Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
1.
Zhongguo Fei Ai Za Zhi ; 24(5): 351-356, 2021 May 20.
Article in Chinese | MEDLINE | ID: mdl-34034459

ABSTRACT

BACKGROUND: Studies have shown that elevated serum lactate dehydrogenase (LDH) concentration can lead to poor prognosis in patients with small cell lung cancer and lung adenocarcinoma, but its relationship with the prognosis of patients with lung large-cell neuroendocrine carcinoma (L-LCNEC) is not clear. This study aims to explore the influence of L-LCNEC preoperative serum LDH concentration and postoperative LDH concentration change trend on the disease-free survival (DFS) of patients after surgery, so as to judge the clinical prognosis of L-LCNEC provides new ideas. METHODS: Collected the clinical data. The receiver operating characteristic (ROC) curve was used to determine the optimal cut-off value, while the Kaplan-Meier and Cox proportional hazard model were used to analyze data. RESULTS: DFS was shortened in patients with high serum LDH concentration before operation and increased LDH concentration after operation (P<0.001, P<0.001). The preoperative LDH concentration and postoperative LDH concentration change trend were independent prognostic factors for patients (P<0.001, P=0.037). CONCLUSIONS: Preoperative LDH concentration and its postoperative concentration change trend in patients with L-LCNEC are independent prognostic factors for DFS of patients.


Subject(s)
Carcinoma, Large Cell/enzymology , Carcinoma, Neuroendocrine/enzymology , L-Lactate Dehydrogenase/blood , Lung Neoplasms/enzymology , Adult , Aged , Aged, 80 and over , Carcinoma, Large Cell/blood , Carcinoma, Large Cell/mortality , Carcinoma, Large Cell/surgery , Carcinoma, Neuroendocrine/blood , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/surgery , Disease-Free Survival , Female , Humans , Lung Neoplasms/blood , Lung Neoplasms/mortality , Lung Neoplasms/surgery , Male , Middle Aged , ROC Curve , Retrospective Studies
2.
Sci Rep ; 11(1): 9785, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33963267

ABSTRACT

In the last two decades, various therapies have been introduced for lung carcinoma patients, including tyrosine-kinase inhibitors for different mutations. While some of them are specific to specific tumor types, others, like NTRK1-3 fusions, are found in various solid tumors. The occurrence of an NTRK1,2 or 3 fusion acts as a biomarker for efficient treatment with NTRK inhibitors, irrespectively of the tumor type. However, the occurrence of the NTRK1-3 fusions in lung carcinomas is extremely rare. We performed a retrospective analysis to evaluate the applicability of immunohistochemistry with the pan-TRK antibody in the detection of NTRK fusions in lung carcinomas. The study cohort included 176 adenocarcinomas (AC), 161 squamous cell carcinomas (SCC), 31 large-cell neuroendocrine carcinomas (LCNEC), and 19 small cell lung carcinomas (SCLC). Immunohistochemistry (IHC) was performed using the pan-TRK antibody (clone EPR17341, Ventana) on tissue microarrays, while confirmation for all positive cases was done using RNA-based Archer FusionPlex MUG Lung Panel. On IHC staining, 12/387 samples (3.1%) demonstrated a positive reaction. Ten SCC cases (10/161, 6.2%), and two LCNEC cases (2/31, 6.5%) were positive. Positive cases demonstrated heterogeneous staining of tumor cells, mostly membranous with some cytoplasmic and in one case nuclear pattern. RNA-based sequencing did not demonstrate any NTRK1-3 fusion in our patients' collective. Our study demonstrates that pan-TRK expression in lung carcinoma is very low across different histologic types. NTRK1-3 fusions using an RNA-based sequencing approached could not be detected. This stresses the importance of confirmation of immunohistochemistry results by molecular methods.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Neuroendocrine , Carcinoma, Squamous Cell , Lung Neoplasms , Oncogene Proteins, Fusion , Receptor Protein-Tyrosine Kinases , Adenocarcinoma of Lung/enzymology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/pathology , Aged , Aged, 80 and over , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Carcinoma, Squamous Cell/enzymology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Female , Humans , Immunohistochemistry , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Retrospective Studies
3.
Cancer Sci ; 112(5): 1936-1942, 2021 May.
Article in English | MEDLINE | ID: mdl-33453146

