Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
2.
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
3.
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.

4.
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
6.
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.

7.
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
8.
Cell Host Microbe ; 14(2): 125-35, 2013 Aug 14.
Article in English | MEDLINE | ID: mdl-23954152

ABSTRACT

Merkel cell polyomavirus (MCV) causes an aggressive human skin cancer, Merkel cell carcinoma, through expression of small T (sT) and large T (LT) viral oncoproteins. MCV sT is also required for efficient MCV DNA replication by the multifunctional MCV LT helicase protein. We find that LT is targeted for proteasomal degradation by the cellular SCF(Fbw7) E3 ligase, which can be inhibited by sT through its LT-stabilization domain (LSD). Consequently, sT also stabilizes cellular SCF(Fbw7) targets, including the cell-cycle regulators c-Myc and cyclin E. Mutating the sT LSD decreases LT protein levels and eliminates synergism in MCV DNA replication as well as sT-induced cell transformation. SCF(Fbw7) knockdown mimics sT-mediated stabilization of LT, but this knockdown is insufficient to fully reconstitute the transforming activity of a mutant LSD sT protein. Thus, MCV has evolved a regulatory system involving SCF(Fbw7) that controls viral replication but also contributes to host cell transformation.


Subject(s)
Antigens, Viral, Tumor/metabolism , Cell Cycle Proteins/antagonists & inhibitors , F-Box Proteins/antagonists & inhibitors , Host-Pathogen Interactions , Merkel cell polyomavirus/physiology , Oncogene Proteins/biosynthesis , Ubiquitin-Protein Ligases/antagonists & inhibitors , Virus Replication , Cell Line , F-Box-WD Repeat-Containing Protein 7 , Humans
9.
J Clin Invest ; 123(6): 2694-702, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23728175

ABSTRACT

MicroRNAs (miRNAs) are excellent tumor biomarkers because of their cell-type specificity and abundance. However, many miRNA detection methods, such as real-time PCR, obliterate valuable visuospatial information in tissue samples. To enable miRNA visualization in formalin-fixed paraffin-embedded (FFPE) tissues, we developed multicolor miRNA FISH. As a proof of concept, we used this method to differentiate two skin tumors, basal cell carcinoma (BCC) and Merkel cell carcinoma (MCC), with overlapping histologic features but distinct cellular origins. Using sequencing-based miRNA profiling and discriminant analysis, we identified the tumor-specific miRNAs miR-205 and miR-375 in BCC and MCC, respectively. We addressed three major shortcomings in miRNA FISH, identifying optimal conditions for miRNA fixation and ribosomal RNA (rRNA) retention using model compounds and high-pressure liquid chromatography (HPLC) analyses, enhancing signal amplification and detection by increasing probe-hapten linker lengths, and improving probe specificity using shortened probes with minimal rRNA sequence complementarity. We validated our method on 4 BCC and 12 MCC tumors. Amplified miR-205 and miR-375 signals were normalized against directly detectable reference rRNA signals. Tumors were classified using predefined cutoff values, and all were correctly identified in blinded analysis. Our study establishes a reliable miRNA FISH technique for parallel visualization of differentially expressed miRNAs in FFPE tumor tissues.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Basal Cell/diagnosis , Carcinoma, Merkel Cell/diagnosis , MicroRNAs/metabolism , Skin Neoplasms/diagnosis , Animals , Biomarkers, Tumor/genetics , Carcinoma, Basal Cell/metabolism , Carcinoma, Merkel Cell/metabolism , Cluster Analysis , Diagnosis, Differential , Fixatives/chemistry , Fluorescent Dyes/chemistry , Formaldehyde/chemistry , Gene Expression , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/isolation & purification , Molecular Diagnostic Techniques , Paraffin Embedding , RNA, Ribosomal, 28S/metabolism , Sequence Analysis, RNA , Signal-To-Noise Ratio , Skin Neoplasms/metabolism , Tissue Fixation
10.
J Virol Methods ; 187(1): 6-14, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23085629

ABSTRACT

Merkel cell carcinoma (MCC) is an aggressive skin cancer with a high mortality rate. The majority of MCC (70-80%) harbor clonally integrated Merkel cell polyomavirus (MCV) in the tumor genome and express viral T antigen oncoproteins. The characterization of an early passage MCV-positive MCC cell line MS-1 is described, and its cellular, immunohistochemical, and virological features to MCV-negative (UISO, MCC13, and MCC26) and MCV-positive cell lines (MKL-1 and MKL-2) were compared. The MS-1 cellular genome harbors integrated MCV, which preserves an identical viral sequence from its parental tumor. Neither VP2 gene transcripts nor VP1 protein are detectable in MS-1 or other MCV-positive MCC cell lines tested. Mapping of viral and cellular integration sites in MS-1 and MCC tumor samples demonstrates no consistent viral or cellular gene integration locus. All MCV-positive cell lines show cytokeratin 20 positivity and grow in suspension. When injected subcutaneously into NOD scid gamma (NSG) mice, MS-1 forms a discrete macroscopic tumor. Immunophenotypic analysis of the MS-1 cell line and xenografts in mice show identical profiles to the parental tumor biopsy. Hence, MS-1 is an early passage cell line that provides a useful in vitro model to characterize MCV-positive MCC.


Subject(s)
Carcinoma, Merkel Cell/virology , Merkel cell polyomavirus/growth & development , Skin Neoplasms/virology , Animals , Antigens, Viral, Tumor/analysis , Cell Line, Tumor/virology , Cell Lineage , Humans , Keratin-20/biosynthesis , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Polyomavirus Infections/virology , Transplantation, Heterologous , Tumor Virus Infections/virology , Viral Structural Proteins/analysis , Virus Integration/genetics
11.
Sci Transl Med ; 4(133): 133ra56, 2012 May 09.
Article in English | MEDLINE | ID: mdl-22572880

ABSTRACT

Merkel cell polyomavirus (MCV) causes ~80% of primary and metastatic Merkel cell carcinomas (MCCs). By comparing digital transcriptome subtraction deep-sequencing profiles, we found that transcripts of the cellular survivin oncoprotein [BIRC5a (baculoviral inhibitor of apoptosis repeat-containing 5)] were up-regulated sevenfold in virus-positive compared to virus-negative MCC tumors. Knockdown of MCV large T antigen in MCV-positive MCC cell lines decreased survivin mRNA and protein expression. Exogenously expressed MCV large T antigen increased survivin protein expression in non-MCC primary cells. This required an intact retinoblastoma protein-targeting domain that activated survivin gene transcription as well as expression of other G(1)-S-phase proteins including E2F1 and cyclin E. Survivin expression is critical to the survival of MCV-positive MCC cells. A small-molecule survivin inhibitor, YM155, potently and selectively initiates irreversible, nonapoptotic, programmed MCV-positive MCC cell death. Of 1360 other chemotherapeutic and pharmacologically active compounds screened in vitro, only bortezomib (Velcade) was found to be similarly potent, but was not selective in killing MCV-positive MCC cells. YM155 halted the growth of MCV-positive MCC xenograft tumors and was nontoxic in mice, whereas bortezomib was not active in vivo and mice displayed serious morbidity. Xenograft tumors resumed growth once YM155 treatment was stopped, suggesting that YM155 may be cytostatic rather than cytotoxic in vivo. Identifying the cellular pathways, such as those involving survivin, that are targeted by tumor viruses can lead to rapid and rational identification of drug candidates for treating virus-induced cancers.


Subject(s)
Carcinoma, Merkel Cell/drug therapy , Carcinoma, Merkel Cell/metabolism , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Inhibitor of Apoptosis Proteins/metabolism , Polyomavirus Infections/metabolism , Tumor Virus Infections/metabolism , Animals , Antigens, Viral, Tumor/genetics , Antineoplastic Agents/pharmacology , Base Sequence , Boronic Acids/pharmacology , Bortezomib , Carcinoma, Merkel Cell/genetics , Carcinoma, Merkel Cell/virology , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Imidazoles/pharmacology , Inhibitor of Apoptosis Proteins/genetics , Merkel cell polyomavirus/genetics , Merkel cell polyomavirus/pathogenicity , Mice , Mice, Inbred NOD , Mice, SCID , Naphthoquinones/pharmacology , Polyomavirus Infections/genetics , Polyomavirus Infections/therapy , Polyomavirus Infections/virology , Pyrazines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Survivin , Translational Research, Biomedical , Tumor Virus Infections/genetics , Tumor Virus Infections/therapy , Tumor Virus Infections/virology , Xenograft Model Antitumor Assays
12.
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
13.
J Clin Invest ; 121(9): 3623-34, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21841310

ABSTRACT

Merkel cell polyomavirus (MCV) is the recently discovered cause of most Merkel cell carcinomas (MCCs), an aggressive form of nonmelanoma skin cancer. Although MCV is known to integrate into the tumor cell genome and to undergo mutation, the molecular mechanisms used by this virus to cause cancer are unknown. Here, we show that MCV small T (sT) antigen is expressed in most MCC tumors, where it is required for tumor cell growth. Unlike the closely related SV40 sT, MCV sT transformed rodent fibroblasts to anchorage- and contact-independent growth and promoted serum-free proliferation of human cells. These effects did not involve protein phosphatase 2A (PP2A) inhibition. MCV sT was found to act downstream in the mammalian target of rapamycin (mTOR) signaling pathway to preserve eukaryotic translation initiation factor 4E-binding protein 1 (4E-BP1) hyperphosphorylation, resulting in dysregulated cap-dependent translation. MCV sT-associated 4E-BP1 serine 65 hyperphosphorylation was resistant to mTOR complex (mTORC1) and mTORC2 inhibitors. Steady-state phosphorylation of other downstream Akt-mTOR targets, including S6K and 4E-BP2, was also increased by MCV sT. Expression of a constitutively active 4E-BP1 that could not be phosphorylated antagonized the cell transformation activity of MCV sT. Taken together, these experiments showed that 4E-BP1 inhibition is required for MCV transformation. Thus, MCV sT is an oncoprotein, and its effects on dysregulated cap-dependent translation have clinical implications for the prevention, diagnosis, and treatment of MCV-related cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antigens, Polyomavirus Transforming/metabolism , Carcinoma, Merkel Cell/metabolism , Merkel Cells/virology , Oncogene Proteins/metabolism , Phosphoproteins/metabolism , Polyomavirus/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antigens, Polyomavirus Transforming/genetics , Carcinoma, Merkel Cell/pathology , Carcinoma, Merkel Cell/virology , Cell Cycle Proteins , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic , Humans , Oncogene Proteins/genetics , Phosphoproteins/genetics , Phosphorylation , Polyomavirus/pathogenicity , Protein Biosynthesis , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , TOR Serine-Threonine Kinases/metabolism
14.
PLoS One ; 6(7): e22468, 2011.
Article in English | MEDLINE | ID: mdl-21799863

ABSTRACT

Merkel cell polyomavirus (MCV), a previously unrecognized component of the human viral skin flora, was discovered as a mutated and clonally-integrated virus inserted into Merkel cell carcinoma (MCC) genomes. We reconstructed a replicating MCV clone (MCV-HF), and then mutated viral sites required for replication or interaction with cellular proteins to examine replication efficiency and viral gene expression. Three days after MCV-HF transfection into 293 cells, although replication is not robust, encapsidated viral DNA and protein can be readily isolated by density gradient centrifugation and typical ∼40 nm diameter polyomavirus virions are identified by electron microscopy. The virus has an orderly gene expression cascade during replication in which large T (LT) and 57kT proteins are first expressed by day 2, followed by expression of small T (sT) and VP1 proteins. VP1 and sT proteins are not detected, and spliced 57kT is markedly diminished, in the replication-defective virus suggesting that early gene splicing and late gene transcription may be dependent on viral DNA replication. MCV replication and encapsidation is increased by overexpression of MCV sT, consistent with sT being a limiting factor during virus replication. Mutation of the MCV LT vacuolar sorting protein hVam6p (Vps39) binding site also enhances MCV replication while exogenous hVam6p overexpression reduces MCV virion production by >90%. Although MCV-HF generates encapsidated wild-type MCV virions, we did not find conditions for persistent transmission to recipient cell lines suggesting that MCV has a highly restricted tropism. These studies identify and highlight the role of polyomavirus DNA replication in viral gene expression and show that viral sT and cellular hVam6p are important factors regulating MCV replication. MCV-HF is a molecular clone that can be readily manipulated to investigate factors affecting MCV replication.


Subject(s)
Merkel cell polyomavirus/physiology , Virus Replication , Antigens, Polyomavirus Transforming/chemistry , Antigens, Polyomavirus Transforming/genetics , Antigens, Polyomavirus Transforming/metabolism , Autophagy-Related Proteins , Cloning, Molecular , Conserved Sequence , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Merkel cell polyomavirus/genetics , Merkel cell polyomavirus/immunology , Merkel cell polyomavirus/metabolism , Mutation , Protein Structure, Tertiary , Vesicular Transport Proteins/metabolism , Virus Replication/genetics
15.
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
16.
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
17.
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
18.
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
19.
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
20.
Science ; 319(5866): 1096-100, 2008 Feb 22.
Article in English | MEDLINE | ID: mdl-18202256

ABSTRACT

Merkel cell carcinoma (MCC) is a rare but aggressive human skin cancer that typically affects elderly and immunosuppressed individuals, a feature suggestive of an infectious origin. We studied MCC samples by digital transcriptome subtraction and detected a fusion transcript between a previously undescribed virus T antigen and a human receptor tyrosine phosphatase. Further investigation led to identification and sequence analysis of the 5387-base-pair genome of a previously unknown polyomavirus that we call Merkel cell polyomavirus (MCV or MCPyV). MCV sequences were detected in 8 of 10 (80%) MCC tumors but only 5 of 59 (8%) control tissues from various body sites and 4 of 25 (16%) control skin tissues. In six of eight MCV-positive MCCs, viral DNA was integrated within the tumor genome in a clonal pattern, suggesting that MCV infection and integration preceded clonal expansion of the tumor cells. Thus, MCV may be a contributing factor in the pathogenesis of MCC.


Subject(s)
Carcinoma, Merkel Cell/virology , Polyomavirus Infections/virology , Polyomavirus/genetics , Skin Neoplasms/virology , Tumor Virus Infections/virology , Virus Integration , Antigens, Polyomavirus Transforming/genetics , Carcinoma, Merkel Cell/genetics , Gene Library , Genetic Techniques , Genome, Viral , Humans , Polyomavirus/isolation & purification , Polyomavirus/pathogenicity , Polyomavirus/physiology , Polyomavirus Infections/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 5/genetics , Skin Neoplasms/genetics , Tumor Virus Infections/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...