Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cancer Sci ; 2024 May 27.
Article in English | MEDLINE | ID: mdl-38801833

ABSTRACT

Lung cancer is the leading cause of cancer death and includes two major types: non-small-cell lung cancer (NSCLC) and small-cell lung cancer (SCLC), accounting for 85% and 15% of cases, respectively. Non-small-cell lung cancer harboring actionable driver mutations is generally treated with tyrosine kinase inhibitors (TKIs) molecularly targeting individual oncogenes. Although TKIs have greatly contributed to better clinical outcomes, acquired resistance to them inevitably occurs. Histologic or lineage transformation is a rare but well-documented off-target mechanism associated with acquired resistance, and has been identified in settings following treatment with multiple different TKIs and other drugs. It includes neuroendocrine transformation, squamous cell transformation, and epithelial-to-mesenchymal transition. Here, we review the clinicopathologic features of transformed tumors and current understanding of the key genetic alterations and biologic mechanism of lineage transformation in NSCLC, particularly TKI-triggered transformation.

2.
Pathol Int ; 72(9): 457-463, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35801418

ABSTRACT

Angiosarcoma is a rare malignant tumor derived from vascular endothelial cells and has a poor prognosis. We have experienced a case of multiple breast angiosarcoma for which multiple resections had been performed during the course of its progression over a period of more than 15 years, allowing comprehensive genetic mutation analysis. Somatic mutations in several cancer-related genes were detected, but no previously reported driver gene mutations of angiosarcoma were evident. Several germline mutations associated with malignancy, such as single nucleotide polymorphisms in Fibroblast Growth Factor Receptor 4 (FGFR4) (p.Gly388Arg, rs351855), Kinase Insert Domain Receptor (KDR) (Gln472His, rs1870377) and tumor protein p53 (TP53) (p.Pro72Arg, rs1042522) were detected. Common signatures and genetic mutations were scarce in the tumor samples subjected to genetic mutational analysis. These findings suggested that this case was very probably a multiprimary angiosarcoma.


Subject(s)
Hemangiosarcoma , Breast Neoplasms , Endothelial Cells/pathology , Hemangiosarcoma/genetics , Hemangiosarcoma/pathology , Humans , Mutation , Receptor, Fibroblast Growth Factor, Type 4/genetics , Tumor Suppressor Protein p53/genetics , Vascular Endothelial Growth Factor Receptor-2/genetics
3.
Lab Invest ; 101(5): 543-553, 2021 05.
Article in English | MEDLINE | ID: mdl-33495573

ABSTRACT

Patient-derived xenograft (PDX) murine models are employed for preclinical research on cancers, including non-small cell lung cancers (NSCLCs). Even though lung squamous cell carcinomas (LUSCs) show the highest engraftment rate among NSCLCs, half of them nevertheless show PDX failure in immunodeficient mice. Here, using immunohistochemistry and RNA sequencing, we evaluated the distinct immunohistochemical and gene expression profiles of resected LUSCs that showed successful engraftment. Among various LUSCs, including the basal, classical, secretory, and primitive subtypes, those in the non-engrafting (NEG) group showed gene expression profiles similar to the pure secretory subtype with positivity for CK7, whereas those in the engrafting (EG) group were similar to the mixed secretory subtype with positivity for p63. Pathway analysis of 295 genes that demonstrated significant differences in expression between NEG and EG tumors revealed that the former had enriched expression of genes related to the immune system, whereas the latter had enriched expression of genes related to the cell cycle and DNA replication. Interestingly, NEG tumors showed higher infiltration of B cells (CD19+) and follicular dendritic cells (CD23+) in lymph follicles than EG tumors. Taken together, these findings suggest that the PDX cancer model of LUSC represents only a certain population of LUSCs and that CD19- and CD23-positive tumor-infiltrating immune cells in the original tumors may negatively influence PDX engraftment in immunodeficient mice.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Lung Neoplasms/metabolism , Neoplasm Transplantation , Animals , Antigens, CD19/metabolism , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/pathology , Gene Expression Profiling , Lung/pathology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Mice, SCID , Neoplasms, Experimental , Receptors, IgE/metabolism
4.
Cancer Sci ; 112(12): 5114-5126, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34628698

ABSTRACT

Ovarian carcinoma immunoreactive antigen domain 2 (OCIAD2) has been reported to show significantly higher expression in invasive lung adenocarcinoma than in lung adenocarcinoma in situ, and its abnormal expression is associated with poorer prognosis of the patients. However, the cellular function of OCIAD2 in this tumor remains poorly understood. In the present study, we first validated that OCIAD2 showed higher expression in human lung adenocarcinoma tissues or cell lines than in normal lung tissue or immortalized normal bronchial epithelial cells. OCIAD2 was localized predominantly at the mitochondrial membrane in lung adenocarcinoma cells. Interestingly, suppression of OCIAD2 led to loss of mitochondrial structure and a reduction in the number of mitochondria. Moreover, OCIAD2 suppression led to downregulation of cellular growth, proliferation, migration, and invasion, and upregulation of mitochondria-related apoptosis. We also showed that OCIAD2 suppression induced a decrease in mitochondrial membrane potential and release of cytochrome c. Transcriptional profiling using RNA sequencing revealed a total of 137 genes whose expression was commonly altered after OCIAD2 knockdown in three lung adenocarcinoma cell lines (A549, HCC827, and PC9). Pathway enrichment analysis of those genes demonstrated significant enrichment in apoptotic signaling or endoplasmic reticulum (ER) stress pathways. Our data suggest that OCIAD2 inhibits the mitochondria-initiated apoptosis and thus promotes the survival of lung cancer cells. Therefore, OCIAD2 may be an effective target for treatment of lung adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Apoptosis/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Mitochondria/genetics , Neoplasm Proteins/genetics , A549 Cells , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Blotting, Western , Cell Line, Tumor , Gene Ontology , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Membrane Potential, Mitochondrial/genetics , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/ultrastructure , Neoplasm Proteins/metabolism , RNA-Seq/methods , Reverse Transcriptase Polymerase Chain Reaction
5.
Pathol Int ; 71(10): 655-665, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34324245

ABSTRACT

Lung cancer is the most common cause of global cancer-related mortality, and the main histological type is adenocarcinoma, accounting for 50% of non-small cell lung cancer. In 2015, the World Health Organization (WHO) histological classification defined the concepts of "adenocarcinoma in situ" (AIS) and "minimally invasive adenocarcinoma" (MIA), which are considered to be adenocarcinomas at a very early stage. Although AIS and MIA have a very favorable outcome, once they progress to early but invasive adenocarcinoma (eIA), they can sometimes have a fatal outcome. We previously compared the expression profiles of eIA and AIS, and identified stratifin (SFN; 14-3-3 sigma) as a protein showing significantly higher expression in eIA than in AIS. Expression of SFN is controlled epigenetically by DNA demethylation, and its overexpression is significantly correlated with poorer outcome. In vitro and in vivo analyses have shown that SFN facilitates early progression of adenocarcinoma by enhancing cell proliferation. This review summarizes genetic and epigenetic abnormalities that can occur in early-stage lung adenocarcinoma and introduces recent findings regarding the biological significance of SFN overexpression during the course of lung adenocarcinoma progression. Therapeutic strategies for targeting SFN are also discussed.


Subject(s)
14-3-3 Proteins/genetics , Adenocarcinoma of Lung/genetics , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Exoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , 14-3-3 Proteins/metabolism , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/genetics , Disease Progression , Epigenesis, Genetic , Exoribonucleases/metabolism , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Lung Neoplasms/pathology
6.
Cancer Sci ; 110(8): 2431-2441, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31144406

ABSTRACT

The landscape of genetic alterations in disease models such as transgenic mice or mice with carcinogen-induced tumors has provided a huge amount of information that has shed light on the process of tumorigenesis in human non-small-cell lung cancer (NSCLC). We have previously identified stratifin (SFN) as a potent oncogene, and generated SFN-transgenic (Tg-SPC-SFN+/- ) mice, which express human SFN (hSFN) only in the lung. Here, we have found that carcinogen nicotine-derived nitrosaminoketone (NNK)-induced tumors developing in Tg-SPC-SFN+/- mice show a similar histology to human lung adenocarcinoma and exhibit high hSFN expression. In order to compare the genetic characteristics of Tg-SPC-SFN+/- tumors and human lung adenocarcinoma, the former were subjected to whole-exome sequencing. Interestingly, Tg-SPC-SFN+/- tumors showed the distinct distribution of exonic mutations and high number of mutated genes and transversion. Moreover, Tg-SPC-SFN+/- tumors showed 73 genes that were commonly detected in more than 2 tumors, mutations of which were also found in human lung adenocarcinoma. The expression levels of some of these genes were significantly associated with the clinical outcome of lung adenocarcinoma patients. Additionally, mutated genes in Tg-SPC-SFN+/- tumors were closely associated with key canonical pathways such as PI3K/AKT signaling and apoptosis signaling. These results suggest that SFN overexpression is a universal abnormality in human lung adenocarcinogenesis and Tg-SPC-SFN+/- tumors recapitulate key features of major human lung adenocarcinoma. Therefore, Tg-SPC-SFN+/- mice provide a useful model for clarifying the molecular mechanism underlying lung adenocarcinogenesis.


Subject(s)
14-3-3 Proteins/genetics , Adenocarcinoma of Lung/chemically induced , Adenocarcinoma of Lung/genetics , Biomarkers, Tumor/genetics , Carcinogens/pharmacology , Exoribonucleases/genetics , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Mutation/genetics , A549 Cells , Animals , Apoptosis/genetics , Carcinoma, Non-Small-Cell Lung/chemically induced , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mice , Mice, Inbred ICR , Mice, Transgenic , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics , Exome Sequencing/methods
7.
Pathol Int ; 68(11): 596-604, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30320419

ABSTRACT

The clinicopathological implications of ovarian cancer immunoreactive antigen domain containing 2 (OCIAD2) in lung adenocarcinoma were investigated. The expression of OCIAD2 in 191 surgically resected lung adenocarcinomas was examined using immunohistochemistry. OCIAD2 expression was quantified using the H-score and dichotomized as high or low. High OCIAD2 protein expression was significantly correlated with vascular invasion (P = 0.0018), lymphatic permeation (P = 0.049), T factor (P = 0.0024), and pathological stage (P = 0.0003). High OCIAD2 expression was significantly associated with poorer overall survival (OS) (n = 191, P = 0.0325). In peripheral-type lung adenocarcinomas (n = 161), high OCIAD2 expression was significantly associated with both poorer OS (P = 0.0214) and poorer disease-free survival (P = 0.0496). Adenocarcinoma in situ (AIS) and minimally invasive adenocarcinoma (MIA) showed weaker OCIAD2 expression than invasive adenocarcinoma. Among small adenocarcinomas measuring 2 cm or less in greatest dimension classified according to the Noguchi's classification (n = 79), invasive adenocarcinomas showed significantly higher OCIAD2 expression than non-invasive adenocarcinomas (P = 0.0007). Interestingly, OCIAD2 was expressed heterogeneously even within a tumor, and its expression was higher in areas of invasion than in areas of in situ spread. Our results suggest that OCIAD2 could be a useful prognostic biomarker of lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung/pathology , Biomarkers, Tumor/analysis , Neoplasm Proteins/biosynthesis , Adenocarcinoma of Lung/mortality , Aged , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Proteins/analysis , Prognosis , Proportional Hazards Models
8.
Cancer Sci ; 108(3): 536-542, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28012229

ABSTRACT

Immunoglobulin (CD79a) binding protein 1 (IGBP1) is universally overexpressed in lung adenocarcinoma and exerts an anti-apoptotic effect by binding to PP2Ac. However, the molecular mechanism of IGBP1 overexpression is still unclear. In the present study, we used a microRNA (miRNA) array and TargetScan Human software to detect IGBP1-related miRNAs that regulate IGBP1 expression. The miRNA array analysis revealed more than 100 miRNAs that are dysregulated in early invasive adenocarcinoma. On the other hand, in silico analysis using TargetScan Human revealed 79 miRNAs that are associated with IGBP1 protein expression. Among the miRNAs selected by miRNA array analysis, six (miR-34b, miR-138, miR-374a, miR-374b, miR-1909, miR-3941) were also included among those selected by TargetScan analysis. Real-time reverse transcription PCR (real-time RT-PCR) showed that the six microRNAs were downregulated in invasive adenocarcinoma (IGBP1+) relative to adjacent normal lung tissue (IGBP1-). Among these microRNAs, only miR-34b and miR-3941 depressed luciferase activity by targeting 3'UTR-IGBP1 in the luciferase vector. We transfected miR-34b and miR-3941 into lung adenocarcinoma cell lines (A549, PC-9), and both of them suppressed IGBP1 expression and cell proliferation. Moreover, the transfected miR-34b and miR-3941 induced apoptosis of a lung adenocarcinoma cell line, similarly to the effect of siIGBP1 RNA. As well as miR-34b, we found that miR-3941 targeted IGBP1 specifically and was able to exclusively downregulate IGBP1 expression. These findings indicate that suppression of miR-3941 has an important role in the progression of lung adenocarcinoma at an early stage.


Subject(s)
Adenocarcinoma/pathology , Apoptosis/genetics , Gene Expression Regulation, Neoplastic/genetics , Intracellular Signaling Peptides and Proteins/biosynthesis , Lung Neoplasms/pathology , MicroRNAs/genetics , A549 Cells , Adaptor Proteins, Signal Transducing , Adenocarcinoma/genetics , Adenocarcinoma of Lung , Cell Line, Tumor , Cell Proliferation/genetics , Disease Progression , Humans , Intracellular Signaling Peptides and Proteins/genetics , Lung Neoplasms/genetics , Molecular Chaperones , Real-Time Polymerase Chain Reaction , Transfection
9.
Pathol Int ; 67(11): 555-563, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29027312

ABSTRACT

Cyclophilin A (CypA) has been reported to be upregulated in malignant tumors. CypA expression is thought to be associated with acquisition of tumor growth and anti-apoptotic function. Although upregulation of CypA has been reported in lung adenocarcinoma, its clinicopathological significance and roles in malignant progression remain unclear. Here we investigated the implications of CypA expression for outcome in patients with lung adenocarcinoma. Lung adenocarcinoma specimens from 198 cases were selected and reclassified according to the World Health Organization classification (4th edition) and the Noguchi classification. CypA expression was assessed by immunohistochemistry, and the H-score was calculated on the basis of intensity and proportion. The specificity of the antibody used was confirmed by Western blotting and the cut-off point was determined from the ROC curve. Sixty-seven cases (33.8%) had low CypA expression (CypA-L group) and 131 (66.2%) had high CypA expression (CypA-H group). Many cases of adenocarcinoma in situ were CypA-L, and advanced adenocarcinomas tended to be classified as CypA-H. Clinically, patients with CypA-H tumors showed a significantly poorer prognosis than those with CypA-L tumors. This is the first investigation of the implications of the CypA expression level in terms of the clinical characteristics of resected lung adenocarcinomas.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/analysis , Cyclophilin A/biosynthesis , Lung Neoplasms/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adenocarcinoma of Lung , Adult , Aged , Cyclophilin A/analysis , Female , Humans , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Middle Aged , Prognosis
10.
Pathol Int ; 67(6): 292-301, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28544031

ABSTRACT

Alterations of epidermal growth factor receptor (EGFR) expression frequently occur in early-stage lung adenocarcinoma. Ubiquitin-specific protease 8 (USP8) has been reported to stabilize EGFR protein at the plasma membrane through the recycling pathway. Here, we examined the correlation between USP8 expression and the expression or mutation status of EGFR, as well as the clinicopathological features of lung adenocarcinoma and patient outcome. Expression of EGFR and USP8 in surgically resected specimens of lung adenocarcinoma (82 cases) was examined by immunohistochemistry. Overexpression of EGFR was mutually correlated with that of USP8, and was also associated with clinicopathological features including pathological subtype, lymphatic permeation, and vascular invasion. Moreover, patients who had USP8-positive tumors had a significantly poorer outcome than those who were USP8-negative, not only overall but also patients who were EGFR-negative. Although EGFR was expressed in invasive adenocarcinoma but not in adenocarcinoma in situ (AIS), USP8 was overexpressed in not only invasive adenocarcinoma but also 38.1% of AIS cases. In vitro, USP8 regulated the expression and half-life of EGFR in immortalized AIS cells, and also cell proliferation. Our findings indicate that overexpression of USP8 in lung adenocarcinoma is an early event during the course of tumor progression, and is related to EGFR expression.


Subject(s)
Adenocarcinoma/diagnosis , Biomarkers, Tumor/metabolism , Endopeptidases/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Lung Neoplasms/diagnosis , Ubiquitin Thiolesterase/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Biomarkers, Tumor/genetics , Cell Line , Disease-Free Survival , Early Diagnosis , Endopeptidases/genetics , Endosomal Sorting Complexes Required for Transport/genetics , ErbB Receptors/genetics , Female , Humans , Immunohistochemistry , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Prognosis , Ubiquitin Thiolesterase/genetics
11.
Mol Cancer ; 14: 142, 2015 Jul 30.
Article in English | MEDLINE | ID: mdl-26223682

ABSTRACT

BACKGROUNDS: Adenocarcinoma in situ (AIS) of the lung has an extremely favorable prognosis. However, early but invasive adenocarcinoma (eIA) sometimes has a fatal outcome. We had previously compared the expression profiles of AIS with those of eIA showing lymph node metastasis or a fatal outcome, and found that stratifin (SFN, 14-3-3 sigma) was a differentially expressed gene related to cell proliferation. Here, we performed an in vivo study to clarify the role of SFN in initiation and progression of lung adenocarcinoma. FINDINGS: Suppression of SFN expression in A549 (a human lung adenocarcinoma cell line) by siSFN significantly reduced cell proliferation activity and the S-phase subpopulation. In vivo, tumor development or metastasis to the lung was reduced in shSFN-transfected A549 cells. Moreover, we generated SFN-transgenic mice (Tg-SPC-SFN(+/-)) showing lung-specific expression of human SFN under the control of a tissue-specific enhancer, the SPC promoter. We found that Tg-SPC-SFN(+/-) mice developed lung tumors at a significantly higher rate than control mice after administration of chemical carcinogen, NNK. Interestingly, several Tg-SPC-SFN(+/-) mice developed tumors without NNK. These tumor cells showed high hSFN expression. CONCLUSION: These results suggest that SFN facilitates lung tumor development and progression. SFN appears to be a novel oncogene with potential as a therapeutic target.


Subject(s)
14-3-3 Proteins/genetics , Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , Cell Proliferation/genetics , Exoribonucleases/genetics , Lung Neoplasms/genetics , 14-3-3 Proteins/biosynthesis , Adenocarcinoma/chemically induced , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Biomarkers, Tumor/biosynthesis , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Exoribonucleases/biosynthesis , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mice , Mice, Transgenic , Neoplasm Metastasis , Neoplasm Staging , Nitrosamines/toxicity , Xenograft Model Antitumor Assays
12.
Pathol Int ; 64(3): 115-22, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24698421

ABSTRACT

Adenomyosis is characterized by extension of endometrial glands and stromal cells into the myometrium. Here we proved that 'moesin' is a unique biomarker of adenomyosis. We selected two cases of adenomyosis that had been surgically resected and fixed with formalin. Proteins were extracted from the infiltrating adenomyosis lesions and normal endometrium by tissue microdissection. The extracted proteins were examined using a LC-MS/MS system and the expression profiles of each region were compared. Two hundred and sixty proteins were detected, among which 73 were expressed more in adenomyosis than in normal endometrium. Among these proteins, we focused on overexpression of moesin in adenomyosis. Expression of moesin estimated semiquantitatively using an immunohistochemistry score was higher in adenomyosis than in normal endometrium. In particular, moesin was significanly overexpressed in stromal cells of adenomyosis than in those of normal endometrium. Relative to normal endometrium, moesin was also overexpressed at the RNA level in 9 of 14 cases of adenomyosis and at the protein level in all 14 cases. We also detected activated (phosphorylated) moesin in adenomyosis lesions. The present findings suggest that moesin is characteristically overexpressed and activated in adenomyosis, and that moesin activation may be related to extension of adenomyosis in the myometrium.


Subject(s)
Adenomyosis/diagnosis , Endometrium/metabolism , Microfilament Proteins/metabolism , Myometrium/metabolism , Adenomyosis/metabolism , Adenomyosis/pathology , Biomarkers/metabolism , Endometrium/pathology , Female , Humans , Myometrium/pathology , Phosphorylation
13.
Virchows Arch ; 2024 May 06.
Article in English | MEDLINE | ID: mdl-38710944

ABSTRACT

INTRODUCTION: HNF4α expression and SMARCA4 loss were thought to be features of non-terminal respiratory unit (TRU)-type lung adenocarcinomas, but their relationships remained unclear. MATERIALS AND METHODS: HNF4α-positive cases among 241 lung adenocarcinomas were stratified based on TTF-1 and SMARCA4 expressions, histological subtypes, and driver mutations. Immunohistochemical analysis was performed using xenograft tumors of lung adenocarcinoma cell lines with high HNF4A expression. RESULT: HNF4α-positive adenocarcinomas(n = 33) were divided into two groups: the variant group(15 mucinous, 2 enteric, and 1 colloid), where SMARCA4 was retained in all cases, and the conventional non-mucinous group(6 papillary, 5 solid, and 4 acinar), where SMARCA4 was lost in 3/15 cases(20%). All variant cases were negative for TTF-1 and showed wild-type EGFR and frequent KRAS mutations(10/18, 56%). The non-mucinous group was further divided into two groups: TRU-type(n = 7), which was positive for TTF-1 and showed predominantly papillary histology(6/7, 86%) and EGFR mutations(3/7, 43%), and non-TRU-type(n = 8), which was negative for TTF-1, showed frequent loss of SMARCA4(2/8, 25%) and predominantly solid histology(4/8, 50%), and never harbored EGFR mutations. Survival analysis of 230 cases based on histological grading and HNF4α expression revealed that HNF4α-positive poorly differentiated (grade 3) adenocarcinoma showed the worst prognosis. Among 39 cell lines, A549 showed the highest level of HNF4A, immunohistochemically HNF4α expression positive and SMARCA4 lost, and exhibited non-mucinous, high-grade morphology in xenograft tumors. CONCLUSION: HNF4α-positive non-mucinous adenocarcinomas included TRU-type and non-TRU-type cases; the latter tended to exhibit the high-grade phenotype with frequent loss of SMARCA4, and A549 was a representative cell line.

14.
Biochim Biophys Acta Mol Basis Dis ; 1870(6): 167249, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38768929

ABSTRACT

RET fusion is an oncogenic driver in 1-2 % of patients with non-small cell lung cancer (NSCLC). Although RET-positive tumors have been treated with multikinase inhibitors such as vandetanib or RET-selective inhibitors, ultimately resistance to them develops. Here we established vandetanib resistance (VR) clones from LC-2/ad cells harboring CCDC6-RET fusion and explored the molecular mechanism of the resistance. Each VR clone had a distinct phenotype, implying they had acquired resistance via different mechanisms. Consistently, whole exome-seq and RNA-seq revealed that the VR clones had unique mutational signatures and expression profiles, and shared only a few common remarkable events. AXL and IGF-1R were activated as bypass pathway in different VR clones, and sensitive to a combination of RET and AXL inhibitors or IGF-1R inhibitors, respectively. SMARCA4 loss was also found in a particular VR clone and 55 % of post-TKI lung tumor tissues, being correlated with higher sensitivity to SMARCA4/SMARCA2 dual inhibition and shorter PFS after subsequent treatments. Finally, we detected an increased number of damaged mitochondria in one VR clone, which conferred sensitivity to mitochondrial electron transfer chain inhibitors. Increased mitochondria were also observed in post-TKI biopsy specimens in 13/20 cases of NSCLC, suggesting a potential strategy targeting mitochondria to treat resistant tumors. Our data propose new promising therapeutic options to combat resistance to RET inhibitors in NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Drug Resistance, Neoplasm , Lung Neoplasms , Mitochondria , Piperidines , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-ret , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Proto-Oncogene Proteins c-ret/antagonists & inhibitors , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins c-ret/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Piperidines/pharmacology , Piperidines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Protein Kinase Inhibitors/pharmacology , Cell Line, Tumor , Quinazolines/pharmacology , Quinazolines/therapeutic use , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/antagonists & inhibitors , Signal Transduction/drug effects , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/antagonists & inhibitors , DNA Helicases/genetics , DNA Helicases/metabolism , DNA Helicases/antagonists & inhibitors , Cytoskeletal Proteins
15.
Am J Pathol ; 180(4): 1653-62, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22310466

ABSTRACT

We previously have shown the aberrant overexpression of stratifin (SFN, 14-3-3 ς) in lung adenocarcinoma. Although SFN is known to facilitate tumor cell proliferation, the mechanism that underlies its aberrant expression has remained unclear. SFN, the downstream target of p53, often has been reported to be hypermethylated and subsequently silenced in certain cancers; however, its hypomethylation-linked reactivation has not yet been validated. In this study, we investigated the DNA methylation status of the SFN promoter region using 8 lung cancer cell lines and 32 specimens of adenocarcinoma tissue. Real-time methylation-specific PCR analysis showed that although both normal lung tissue and adenocarcinoma in situ bore a completely methylated SFN promoter, the promoter region in almost all invasive adenocarcinomas was at least partially methylated. The expression of SFN and its level of methylation were correlated strongly. Furthermore, statistical analysis revealed that the level of methylation became reduced with progression of the pathologic stage, although no clear relationship between methylation level and p53 abnormality was found. These results suggest that methylation-related silencing of SFN occurs in both normal lung tissues and adenocarcinoma in situ, and that demethylation of the SFN promoter participates in the aberrant expression of SFN in invasive adenocarcinoma cells, independently of p53 alteration. This novel finding might be informative for clarifying the mechanism that underlies the progression of early lung adenocarcinoma.


Subject(s)
14-3-3 Proteins/genetics , Adenocarcinoma/genetics , Biomarkers, Tumor/genetics , CpG Islands/genetics , DNA Methylation , Exonucleases/genetics , Lung Neoplasms/genetics , 14-3-3 Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Base Sequence , Biomarkers, Tumor/metabolism , DNA, Neoplasm/genetics , Exonucleases/metabolism , Exoribonucleases , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Lung/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Staging , Promoter Regions, Genetic/genetics , Real-Time Polymerase Chain Reaction/methods , Tumor Cells, Cultured
16.
Nat Cancer ; 3(6): 710-722, 2022 06.
Article in English | MEDLINE | ID: mdl-35726063

ABSTRACT

Lorlatinib is currently the most advanced, potent and selective anaplastic lymphoma kinase (ALK) tyrosine kinase inhibitor for the treatment of ALK-positive non-small cell lung cancer in the clinic; however, diverse compound ALK mutations driving therapy resistance emerge. Here, we determine the spectrum of lorlatinib-resistant compound ALK mutations in patients, following treatment with lorlatinib, the majority of which involve ALK G1202R or I1171N/S/T. We further identify structurally diverse lorlatinib analogs that harbor differential selective profiles against G1202R versus I1171N/S/T compound ALK mutations. Structural analysis revealed increased potency against compound mutations through improved inhibition of either G1202R or I1171N/S/T mutant kinases. Overall, we propose a classification of heterogenous ALK compound mutations enabling the development of distinct therapeutic strategies for precision targeting following sequential tyrosine kinase inhibitors.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Aminopyridines , Anaplastic Lymphoma Kinase/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Drug Resistance, Neoplasm/genetics , Humans , Lactams , Lactams, Macrocyclic/pharmacology , Lung Neoplasms/drug therapy , Mutation , Protein Kinase Inhibitors/pharmacology , Pyrazoles
17.
Int J Cancer ; 129(10): 2445-53, 2011 Nov 15.
Article in English | MEDLINE | ID: mdl-21207417

ABSTRACT

Adenocarcinoma in situ (AIS) of the lung has an extremely favorable prognosis, with a 5-year survival rate of 100%. However, early invasive adenocarcinoma (EIA) often has a fatal outcome. In this study, we compared the expression profiles of AIS with those of EIA showing lymph node metastasis or a fatal outcome, and screened the differentially expressed genes by cDNA microarray. From the genes selected, we focused on Stratifin (SFN, 14-3-3 σ), which showed significantly higher expression in EIA than in AIS. Immunohistochemistry for SFN revealed that more than 95% of EIAs were immunopositive for SFN, in comparison to only 13% of AISs (p <0.05). Interestingly, positivity was detected not only in the invasive region but also in the in situ spreading component of EIA. Functionally, SFN facilitates the cell proliferation capacity of lung adenocarcinoma. These results indicate that SFN overexpression is a universal abnormality during the stepwise progression from in situ to invasive adenocarcinoma of the lung.


Subject(s)
14-3-3 Proteins/metabolism , Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , Exonucleases/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Disease Progression , Exoribonucleases , Lung Neoplasms/pathology , Lymphatic Metastasis , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation
18.
Histopathology ; 58(5): 729-38, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21457161

ABSTRACT

AIMS: To investigate the specific expression of insulin-like growth factor binding protein-1 (IGFBP-1) in ovarian clear cell adenocarcinoma (CCA). METHODS AND RESULTS: Immunohistochemistry and in situ hybridization for IGFBP-1 were performed in normal endometrium, placenta, and 100 surgically resected cases of ovarian cancer including 31 CCAs and 69 non-CCAs. Immunohistochemistry for hepatocyte nuclear factor-1 (HNF-1ß) was also examined in all cases. Specific expression of IGFBP-1 was confirmed in secretory endometrium, decidua of placenta and Arias-Stella glands of miscarriage material. Among ovarian cancers, almost all cases of CCA showed expression of both IGFBP-1 protein and mRNA, but non-CCA hardly expressed IGFBP-1. There was a significant difference between CCA and non-CCA in the expression of IGFBP-1 protein and mRNA. No correlation was found between the rate of IGFBP-1 expression and pathological T and N factors of the tumour-node-metastasis (TNM) classification. All CCA cases except for one exhibited expression of HNF-1ß protein, whereas only 15.9% of non-CCAs did so. CONCLUSION: The expression of IGFBP-1 in CCA is more specific than that of HNF-1ß. IGFBP-1 shows expression by decidual endometrium and Arias-Stella glands, and CCA also exhibits characteristic expression. These results indicate that IGFBP-1 is a immunohistochemical marker for CCA.


Subject(s)
Adenocarcinoma, Clear Cell/metabolism , Insulin-Like Growth Factor Binding Protein 1/metabolism , Ovarian Neoplasms/metabolism , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/pathology , Adult , Female , Humans , Immunohistochemistry , Middle Aged , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , RNA, Messenger/metabolism
19.
Oncotarget ; 10(17): 1625-1636, 2019 Feb 26.
Article in English | MEDLINE | ID: mdl-30899432

ABSTRACT

Although alteration of DNA methylation in advanced cancer has been extensively investigated, few data for early-stage lung adenocarcinoma are available. Here, we compared DNA methylation profiles between adenocarcinoma in situ (AIS) and early invasive adenocarcinoma using the Infinium methylation array to investigate methylation abnormalities causing early progression of adenocarcinomas. We focused on differentially methylated sites which were located in promoter CpG islands or shore regions, and identified 579 hypermethylated sites and 23 hypomethylated sites in early invasive adenocarcinoma relative to AIS and normal lung. These hypermethylated genes were significantly associated with neuronal pathways such as the GABA receptor and serotonin signaling pathways. Among the hypomethylated genes, we found that GORASP2, ZYG11A, and SFN had significantly lower methylation rates at the shore regions and significantly higher protein expression in invasive adenocarcinoma. Moreover, overexpression of those proteins was strongly associated with patient's poor outcome. Despite DNA demethylation at the promoter region might be rare relative to DNA hypermethylation, we identified 2 new genes, GORASP2 and ZYG11A, which show hypomethylation and overexpression in invasive adenocarcinoma, suggesting that they have important functions in tumor cells. These genes may be clinically applicable as prognostic indicators and could be potential novel target molecules for drug development.

20.
Clin Cancer Res ; 25(9): 2809-2820, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30728155

ABSTRACT

PURPOSE: Aberrant overexpression of SFN (stratifin) plays an oncogenic role in lung adenocarcinoma. We have shown previously that SKP1, an adapter component of E3 ubiquitin ligase forming an SCF complex, is a unique SFN-binding protein in lung adenocarcinoma cells. EXPERIMENTAL DESIGN: In silico simulation and in vitro mutagenesis analysis were performed to identify the SFN-binding domain on SKP1. We examined expression, localization, and stability of SKP1 after knockdown of SFN using lung adenocarcinoma cells including A549. In silico library screening and experimental validation were used for drug screening. Daily oral administration of each candidate drugs to A549-injected tumor-bearing mice was performed to evaluate their in vivo antitumor efficacy. RESULTS: Suppression of SFN upregulated the stability of SKP1 and accelerated its cytoplasm-to-nucleus translocation. Consistently, IHC analysis revealed that cytoplasmic expression of SKP1 was significantly associated with SFN positivity, tumor malignancy, and poorer patient outcome. After SFN suppression, ubiquitination of oncoproteins, including p-cyclin E1, p-c-Myc, p-c-Jun, and cleaved Notch 1, which are target proteins of SCFFBW7, was strongly induced. These results indicate that SFN-SKP1 binding results in SCFFBW7 dysfunction and allows several oncoproteins to evade ubiquitination and subsequent degradation. Because inhibition of SFN-SKP1 binding was expected to have antitumor efficacy, we next searched for candidate SFN inhibitors. Aprepitant and ticagrelor were finally selected as potential SFN inhibitors that dose dependently reduced SFN-SKP1 binding and tumor progression in vivo. CONCLUSIONS: As overexpression of SFN is detectable in most adenocarcinoma, we believe that SFN inhibitors would be novel and promising antitumor drugs for lung adenocarcinoma.


Subject(s)
14-3-3 Proteins/metabolism , Adenocarcinoma of Lung/pathology , Biomarkers, Tumor/metabolism , Exoribonucleases/metabolism , Lung Neoplasms/pathology , Oncogene Proteins/metabolism , S-Phase Kinase-Associated Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin/metabolism , 14-3-3 Proteins/genetics , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Exoribonucleases/genetics , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Oncogene Proteins/genetics , Prognosis , Protein Stability , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/genetics , SKP Cullin F-Box Protein Ligases/genetics , Survival Rate , Tumor Cells, Cultured , Ubiquitination , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL