naïve or enrollment biopsies. Examination of gene-expression, total protein, and protein phosphorylation levels showed relative upregulation of ≥2 members of the HER-family receptors or ligands upon enrollment versus matched treatment-naïve samples. CONCLUSIONS: The RP2D of neratinib in this combination was 240 mg/day, which was well tolerated with low incidence of G3 AEs. There were no objective responses; SD was seen at all neratinib doses. HER2 amplification, detectable in both tissue and blood, was more frequent post-anti-EGFR therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Mutation , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cetuximab/administration & dosage , Class I Phosphatidylinositol 3-Kinases/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Female , Follow-Up Studies , GTP Phosphohydrolases/genetics , Humans , Male , Maximum Tolerated Dose , Membrane Proteins/genetics , Middle Aged , Neoplasm Metastasis , Panitumumab/administration & dosage , Prognosis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Quinolines/administration & dosage , Retrospective Studies , Tissue Distribution
11.
Int J Cancer ; 125(6): 1250-6, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19499548

ABSTRACT

Merkel cell polyomavirus (MCV) is a newly-discovered human tumor virus found in approximately 80% of Merkel cell carcinoma (MCC). The rate of MCV infection among persons without MCC is unknown. We developed a MCV virus-like particle (VLP) enzyme-linked immunoassay (EIA) that does not cross-react with human BK or murine polyomaviruses. Peptide mapping of the MCV VP1 gene and immunoblotting with denatured MCV VLP are less sensitive than the MCV EIA in detecting MCV antibodies suggesting antibody reactivity in this assay primarily targets conformational but not linear epitopes. Among MCC patients, all 21 (100%) patients tested with MCV-positive tumors had high serum MCV IgG but not high MCV IgM levels. Only 3 of 6 (50%) MCC patients with MCV-negative tumors were positive for MCV antibodies. Sera from most adults, including 107 of 166 (64%) blood donors, 63 of 100 (63%) commercial donors and 37 of 50 (74%) systemic lupus erythematosus patients, show evidence for prior MCV exposure. Age-specific MCV prevalence was determined by examining a cross-sectional distribution of 150 Langerhans cell histiocytosis (an unrelated neoplasm) patient sera. MCV prevalence increases from 50% among children age 15 years or younger to 80% among persons older than 50 years. We did not find evidence for vertical transmission among infants. Although past exposure to MCV is common among all adult groups, MCC patients have a markedly elevated MCV IgG response compared with control patients. Our study demonstrates that MCV is a widespread but previously unrecognized human infection.


Subject(s)
Capsid Proteins/immunology , Carcinoma, Merkel Cell/virology , Epitopes/immunology , Polyomavirus Infections/immunology , Polyomavirus/immunology , Skin Neoplasms/virology , Tumor Virus Infections/immunology , Adolescent , Adult , Aged , Aged, 80 and over , Antibodies, Viral/blood , BK Virus/physiology , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin G/blood , Middle Aged , Molecular Conformation
12.
Int J Cancer ; 125(6): 1243-9, 2009 Sep 15.
Article in English | MEDLINE | ID: mdl-19499546

ABSTRACT

Merkel cell polyomavirus (MCV) is a recently discovered human virus closely related to African green monkey lymphotropic polyomavirus. MCV DNA is integrated in approximately 80% of Merkel cell carcinomas (MCC), a neuroendocrine skin cancer linked to lymphoid malignancies such as chronic lymphocytic leukemia (CLL). To assess MCV infection and its association with human diseases, we developed a monoclonal antibody that specifically recognizes endogenous and transfected MCV large T (LT) antigen. We show expression of MCV LT protein localized to nuclei of tumor cells from MCC having PCR quantified MCV genome at an average of 5.2 (range 0.8-14.3) T antigen DNA copies per cell. Expression of this putative viral oncoprotein in tumor cells provides the mechanistic underpinning supporting the notion that MCV causes a subset of MCC. In contrast, although 2.2% of 325 hematolymphoid malignancies surveyed also showed evidence for MCV infection by DNA PCR, none were positive at high viral copy numbers, and none of 173 lymphoid malignancies examined on tissue microarrays expressed MCV LT protein in tumor cells. As with some of the other human polyomaviruses, lymphocytes may serve as a tissue reservoir for MCV infection, but hematolymphoid malignancies associated with MCC are unlikely to be caused by MCV.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Carcinoma, Merkel Cell/virology , Gene Expression Regulation, Viral , Lymphoid Tissue/virology , Lymphoma/virology , Polyomavirus Infections/genetics , Polyomavirus/pathogenicity , Skin Neoplasms/virology , Amino Acid Sequence , Carcinoma, Merkel Cell/pathology , DNA, Viral/analysis , Fluorescent Antibody Technique , Gene Dosage , Humans , Immunoblotting , Immunoenzyme Techniques , Molecular Sequence Data , Polymerase Chain Reaction , Polyomavirus Infections/pathology , Sequence Homology, Amino Acid , Tumor Virus Infections/genetics , Tumor Virus Infections/pathology
13.
J Gastroenterol Hepatol ; 23(10): 1613-20, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18717758

ABSTRACT

BACKGROUND AND AIM: Cyclooxygenase-2 (COX-2) plays an important role in the carcinogenesis of esophageal squamous cell carcinoma (ESCC). However, it is not clear whether COX-2 is involved in the early or late stage of the development of ESCC. The aim of this study was to investigate the role of COX-2 in the carcinogenesis of ESCC by an immortalized esophageal epithelial cell line. METHODS: Human papillomavirus type 16 (HPV16)-E6/E7 and human telomerase reverse transcriptase (hTERT) transfection were used for immortalization of esophageal epithelial cells. COX-2-specific RNA interference was used for the inhibition of COX-2 expression. RESULTS: An immortalized esophageal epithelial cell line, NE6-E6E7/hTERT, was established, which had high proliferation activity but failed to induce colony formation in soft agar. COX-2 expression was upregulated in the early process of immortalization, while COX-2 small interfering RNA (siRNA) decreased the Bcl-2 expression, increased the expression of Bax, and induced cell-cycle arrest at the G0/G1 phase in NE6-E6E7/hTERT cells. Expressions of p53, cyclinD1, and the ratio of hyperphosphorylated-RB/hypophosphorylated-RB were progressively increased after E6E7 and the subsequent hTERT transfections. These changes were accompanied by the alteration of COX-2 expression, but could be reversed by COX-2 siRNA (P < 0.05). P16 expression was significantly downregulated in NE6-E6E7 or NE6-E6E7/hTERT cells (P < 0.05), and was not affected by COX-2 siRNA. CONCLUSIONS: Our results suggest that induction of cyclooxygenase-2 is essential in the human papillomavirus type 16 and hTERT-induced immortalization of human esophageal epithelial cells, and that COX-2 inhibition may be a potential target to block the carcinogenesis of ESCC at the precancerous stage.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Viral , Cyclooxygenase 2/metabolism , Epithelial Cells/enzymology , Esophagus/enzymology , Oncogene Proteins, Viral/genetics , Repressor Proteins/genetics , Telomerase/genetics , Apoptosis , Cell Cycle , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Cyclooxygenase 2/genetics , Dinoprostone/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Esophagus/pathology , Esophagus/virology , Humans , Karyotyping , Neoplastic Stem Cells/enzymology , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Repressor Proteins/metabolism , Retinoblastoma Protein/metabolism , Telomerase/metabolism , Time Factors , Transfection , Tumor Suppressor Protein p53/metabolism , Up-Regulation , bcl-2-Associated X Protein/metabolism
14.
Cancer Res ; 62(6): 1662-8, 2002 Mar 15.
Article in English | MEDLINE | ID: mdl-11912137

ABSTRACT

Chromosome instability is a commonly observed feature in ovarian carcinoma. Mitotic checkpoint controls are thought to be essential for accurate chromosomal segregation, and MAD2 is a key component of this checkpoint. In this study, we investigated the competence of the mitotic checkpoint and its relationship to the expression of MAD2 protein in seven ovarian cancer cell lines. We found that a significant number (43%, three of seven cell lines) of the tested ovarian cancer cells failed to arrest in the G(2)-M phase of the cell cycle in response to microtubule disruption. This loss of mitotic checkpoint control was associated with reduced expression of the MAD2 protein. To additionally understand the significance of the MAD2 to mitotic checkpoint control, we established an inducible expression system in which MAD2 was induced by the addition of ponasterone A. Notably, the induced expression of MAD2 in two checkpoint-defective ovarian cancer cell lines led to the restoration of mitotic checkpoint response to spindle-disrupting agents. Taken together, our findings suggest that the steady-state amount of MAD2 inside cells may represent a molecular switch for mitotic checkpoint control. This provides a novel insight into the molecular basis of CIN in ovarian carcinoma and has implications for effective use of checkpoint-targeting drugs.


Subject(s)
Calcium-Binding Proteins/biosynthesis , Mitosis/physiology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Calcium-Binding Proteins/genetics , Cell Cycle/physiology , Cell Cycle Proteins , Female , Humans , Mad2 Proteins , Ovarian Neoplasms/genetics , Plasmids/genetics , Repressor Proteins , Transfection , Tumor Cells, Cultured
15.
Oncogene ; 22(1): 109-16, 2003 Jan 09.
Article in English | MEDLINE | ID: mdl-12527913

ABSTRACT

Mitotic arrest deficient 2 (MAD2) is thought to be a key component of the mitotic checkpoint, which ensures accurate chromosome segregation. Reduced expression of MAD2 protein is associated with mitotic checkpoint abrogation and chromosomal instability in certain types of human cancers. To explore the possibility of developing a novel strategy for the treatment of cancer based on selective killing of mitotic checkpoint-defective or -competent cells, here we have investigated the effect of MAD2 expression on cellular sensitivity to checkpoint-targeting anticancer drugs. We reintroduced MAD2 protein in a mitotic checkpoint-defective nasopharyngeal carcinoma cell line, CNE2, using an inducible expression vector. We found that overexpression of MAD2 led to an increased sensitivity to vincristine, which was accompanied by increased mitotic index and G2/M cell cycle arrest. In addition, increased phosphorylation of Raf, MEK1/2 and Bcl-2 was observed in MAD2-overexpressing cells in response to vincristine. Furthermore, inhibition of phosphorylation of MEK1/2 by its inhibitor PD098059 led to reduced sensitivity to vincristine, which was associated with decreased Bcl-2 phosphorylation. Our data suggest a role for MAD2 in the sensitization of cancer cells to certain mitotic checkpoint-targeting anticancer drugs.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Calcium-Binding Proteins/physiology , Carrier Proteins , Fungal Proteins/physiology , Mitosis/drug effects , Nasopharyngeal Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-raf/metabolism , Vincristine/pharmacology , Cell Cycle Proteins , Humans , Mitogen-Activated Protein Kinases/metabolism , Nuclear Proteins , Phosphorylation , Tumor Cells, Cultured
16.
Biochim Biophys Acta ; 1590(1-3): 150-8, 2002 Jun 12.
Article in English | MEDLINE | ID: mdl-12063178

ABSTRACT

Nasopharyngeal carcinoma (NPC) is a common cancer in Southeast Asia, especially in southern China. One of the most striking features of this disease is its close relationship with Epstein-Barr Virus (EBV). However, to date there is no direct study on the mechanisms involved in the role of EBV in the tumorigenesis of NPC, largely due to lack of an experimental model. Available hypotheses on the association between EBV and NPC are generated from non-nasopharyngeal epithelial cell systems such as human keratinocytes or mouse epithelial cells, which may not truly represent the biological properties of nasopharyngeal epithelial (NP) cells. In this study, we report the establishment of two immortalized NP cell lines, NP69SV40T and NP39E6/E7, using SV40T and HPV16E6/E7 oncogenes. We found that NP60SV40T and NP39E6/E7 cell lines not only maintained many characteristics of normal NP cells (i.e. keratin profile and responsive to TGFbeta inhibition) but also highly responsive to one of the EBV encoded genes, LMP1. Comparative genome hybridization (CGH) analysis showed that these two cell lines contained multiple genetic alterations, some of which have been described in NPC. The immortalized NP cell lines are non-tumorigenic and exhibit anchorage-dependent growth. These cell lines may provide a possible cell model system for studying the mechanisms involved in the tumorigenesis of NPC.