ABSTRACT

Extrapulmonary neuroendocrine carcinoma (EPNEC) is a lethal disease with a poor prognosis. Platinum-based chemotherapy is used as the standard first-line treatment for unresectable EPNEC. Several retrospective studies have reported the results of the utilization of temozolomide (TMZ) as a drug for the second-line treatment for EPNEC. Patients with unresectable EPNEC that were resistant to platinum-based combination chemotherapy were recruited for a prospective phase II study of TMZ monotherapy. A 200 mg/m2 dose of TMZ was given from day 1 to day 5, every 4 weeks. Response rate (RR) was evaluated as the primary end-point. The presence of O6 -methylguanine DNA methyltransferase (MGMT) in EPNEC patients was also evaluated as exploratory research. Thirteen patients were enrolled in this study. Primary lesions were pancreas (n = 3), stomach (n = 3), duodenum (n = 1), colon (n = 1), gallbladder (n = 1), liver (n = 1), uterus (n = 1), bladder (n = 1), and primary unknown (n = 1). Each case was defined as pathological poorly differentiated neuroendocrine carcinoma from surgically resected and/or biopsied specimens. The median Ki-67 labeling index was 60% (range, 22%-90%). The RR was 15.4%, progression-free survival was 1.8 months (95% confidence interval [CI], 1.0-2.7), overall survival (OS) was 7.8 months (95% CI, 6.0-9.5), and OS from first-line treatment was 19.2 months (95% CI, 15.1-23.3). No grade 3 or 4 hematological toxicity had occurred and there was one case of grade 3 nausea. One case presented MGMT deficiency and this case showed partial response. Temozolomide monotherapy is a feasible, modestly effective, and safe treatment for patients with unresectable EPNEC following platinum-based chemotherapy, especially those with MGMT deficiency.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Temozolomide/therapeutic use , Adult , Aged , Antineoplastic Agents, Alkylating/administration & dosage , Antineoplastic Agents, Alkylating/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Colonic Neoplasms/pathology , Drug Administration Schedule , Duodenal Neoplasms/drug therapy , Duodenal Neoplasms/enzymology , Duodenal Neoplasms/pathology , Female , Gallbladder Neoplasms/drug therapy , Gallbladder Neoplasms/enzymology , Gallbladder Neoplasms/pathology , Humans , Ki-67 Antigen/analysis , Liver Neoplasms/drug therapy , Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Male , Middle Aged , O(6)-Methylguanine-DNA Methyltransferase/analysis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Progression-Free Survival , Prospective Studies , Stomach Neoplasms/drug therapy , Stomach Neoplasms/enzymology , Stomach Neoplasms/pathology , Temozolomide/administration & dosage , Temozolomide/adverse effects , Uterine Neoplasms/drug therapy , Uterine Neoplasms/enzymology , Uterine Neoplasms/pathology
4.
J Pathol ; 253(1): 106-118, 2021 01.
Article in English | MEDLINE | ID: mdl-33009820

ABSTRACT

Neuroendocrine prostate cancer (NEPC) is a more aggressive subtype of castration-resistant prostate cancer (CRPC). Although it is well established that PHF8 can enhance prostate cancer cell proliferation, whether PHF8 is involved in prostate cancer initiation and progression is relatively unclear. By comparing the transgenic adenocarcinoma of the mouse prostate (TRAMP) mice with or without Phf8 knockout, we systemically examined the role of PHF8 in prostate cancer development. We found that PHF8 plays a minimum role in initiation and progression of adenocarcinoma. However, PHF8 is essential for NEPC because not only is PHF8 highly expressed in NEPC but also animals without Phf8 failed to develop NEPC. Mechanistically, PHF8 transcriptionally upregulates FOXA2 by demethylating and removing the repressive histone markers on the promoter region of the FOXA2 gene, and the upregulated FOXA2 subsequently regulates the expression of genes involved in NEPC development. Since both PHF8 and FOXA2 are highly expressed in NEPC tissues from patients or patient-derived xenografts, the levels of PHF8 and FOXA2 can either individually or in combination serve as NEPC biomarkers and targeting either PHF8 or FOXA2 could be potential therapeutic strategies for NEPC treatment. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.


Subject(s)
Adenocarcinoma/enzymology , Biomarkers, Tumor/metabolism , Carcinoma, Neuroendocrine/enzymology , Epigenesis, Genetic , Hepatocyte Nuclear Factor 3-beta/metabolism , Histone Demethylases/metabolism , Prostatic Neoplasms/enzymology , Transcription Factors/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Animals , Biomarkers, Tumor/genetics , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/secondary , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Hepatocyte Nuclear Factor 3-beta/genetics , Histone Demethylases/genetics , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , PC-3 Cells , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Transcription Factors/genetics , Transcription, Genetic , Up-Regulation
6.
PLoS One ; 15(5): e0233720, 2020.
Article in English | MEDLINE | ID: mdl-32459817

ABSTRACT

Since patients with medullary thyroid cancer (MTC) often have metastatic disease at the time of diagnosis, the development of efficient systemic treatment options for MTC is important. Vandetanib and cabozantinib are two tyrosine kinase inhibitors (TKIs) that were recently approved by FDA and EMA for systemic treatment of metastatic MTC. Additionally, since MTC is of a neuroendocrine tumour type, treatment with radiolabelled somatostatin analogues (e.g. 177Lu-octreotate) is a valid option for patients with MTC. The aim of this study was to investigate the potentially increased therapeutic effect of combining radiation therapy with these TKIs for treatment of MTC in a mouse model. Nude mice carrying patient-derived MTC tumours (GOT2) were treated with external beam radiotherapy (EBRT) and/or one of the two TKIs vandetanib or cabozantinib. The tumour volume was determined and compared with that of mock-treated controls. The treatment doses were chosen to give a moderate effect as monotherapy to be able to detect any increased therapeutic effect from the combination therapy. At the end of follow-up, tumours were processed for immunohistochemical (IHC) analyses. The animals in the combination therapy groups showed the largest reduction in tumour volume and the longest time to tumour progression. Two weeks after start of treatment, the tumour volume for these mice was reduced by about 70-75% compared with controls. Furthermore, also EBRT and TKI monotherapy resulted in a clear anti-tumour effect with a reduced tumour growth compared with controls. The results show that an increased therapeutic effect could be achieved when irradiation is combined with TKIs for treatment of MTC. Future studies should evaluate the potential of using 177Lu-octreotate therapy in combination with TKIs in patients.


Subject(s)
Anilides/pharmacology , Carcinoma, Neuroendocrine/therapy , Chemoradiotherapy , Neoplasm Proteins/antagonists & inhibitors , Piperidines/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyridines/pharmacology , Quinazolines/pharmacology , Thyroid Neoplasms/therapy , Animals , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/pathology , Xenograft Model Antitumor Assays
7.
PLoS Comput Biol ; 15(4): e1006878, 2019 04.
Article in English | MEDLINE | ID: mdl-31026276

ABSTRACT

Drosophila provides an inexpensive and quantitative platform for measuring whole animal drug response. A complementary approach is virtual screening, where chemical libraries can be efficiently screened against protein target(s). Here, we present a unique discovery platform integrating structure-based modeling with Drosophila biology and organic synthesis. We demonstrate this platform by developing chemicals targeting a Drosophila model of Medullary Thyroid Cancer (MTC) characterized by a transformation network activated by oncogenic dRetM955T. Structural models for kinases relevant to MTC were generated for virtual screening to identify unique preliminary hits that suppressed dRetM955T-induced transformation. We then combined features from our hits with those of known inhibitors to create a 'hybrid' molecule with improved suppression of dRetM955T transformation. Our platform provides a framework to efficiently explore novel kinase inhibitors outside of explored inhibitor chemical space that are effective in inhibiting cancer networks while minimizing whole body toxicity.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Neuroendocrine , Drug Evaluation, Preclinical/methods , Protein Kinase Inhibitors/pharmacology , Protein Kinases , Thyroid Neoplasms , Animals , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/metabolism , Computational Biology/methods , Drosophila , Models, Biological , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/metabolism , Protein Kinases/drug effects , Protein Kinases/metabolism , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/metabolism
8.
J Clin Invest ; 129(4): 1641-1653, 2019 03 04.
Article in English | MEDLINE | ID: mdl-30721156

ABSTRACT

Hyperactivated AKT/mTOR signaling is a hallmark of pancreatic neuroendocrine tumors (PNETs). Drugs targeting this pathway are used clinically, but tumor resistance invariably develops. A better understanding of factors regulating AKT/mTOR signaling and PNET pathogenesis is needed to improve current therapies. We discovered that RABL6A, a new oncogenic driver of PNET proliferation, is required for AKT activity. Silencing RABL6A caused PNET cell-cycle arrest that coincided with selective loss of AKT-S473 (not T308) phosphorylation and AKT/mTOR inactivation. Restoration of AKT phosphorylation rescued the G1 phase block triggered by RABL6A silencing. Mechanistically, loss of AKT-S473 phosphorylation in RABL6A-depleted cells was the result of increased protein phosphatase 2A (PP2A) activity. Inhibition of PP2A restored phosphorylation of AKT-S473 in RABL6A-depleted cells, whereas PP2A reactivation using a specific small-molecule activator of PP2A (SMAP) abolished that phosphorylation. Moreover, SMAP treatment effectively killed PNET cells in a RABL6A-dependent manner and suppressed PNET growth in vivo. The present work identifies RABL6A as a new inhibitor of the PP2A tumor suppressor and an essential activator of AKT in PNET cells. Our findings offer what we believe is a novel strategy of PP2A reactivation for treatment of PNETs as well as other human cancers driven by RABL6A overexpression and PP2A inactivation.


Subject(s)
Carcinoma, Neuroendocrine/enzymology , Oncogene Proteins/metabolism , Pancreatic Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Tumor Suppressor Proteins/metabolism , rab GTP-Binding Proteins/metabolism , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor , Enzyme Activators/pharmacology , G1 Phase/drug effects , G1 Phase/genetics , Humans , Oncogene Proteins/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Phosphatase 2/genetics , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/genetics , rab GTP-Binding Proteins/genetics
9.
Anticancer Res ; 38(11): 6085-6090, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30396922

ABSTRACT

BACKGROUND: Cystathione ß-synthase (CBS) catalyzes the conversion of homocysteine and cysteine to hydrogen sulfide (H2S) and cystathione, via the trans-sulfuration pathway. CBS protein expression levels are increased in several different human malignancies, with increased protein expression correlating with parameters such as tumor stage, anaplasia, metastases, and chemotherapy resistance. MATERIALS AND METHODS: This study employed tissue microarrays to examine CBS expression in benign thyroid tissue, thyroid oncocytomas, thyroid follicular adenomas, and in follicular, papillary, anaplastic, and medullary thyroid carcinomas. RESULTS: CBS expression was increased in all thyroid carcinomas types compared to benign thyroid tissue, but not in thyroid follicular adenomas or oncocytomas. A similar pattern was observed for nicotinamide phosphoribosyltransferase (NAMPT) tissue microarray analysis comparing thyroid adenomas and follicular carcinomas. CONCLUSION: For the first time, we showed that an H2S-syntheszing enzyme plays a role in thyroid malignancies. Additionally, our data suggest that CBS and NAMPT immunohistochemistry may be useful in differentiating follicular adenomas from follicular carcinomas.


Subject(s)
Cystathionine beta-Synthase/biosynthesis , Thyroid Neoplasms/enzymology , Adenocarcinoma, Follicular/enzymology , Adenoma, Oxyphilic/enzymology , Carcinoma, Neuroendocrine/enzymology , Cytokines/biosynthesis , Humans , Hydrogen Sulfide/metabolism , Immunohistochemistry , Nicotinamide Phosphoribosyltransferase/biosynthesis , Thyroid Cancer, Papillary/enzymology , Thyroid Carcinoma, Anaplastic/enzymology , Tissue Array Analysis
10.
DNA Cell Biol ; 37(9): 758-766, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29969286

ABSTRACT

Current clinical anti-androgen therapies in advanced prostate cancer (PCa) are driving an increased incidence of neuroendocrine prostate cancer (NEPC), a histological variant exhibiting reduced androgen receptor levels and expression of neuroendocrine markers. The mechanisms underlying the development of NEPC are poorly understood. A set of available data from a well-validated xenograft model of NEPC was used to analyze the exact role of protein kinase (PK) played in the development of NEPC. Fifty-four actionable and druggable PKs, mainly enriched in PI3K-Akt, mTOR, and MAPK signaling pathways, were screened out from the drastically changed PKs during NEPC transdifferentiation. Further analysis based on the crosstalk of these above signaling pathways finally singled out 10 PKs considered drivers and therapeutic targets in the development and treatment of NEPC. In vitro, the variation trend of PK expression observed during NEPC transdifferentiation could be recapitulated in PCa cell lines with different malignant degree. The predicted kinase targets exhibited different sensibilities in the restriction of PC3 cell growth. Selective actionable and druggable PKs may act as drivers in the progression of NEPC, and most of them can be used as potential therapeutic targets in clinical practice.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Neuroendocrine/drug therapy , Computational Biology/methods , Databases, Factual , Gene Expression Regulation, Enzymologic , Prostatic Neoplasms/drug therapy , Protein Kinases/chemistry , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Cell Line, Tumor , Cell Transdifferentiation , Disease Progression , Gene Ontology , Humans , Male , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Protein Kinases/genetics
11.
Hum Pathol ; 79: 212-221, 2018 09.
Article in English | MEDLINE | ID: mdl-29885402

ABSTRACT

Medullary thyroid carcinoma (MTC) is a rare neoplasia derived from neural parafollicular C cells. MicroRNAs (miRNAs) are small regulatory RNAs with essential roles in the biology of cancers such as MTC and can be applied as diagnostic markers. According to previous studies, miR-144 and miR-34 and their two oncogenes target, mammalian target of rapamycin (mTOR) and AXL receptor tyrosine kinase (AXL), were selected for further investigations in our study. Thirty MTC samples as well as thirty adjacent normal thyroid tissues were applied in this study including 28 formalin-fixed, paraffin-embedded (FFPE) and 2 fresh-frozen MTC samples. RNA extraction and complementary DNA (cDNA) synthesis were performed for all samples. After primer pairs and probes were designed, real-time polymerase chain reaction (real-time PCR) method was used, and the results were analyzed using 2-ΔΔCt method. Receiver operating characteristic (ROC) curve analysis was applied to assess the diagnostic value of the two miRNAs. AXL protein level was measured in all clinical samples using enzyme-linked immunosorbent assay (ELISA) method. Both miRNAs were up-regulated in all clinical samples compared to the normal tissues. AXL was up-regulated in most clinical samples while mTOR was down-regulated in most samples. Furthermore, the level of AXL protein increased. ROC curve analysis demonstrated that increased expression of miR-34a and miR-144 in MTC patients had significant predictive value. The results demonstrated that high expression of miR-144 and miR-34a can be considered as biomarkers of MTC. However, there was no statistically significant correlation between the expression of these miRNAs and target genes in MTC clinical samples.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Neuroendocrine/genetics , MicroRNAs/genetics , Thyroid Neoplasms/genetics , Adolescent , Adult , Aged , Biomarkers, Tumor/analysis , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/pathology , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Predictive Value of Tests , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/analysis , Receptor Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , TOR Serine-Threonine Kinases/analysis , TOR Serine-Threonine Kinases/genetics , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/pathology , Up-Regulation , Young Adult , Axl Receptor Tyrosine Kinase
12.
Exp Mol Pathol ; 104(1): 26-28, 2018 02.
Article in English | MEDLINE | ID: mdl-29248665

ABSTRACT

Mixed morphology lung tumors are rare; this is the second report of a combined NSCLC and atypical carcinoid tumor. Next generation sequencing was performed on both histologically distinct patterns which identified that both components harbored a BRAF p.V600E mutation. Molecular studies inform our knowledge of the biology and aid in treatment decisions for mixed morphology lung cancers.


Subject(s)
Adenocarcinoma/genetics , Carcinoid Tumor/genetics , Lung Neoplasms/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Adenocarcinoma/enzymology , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Aged , Carcinoid Tumor/enzymology , Carcinoid Tumor/pathology , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , High-Throughput Nucleotide Sequencing , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Male , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins B-raf/metabolism
14.
Hum Pathol ; 49: 124-34, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26826419

ABSTRACT

The molecular alterations in colorectal poorly differentiated neuroendocrine carcinoma remain incompletely characterized, particularly with respect to mutations in BRAF and KRAS. We analyzed 32 colorectal poorly differentiated neuroendocrine carcinomas and 40 colorectal poorly differentiated conventional adenocarcinomas for mutations in KRAS and BRAF and for DNA mismatch repair protein abnormalities to correlate histopathology with molecular alterations and survival. Compared with poorly differentiated conventional adenocarcinoma, poorly differentiated neuroendocrine carcinoma frequently harbored BRAF mutations (59% versus 5%; P < .001) and less frequently demonstrated KRAS codon 12 or 13 mutations (17% versus 43%; P = .03). BRAF mutations were identified in both pure poorly differentiated neuroendocrine carcinoma (60%) and poorly differentiated neuroendocrine carcinoma associated with a signet ring cell adenocarcinoma component (82%). Most (93%) poorly differentiated neuroendocrine carcinomas demonstrated proficient DNA mismatch repair by either microsatellite instability polymerase chain reaction or DNA mismatch repair immunohistochemistry. Patients with poorly differentiated neuroendocrine carcinoma had a significantly worse overall survival compared with patients with poorly differentiated conventional adenocarcinoma (P < .001). There was no significant difference in overall survival between patients with pure poorly differentiated neuroendocrine carcinoma and patients with both poorly differentiated neuroendocrine carcinoma and adenocarcinoma components (P = .5). In conclusion, colorectal poorly differentiated neuroendocrine carcinomas frequently harbor BRAF mutations and are associated with poor overall survival.


Subject(s)
Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Carcinoma, Neuroendocrine/genetics , Cell Differentiation , Colorectal Neoplasms/genetics , Mutation , Proto-Oncogene Proteins B-raf/genetics , Adenocarcinoma/enzymology , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Biopsy , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/therapy , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , DNA Mismatch Repair , DNA Mutational Analysis , Female , Genetic Predisposition to Disease , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Microsatellite Instability , Middle Aged , Phenotype , Proportional Hazards Models , Proto-Oncogene Proteins p21(ras)/genetics , Time Factors , Treatment Outcome , Young Adult
15.
Genes Dev ; 30(2): 220-32, 2016 Jan 15.
Article in English | MEDLINE | ID: mdl-26773004

ABSTRACT

Proteases are important for regulating multiple tumorigenic processes, including angiogenesis, tumor growth, and invasion. Elevated protease expression is associated with poor patient prognosis across numerous tumor types. Several multigene protease families have been implicated in cancer, including cysteine cathepsins. However, whether individual family members have unique roles or are functionally redundant remains poorly understood. Here we demonstrate stage-dependent effects of simultaneously deleting cathepsin B (CtsB) and CtsS in a murine pancreatic neuroendocrine tumor model. Early in tumorigenesis, the double knockout results in an additive reduction in angiogenic switching, whereas at late stages, several tumorigenic phenotypes are unexpectedly restored to wild-type levels. We identified CtsZ, which is predominantly supplied by tumor-associated macrophages, as the compensatory protease that regulates the acquired tumor-promoting functions of lesions deficient in both CtsB and CtsS. Thus, deletion of multiple cathepsins can lead to stage-dependent, compensatory mechanisms in the tumor microenvironment, which has potential implications for the clinical consideration of selective versus pan-family cathepsin inhibitors in cancer.


Subject(s)
Carcinoma, Neuroendocrine/enzymology , Cathepsins/genetics , Cathepsins/metabolism , Gene Deletion , Pancreatic Neoplasms/enzymology , Animals , Apoptosis/genetics , Carcinogenesis/genetics , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/physiopathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Macrophages/enzymology , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness/genetics , Neovascularization, Pathologic/enzymology , Neovascularization, Pathologic/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/physiopathology
16.
J Cell Physiol ; 231(8): 1784-95, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26660542

ABSTRACT

The aim of this paper is based on the use of a hyaluronic acid hydrogel of Quercetin tested alone and in combination to an inhibitor of Aurora Kinase type A and B (SNS-314) on human medullary and papillary thyroid cancer cells. Biological investigations were focused on the cellular uptake of the hydrogel, cell viability, antioxidant, and cytokines secretion studies. Quercetin delivered from hydrogel show a time and CD44 dependent interaction with both cell lines with significant anti-inflammatory effects. Combination of Quercetin and SNS-314 leads to a synergistic cytotoxic effect on medullary TT and papillary BCPAP cell lines with a significant reduction of the IC50 value. These results, highlights the importance of synergistic effect of the hyaluronic acid hydrogel of Quercetin with SNS-314 in the regulation of human thyroid cancer cell proliferation and emphasize the anti-tumor activity of these molecules. J. Cell. Physiol. 231: 1784-1795, 2016. © 2015 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aurora Kinase A/metabolism , Aurora Kinase B/antagonists & inhibitors , Carcinoma, Neuroendocrine/drug therapy , Carcinoma/drug therapy , Drug Carriers , Hyaluronic Acid/chemistry , Phenylurea Compounds/pharmacology , Protein Kinase Inhibitors/pharmacology , Quercetin/pharmacology , Thiazoles/pharmacology , Thyroid Neoplasms/drug therapy , Anti-Inflammatory Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/metabolism , Antioxidants/metabolism , Aurora Kinase A/genetics , Aurora Kinase B/metabolism , Carcinoma/enzymology , Carcinoma/pathology , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/pathology , Carcinoma, Papillary , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemistry, Pharmaceutical , Cytokines/metabolism , Dose-Response Relationship, Drug , Drug Synergism , Humans , Hyaluronan Receptors/metabolism , Hydrogels , Inhibitory Concentration 50 , Phenylurea Compounds/chemistry , Phenylurea Compounds/metabolism , Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/metabolism , Quercetin/chemistry , Quercetin/metabolism , Thiazoles/chemistry , Thiazoles/metabolism , Thyroid Cancer, Papillary , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/pathology , Time Factors
17.
Thyroid ; 26(3): 414-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26701095

ABSTRACT

BACKGROUND: Treatment of advanced medullary thyroid carcinoma (MTC) was recently improved with the approval of vandetanib and cabozantinib. However, there is still a need to explore sequential therapy with more than one tyrosine kinase inhibitor (TKI) and to explore alternative therapies when vandetanib and cabozantinib are not available. This study reports the authors' experience with sorafenib as a treatment for advanced MTC. METHODS: This is a retrospective longitudinal study of 13 patients with progressive metastatic MTC treated with sorafenib 400 mg twice daily between December 2011 and January 2015. The primary endpoints were to evaluate response and progression-free survival (PFS) in patients treated with sorafenib outside a clinical trial. The secondary endpoint was an assessment of the toxicity profile. One patient was excluded because of a serious allergic skin rash one week after starting sorafenib. RESULTS: The analysis included 12 patients with metastatic MTC (median age 48 years), 10 with sporadic and 2 with hereditary disease. The median duration of treatment was 11 months, and the median follow-up was 15.5 months. At data cutoff, 2/12 (16%) patients were still on treatment for 16 and 34 months. According to Response Evaluation Criteria in Solid Tumors analysis, 10 (83.3%) patients showed stable disease, and two (16.6%) had progression of disease; no partial response was observed. The median PFS was nine months. However, three patients with extensive and rapidly progressive disease died within three months of sorafenib treatment. The median PFS excluding these three patients was 12 months. Adverse events (AE) occurred in nine (75%) patients. The main AEs were skin toxicity, weight loss, and fatigue. Five (41.6%) patients needed dose reduction, and one patient discontinued treatment because of toxicity. CONCLUSIONS: Treatment with sorafenib in progressive metastatic MTC is well tolerated and resulted in disease control and durable clinical benefit in 75% of patients. Sorafenib treatment could be considered when vandetanib and cabozantinib are not available or after failing these drugs.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Thyroid Neoplasms/drug therapy , Adult , Aged , Antineoplastic Agents/adverse effects , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/secondary , Disease Progression , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Longitudinal Studies , Male , Middle Aged , Niacinamide/adverse effects , Niacinamide/therapeutic use , Phenylurea Compounds/adverse effects , Protein Kinase Inhibitors/adverse effects , Retrospective Studies , Sorafenib , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology , Time Factors , Treatment Outcome
18.
Recent Results Cancer Res ; 204: 227-49, 2015.
Article in English | MEDLINE | ID: mdl-26494392

ABSTRACT

Two independent events--the identification of activating mutations of the RET proto-oncogene, a receptor tyrosine kinase, in medullary thyroid carcinoma, and the recognition that small organic molecules could bind to and inhibit phosphorylation of signaling molecules, thereby inactivating the pathway-led to the recognition that kinase inhibitors could be used to treat medullary thyroid carcinoma (MTC). The introduction of these compounds into clinical practice has transformed the treatment of metastatic MTC and provided insight into the mechanisms by which RET causes C-cell transformation. This chapter will review the progress in this field over the past 7 years.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Neuroendocrine/drug therapy , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Thyroid Neoplasms/drug therapy , Animals , Antineoplastic Agents/adverse effects , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/genetics , Carcinoma, Neuroendocrine/pathology , Genetic Predisposition to Disease , Humans , Molecular Targeted Therapy , Mutation , Patient Selection , Phenotype , Protein Kinase Inhibitors/adverse effects , Proto-Oncogene Mas , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Risk Factors , Signal Transduction/drug effects , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/pathology , Treatment Outcome
19.
J Cancer Res Clin Oncol ; 141(12): 2159-70, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25989941

ABSTRACT

BACKGROUND: As the comprehensive genomic analysis of small cell lung cancer (SCLC) progresses, novel treatments for this disease need to be explored. With attention to the direct connection between the receptor tyrosine kinases (RTKs) of tumor cells and the pharmacological effects of specific inhibitors, we systematically assessed the RTK expressions of high-grade neuroendocrine carcinomas of the lung [HGNECs, including SCLC and large cell neuroendocrine carcinoma (LCNEC)]. PATIENTS AND METHODS: Fifty-one LCNEC and 61 SCLC patients who underwent surgical resection were enrolled in this research. As a control group, 202 patients with adenocarcinomas (ADCs) and 122 patients with squamous cell carcinomas (SQCCs) were also analyzed. All the tumors were stained with antibodies for 10 RTKs: c-Kit, EGFR, IGF1R, KDR, ERBB2, FGFR1, c-Met, ALK, RET, and ROS1. RESULTS: The LCNEC and SCLC patients exhibited similar clinicopathological characteristics. The IHC scores for each RTK were almost equivalent between the LCNEC and SCLC groups, but they were significantly different from those of the ADC or SQCC groups. In particular, c-Kit was the only RTK that was remarkably expressed in both LCNECs and SCLCs. On the other hand, about 20 % of the HGNEC tumors exhibited strongly positive RTK expression, and this rate was similar to those for the ADC and SQCC tumors. Intriguingly, strongly positive RTKs were almost mutually exclusive in individual tumors. CONCLUSIONS: Compared with ADC or SQCC, LCNEC and SCLC had similar expression profiles for the major RTKs. The exclusive c-Kit positivity observed among HGNECs suggests that c-Kit might be a distinctive RTK in HGNEC.


Subject(s)
Adenocarcinoma/enzymology , Biomarkers, Tumor/metabolism , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Squamous Cell/enzymology , Lung Neoplasms/enzymology , Proto-Oncogene Proteins c-kit/metabolism , Small Cell Lung Carcinoma/enzymology , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Large Cell/enzymology , Carcinoma, Large Cell/mortality , Carcinoma, Large Cell/pathology , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/pathology , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Prognosis , Small Cell Lung Carcinoma/mortality , Small Cell Lung Carcinoma/pathology , Survival Rate , Young Adult
20.
Oncotarget ; 6(11): 8539-51, 2015 Apr 20.
Article in English | MEDLINE | ID: mdl-25890501

ABSTRACT

Colorectal cancer is a major health problem and the second cause of cancer related death in western countries. Signaling pathways that control tissue homeostasis are often deregulated during tumorigenesis and contribute to tumor development. Studies in mouse models have shown that the p38 MAPK pathway regulates homeostasis in colon epithelial cells but also plays an important role in colon tumor maintenance. In this study, we have investigated the role of p38 MAPK signaling in patient-derived xenografts (PDXs) from three different human colon tumors representing clinical heterogeneity and that recapitulate the human tumor conditions both at histological and molecular levels. We have found that PH797804, a chemical inhibitor of p38 MAPK, reduces tumor growth of the three PDXs, which correlates with impaired colon tumor cell proliferation and survival. The inhibition of p38 MAPK in PDXs results in downregulation of the IL-6/STAT3 signaling pathway, which is a key regulator of colon tumorigenesis. Our results show the importance of p38 MAPK in human colon tumor growth using a preclinical model, and support that inhibition of p38 MAPK signaling may have therapeutic interest for colon cancer treatment.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Carcinoma, Neuroendocrine/pathology , Colonic Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Neoplasm Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/therapeutic use , Pyridones/therapeutic use , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Adenocarcinoma/drug therapy , Adenocarcinoma/enzymology , Adenocarcinoma/secondary , Animals , Carcinoma, Neuroendocrine/drug therapy , Carcinoma, Neuroendocrine/enzymology , Carcinoma, Neuroendocrine/genetics , Cell Differentiation , Colonic Neoplasms/drug therapy , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Cytokines/metabolism , Genes, ras , Humans , Liver Neoplasms/enzymology , Liver Neoplasms/secondary , MAP Kinase Signaling System/physiology , Mice , Mice, Nude , Mutation , Neoplasm Proteins/physiology , Neoplasm Staging , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/physiology
SELECTION OF CITATIONS
SEARCH DETAIL