Subject(s)
Nasopharynx/cytology , Repressor Proteins , Antigens, Polyomavirus Transforming/genetics , Cell Division , Cell Line, Transformed , Chromosomes, Human/genetics , Enzyme Activation , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Genes, Viral , Herpesvirus 4, Human/pathogenicity , Humans , Models, Biological , Nasopharyngeal Neoplasms/etiology , Nasopharynx/drug effects , Nasopharynx/metabolism , Nucleic Acid Hybridization , Oncogene Proteins, Viral/genetics , Papillomavirus E7 Proteins , Simian virus 40/genetics , Telomerase/metabolism , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta1 , Viral Matrix Proteins/metabolism
17.
Cancer Genet Cytogenet ; 163(1): 30-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16271953

ABSTRACT

Extravillous cytotrophoblast (EVCT) cultures from the normal placentas of three pregnant women were transfected by HPVE6E7. Sequential cytogenetic and molecular analyses were performed to delineate genetic events that may be critical for cell immortalization. One line, PE1-E6E7, was immortalized successfully, whereas 2 other lines, PE3-E6E7 and PE4-E6E7, could not be maintained beyond crisis. Before crisis, the majority of cells in all lines were karyotypically normal. During the early stages of crisis, there was progressive telomere shortening. Most cells were karyotypically abnormal, with extreme cytogenetic divergence and a predominance of telomeric association and dicentric chromosomes affecting many chromosomes. At the later stages of crisis, the karyotype became more convergent with a drastic decrease in nonclonal aberrations. In PE1-E6E7, after crisis the karyotype was complex, with frequent centromeric rearrangements in the form of isochromosomes and whole-arm translocations. There were unbalanced structural aberrations and numerical changes, including loss of chromosome 13, that could be traced throughout the evolution of the line. These findings support the concept that immortalization is a relatively rare and nonrandom event that occurs only in cells that have acquired the necessary or critical genetic alterations. Telomeric dysfunction may be an important mechanism leading to the acquisition of complex karyotypical aberrations.


Subject(s)
Chorionic Villi/ultrastructure , Genes, Viral , Papillomaviridae/genetics , Cells, Cultured , Chorionic Villi/virology , Chromosome Aberrations , Chromosome Banding , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Placenta/cytology , Placenta/virology , Pregnancy , Telomere/genetics , Transfection , Translocation, Genetic
18.
J Mol Diagn ; 6(4): 326-34, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15507671

ABSTRACT

The methylation status of genes in hydatidiform mole and choriocarcinoma and its significance is relatively unexplored. We investigated the methylation status of the promoter regions of six genes, p16, HIC-1, TIMP3, GSTP1, death-associated protein kinase (DAPK), and E-cadherin in 54 hydatidiform moles, five choriocarcinomas, and 10 first trimester placenta by methylation-specific polymerase chain reaction (PCR). Immunohistochemical expression of p16, TIMP3, and E-cadherin, and quantitative real-time RT-PCR of p16 was also performed. Among the six genes examined, the promoter region of four genes (E-cadherin, HIC-1, p16, TIMP3) in choriocarcinoma and three genes (E-cadherin, HIC-1, p16) in hydatidiform mole exhibited aberrant methylation whereas none was hypermethylated in normal placenta. There was a significant correlation between methylation and reduced expression of p16, E-cadherin, and TIMP3 (P < 0.001). Fifteen of the 54 patients with hydatidiform mole developed gestational trophoblastic neoplasia requiring chemotherapy. Promoter hypermethylation of p16 alone, or combined with E-cadherin, was significantly correlated to such development (P = 0.001, 0.0005, respectively). Hypermethylation of multiple genes, especially p16, might be related to the subsequent development of gestational trophoblastic neoplasia.


Subject(s)
Choriocarcinoma/genetics , DNA Methylation , Hydatidiform Mole/genetics , Promoter Regions, Genetic , Apoptosis Regulatory Proteins , Cadherins/genetics , Calcium-Calmodulin-Dependent Protein Kinases/genetics , Cyclin-Dependent Kinase Inhibitor p16/genetics , DNA Primers/chemistry , DNA-Binding Proteins , Death-Associated Protein Kinases , Female , Glutathione S-Transferase pi , Glutathione Transferase/genetics , Humans , Immunohistochemistry , Isoenzymes/genetics , Kruppel-Like Transcription Factors , Placenta/metabolism , Polymerase Chain Reaction , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction/methods , Tissue Inhibitor of Metalloproteinase-3/genetics , Transcription Factors/genetics
19.
Anticancer Res ; 24(2B): 881-6, 2004.
Article in English | MEDLINE | ID: mdl-15161041

ABSTRACT

Gastric cancer is the second most common cause of cancer-related death worldwide and the highest incidence of this cancer has been reported in Asia, especially in China. Identification of early stage lesions is vital in achieving high survival rate. However, due to the lack of reliable biomarkers, the majority of gastric cancer is presented at an advanced stage. Recently, it has been reported that Id-1, a helix-loop-helix protein, may be a valuable diagnostic marker in many types of human cancer. In this study, we evaluated Id-1 protein expression in gastric cancer specimens and compared it with non-malignant tissues. In addition, to investigate whether Id-1 expression levels were associated with the aggressiveness of this disease as implicated in other cancer types, we also assessed Id-1 expression levels in primary tumours and their lymph node metastasized lesions. Our results indicate that up-regulation of Id-1 is a frequent event in gastric cancer but its expression levels are not associated with tumour metastasis. Our evidence provides a possible novel marker for the diagnosis of gastric cancer.


Subject(s)
Adenocarcinoma/metabolism , Biomarkers, Tumor/biosynthesis , Repressor Proteins , Stomach Neoplasms/metabolism , Transcription Factors/biosynthesis , Adenocarcinoma/pathology , Adult , Aged , Female , Humans , Immunohistochemistry , Inhibitor of Differentiation Protein 1 , Lymphatic Metastasis , Male , Middle Aged , Stomach Neoplasms/pathology
20.
Yi Chuan Xue Bao ; 29(6): 476-80, 2002 Jun.
Article in Zh | MEDLINE | ID: mdl-12096622

ABSTRACT

Lung cancer is one of the most common malignant tumors in humans. Metastasis is the basic biological feature of malignant tumors, which is the main cause of death. Molecular mechanism of metastasis is still unclear, although lots of studies have been done in tumor metastasis. To study and explore the molecular basis of metastasis in lung cancer, and isolate tumor metastasis-related genes, two human lung adenocarcinoma cell lines AGZY 83-a and Anip 973 were chosen as research materials. The Anip973 was derived from AGZY83-a, but manifested much higher metastasis potential than the parent line. Using mRNA differential display technique, an unknown cDNA fragment, OPB7-1, which is over-expressive in Anip973 cell line, was obtained. It was used as a template to isolate its corresponding cDNA through dbEST searching and PCR. To search and clone lung adenocarcinoma metastasis-related candidate gene, and to explore the molecular basis of development of lung carcinoma, differential expression of OPB7-1 cDNA fragment among 9 human lung adenocarcinoma cell lines and 12 normal human tissues were detected using cell culture, cDNA clone, Northern blot analysis and bioinformation technology. Results showed that there were significant differences in OPB7-1 expression among 9 human lung adenocarcinoma cell lines. High expression tendency was observed in Anip973 cell line with high metastasis potential, TKB-18 cell line with high invasion potential and GLC-82 cell line with low differentiation potential. Besides, a bigger fragment can be found in Anip973 cell line on the Northern blot hybridization. The 3.0 kb transcriptions were found in various tissues. Over-expression in heart and skeletal muscle could be observed, whereas expression in spleen, liver, kidney, placental and lung could be found except colon, thyroid gland and small intestine. These manifests indicate that OPB7-1 gene has a wide-rage expression in human multiple tissues. A 1.0 kb cDNA fragment was acquired by linking up EST fragments homologous match 5' end and PCR. BLAST analysis revealed that OPB7-1 gene has extremely low sequence identity with any known genes from GenBank and any sequences from EST database. The chromosomal localization of it was determined by RH location method. The OPB7-1 fragment was localized to chromosome 1p31-34. That OPB7-1 gene has an extensive expression pattern, may be a novel tumor gene related to lung carcinoma. Further research needs to be done to obtain the full-length cDNA of OPB7-1 gene. It will be helpful to investigate the expression in lung cancer cases and other tumor tissues for further determining the function of OPB7-1 gene in development of tumor.


Subject(s)
Adenocarcinoma/genetics , Lung Neoplasms/genetics , Neoplasm Proteins/genetics , Base Sequence , Blotting, Northern , Chromosome Mapping , DNA Fragmentation , DNA, Complementary , Gene Expression , Humans , Molecular Sequence Data , Tissue Distribution , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL