首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 15 毫秒
1.
Spontaneous regression of malignant tumors is rare especially of lung tumor and biological mechanism of such remission has not been addressed. We report the case of a 79-year-old Korean patient with non-small cell lung cancer, squamous cell cancer with a right hilar tumor and multiple lymph nodes, lung to lung metastasis that spontaneously regressed without any therapies. He has sustained partial remission state for one year and eight months after the first histological diagnosis.  相似文献   

2.
3.
4.
Deciphering the genomic profiles and tumor microenvironment (TME) in large cell carcinomas of the lung (LCC), large cell neuroendocrine of the lung (LCNEC), and small cell lung cancer (SCLC) might contribute to a better understanding of lung cancer and then improve outcomes. Ten LCC patients, 12 LCNEC patients, and 18 SCLC patients were enrolled. Targeted next-generation sequencing was used to investigate the genomic profiles of LCC, LCNEC, and SCLC. Tumor-infiltrating lymphocytes (TILs) within cancer cell nests and in cancer stroma were counted separately. Precise 60% of LCNEC patients harbored classical non-small cell lung cancer driver alterations, occurring in BRAF, KRAS, ROS1, and RET. More than 70% of SCLC patients harbored TP53-RB1 co-alterations. Moreover, 88.9%, 40%, and 77.8% of LCC, LCNEC, and SCLC cases had a high tumor mutation burden level with more than 7 mutations/Mb. Furthermore, high index of CD68+ CD163+ (TILs within cancer cell nests/ TILs within cancer cell nests and in cancer stroma, P = .041, 548 days vs not reached) and CD163+ TILs (P = .041, 548 days vs not reached) predicted a shorter OS in SCLC. Our findings revealed the distinct genomic profiles and TME contexture among LCC, LCNEC, and SCLC. Our findings suggest that stratifying LCNEC/SCLC patients based on TME contexture might help clinical disease management.  相似文献   

5.
The development of angioimmunoblastic lymphadenopathy in a patient with a slowly growing squamous cell carcinoma of the lung is reported. The possible relation between the two concomitant conditions in this rare case is proposed.  相似文献   

6.
Notch1信号通路是许多细胞信号转导通路的交汇点,不仅在正常组织、细胞的分化、发育过程中起重要作用,而且与一些肿瘤的发生、发展相关,如食管癌、胃癌、宫颈癌、结肠癌、肺癌等.Notch1在非小细胞肺癌中的作用多样.了解Notch1和非小细胞肺癌的关系有利于进一步阐明非小细胞肺癌的发生机制,为非小细胞肺癌的治疗提供一个新的有希望的治疗靶点.  相似文献   

7.
8.
9.
Signaling through the store-operated Ca2+ release-activated Ca2+ (CRAC) channel regulates critical cellular functions, including gene expression, cell growth and differentiation, and Ca2+ homeostasis. Loss-of-function mutations in the CRAC channel pore-forming protein ORAI1 or the Ca2+ sensing protein stromal interaction molecule 1 (STIM1) result in severe immune dysfunction and nonprogressive myopathy. Here, we identify gain-of-function mutations in the cytoplasmic domain of STIM1 (p.R304W) associated with thrombocytopenia, bleeding diathesis, miosis, and tubular myopathy in patients with Stormorken syndrome, and in ORAI1 (p.P245L), associated with a Stormorken-like syndrome of congenital miosis and tubular aggregate myopathy but without hematological abnormalities. Heterologous expression of STIM1 p.R304W results in constitutive activation of the CRAC channel in vitro, and spontaneous bleeding accompanied by reduced numbers of thrombocytes in zebrafish embryos, recapitulating key aspects of Stormorken syndrome. p.P245L in ORAI1 does not make a constitutively active CRAC channel, but suppresses the slow Ca2+-dependent inactivation of the CRAC channel, thus also functioning as a gain-of-function mutation. These data expand our understanding of the phenotypic spectrum of dysregulated CRAC channel signaling, advance our knowledge of the molecular function of the CRAC channel, and suggest new therapies aiming at attenuating store-operated Ca2+ entry in the treatment of patients with Stormorken syndrome and related pathologic conditions.Ca2+ influx in response to the depletion of intracellular Ca2+ stores, or store-operated Ca2+ entry, constitutes one of the major routes of Ca2+ entry in all animal cells (1). Under physiological conditions, Ca2+ influx is activated in response to numerous G protein-coupled receptors and receptor tyrosine kinases signaling via inositol-1,4,5-trisphosphate as a second messenger (2). Store-operated Ca2+ entry is mediated primarily by the Ca2+ release-activated Ca2+ (CRAC) channel (3), which consists of the pore-forming subunits ORAI1–3 (or CRAC modulators 1–3) and Ca2+ sensors, STIM1 and STIM2 (47). STIM proteins reside in the membrane of endoplasmic reticulum (ER), whereas ORAI proteins reside in the plasma membrane. STIM1 is a single transmembrane-spanning protein (812) that, in resting cells, exists as a dimer that binds Ca2+ through two EF hand-containing domains located in the ER lumen (13). Depletion of Ca2+ in the ER induces a series of molecular events in the conformation and localization of STIM1, initiated by the formation of higher-order oligomers, protein unfolding, and accumulation at discrete sites in the cell where the ER membrane is in close proximity to the plasma membrane (11, 1316). In these sites, STIM1 binds to the cytosolic C and N termini of ORAI1 (17, 18), resulting in channel activation and generation of a highly Ca2+-selective CRAC current, or ICRAC (3, 19, 20). ICRAC is responsible not only for restoring cytosolic and ER Ca2+ concentration, thus maintaining the cell in a Ca2+ signaling-competent stage (1), but also for many cellular functions such as regulation of gene expression, exocytosis, proliferation, and apoptosis (1).Consistent with a fundamental role of the CRAC channel in cell signaling, loss-of-function mutations in STIM1 or ORAI1 lead to immune deficiency and nonprogressive myopathy (2123). However, evidence that gain-of-function mutations in STIM1 and ORAI1 can affect human health is only recently starting to emerge. It was shown that mutations in the domain of STIM1 that binds Ca2+ (EF hand domain) in resting conditions are associated with nonsyndromic myopathy with tubular aggregates (24). Functional studies demonstrated that these mutations cause hyperactivation of the CRAC channel (24). However, it remains unknown whether myopathy with tubular aggregates is caused by the increased activity of the CRAC channel, increased activity of another Ca2+ channel using STIM1 as a sensor (25), or a function of STIM1 that is unrelated to Ca2+ signaling, as STIM1 can function independently of ORAI1 (26-28).Stormorken syndrome [Mendelian Inheritance in Man (MIM) 185070] is a rare autosomal-dominant condition with a constellation of symptoms, including congenital miosis, bleeding diathesis, thrombocytopenia, functional (or anatomical) asplenia, and proximal muscle weakness (29). Other manifestations include ichthyosis, headaches, and dyslexia (30). Patients typically display increased creatine kinase (CK) levels and histologic evidence of myopathy with tubular aggregates (30, 31). Here, we show that Stormorken syndrome is caused by an activating mutation in STIM1. We also identify a mutation in the STIM1-interacting molecule, ORAI1, in a Stormorken-like syndrome that presented with miosis and tubular myopathy. Functional analyses reveal that both mutations enhance the activity of the CRAC channel, but by different molecular mechanisms. These data expand the phenotypic spectrum of activating mutations in the CRAC channels from myopathy with tubular aggregates to miosis, bleeding diathesis, thrombocytopenia, asplenia, ichthyosis, headaches, and dyslexia.  相似文献   

10.
BackgroundTumor mutational burden (TMB) has been identified as one of the predictors for the response to anti-programmed cell death-1 (anti-PD-1) antibody therapy and reported to correlate with smoking history in lung adenocarcinoma. However, in squamous cell carcinoma of the lung, the association between TMB and clinicopathological background factors, such as smoking history, has not been reported, including in our previous study. The mutational signature is a tool to identify the mutagens that are contributing to the mutational spectrum of a tumor by investigating the pattern of DNA changes. Here, we analyzed the mutational signature in lung squamous cell carcinoma to identify mutagens affecting the TMB.MethodsSeven representative mutational signatures including signature 7 (SI7) [ultraviolet (UV)-related], SI4 (smoking), SI6/15 [mismatch repair (MMR)], SI2/13 [apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like (APOBEC)], and SI5 (clock-like) were analyzed in Japanese patients with lung squamous cell carcinoma (n=67) using data generated by next-generation sequencing consisting of a 415-gene panel. The relationships between signatures and clinico-pathological data including TMB and programmed death-ligand 1 (PD-L1) expression were analyzed.ResultsAlthough the reconstructed mutational counts were small with targeted sequencing (median: 30.1, range: 13.3–98.7), the distributions of signatures were comparable among samples, with 56 cases containing more than four signatures. The smoking-related SI4 was found in 45 cases and was significantly related with pack-year index (PYI) (P=0.026). The reconstructed mutation counts were highly correlated with SI4 (r=0.51, P<0.0001), whereas the correlation was weak with SI6/15 (MMR-related) and SI2/13 (APOBEC-related). There was no mutational signature related with PD-L1 expression. Some patients exhibited unique signatures; the patient with the highest mutational counts had a MMR signature, and another patient with a prominent UV signature had occupational exposure to UV, as he was employed as a neon sign engineer.ConclusionsMutational signatures can predict the cause of lung squamous cell carcinoma. Tobacco smoking is the mutagen most related with TMB.  相似文献   

11.
白澎  张沪生  王毓洲 《国际呼吸杂志》2007,27(23):1777-1781
目的利用基因表达谱芯片技术,寻找人类肺鳞癌组织和正常肺组织中与生物信号转导或免疫反应有关的差异表达基因。方法分别提取肺鳞癌和正常肺组织总RNA后,逆转录合成cDNA探针,混合后与Biostar H-Ⅰ型基因芯片杂交,ScanArray 4000扫描仪扫描杂交信号,计算机分析差异表达基因。结果筛选出在3组以上标本中存在差异表达的基因77条,其中44条表达上调(r〉2.0),33条表达下调(r〈0.5)。结论利用基因芯片技术实现了高通量筛选与肺癌发生发展密切相关的基因,发现了77条在肺鳞癌中与细胞信号转导或免疫反应有关的差异表达基因,为今后更深入的研究指明了方向。  相似文献   

12.
Lung cancer metastasis is typically determined by histologic similarity between distant and primary lesions. Herein, we present a 70-year-old Japanese woman with an adenocarcinoma in her lung and a squamous cell carcinoma in her femur; both tumors had an identical epidermal growth factor receptor mutation, G719S. This indicated that both tumors had a common origin, despite their histologic dissimilarity. The tumor in the femur was thus identified genetically as a lung cancer metastasis. This case suggests that genetic analysis can determine whether a distant lesion is a lung cancer metastasis, particularly when the histology differs from that of the primary lesion.  相似文献   

13.

Background

Immunohistochemical staining has been widely used in distinguishing lung adenocarcinoma (LUAD) from lung squamous cell carcinoma (LUSC), which is of vital importance for the diagnosis and treatment of lung cancer. Due to the lack of a comprehensive analysis of different lung cancer subtypes, there may still be undiscovered markers with higher diagnostic accuracy.

Methods

Herein first, we systematically analyzed high-throughput data obtained from The Cancer Genome Atlas (TCGA) database. Combining differently expressed gene screening and receiver operating characteristic (ROC) curve analysis, we attempted to identify the genes which might be suitable as immunohistochemical markers in distinguishing LUAD from LUSC. Then we detected the expression of six of these genes (MLPH, TMC5, SFTA3, DSG3, DSC3 and CALML3) in lung cancer sections using immunohistochemical staining.

Results

A number of genes were identified as candidate immunohistochemical markers with high sensitivity and specificity in distinguishing LUAD from LUSC. Then the staining results confirmed the potentials of the six genes (MLPH, TMC5, SFTA3, DSG3, DSC3 and CALML3) in distinguishing LUAD from LUSC, and their sensitivity and specificity were not less than many commonly used markers.

Conclusions

The results revealed that the six genes (MLPH, TMC5, SFTA3, DSG3, DSC3 and CALML3) might be suitable markers in distinguishing LUAD from LUSC, and also validated the feasibility of our methods for identification of candidate markers from high-throughput data.  相似文献   

14.
BackgroundFor metachronous second pulmonary squamous cell carcinoma (msPSC) in patients with resected PSC, the method to distinguish tumour clonality has not yet been well established, which makes it difficult to determine accurate staging and predict prognosis.MethodsPatients who underwent surgery for first PSC and encountered msPSC were recruited from the Surveillance, Epidemiology, and End Results (SEER) database. We extracted overall survival 1 (OS1) for the first PSC, overall survival 2 (OS2) for msPSC, and interval survival for the time interval between the first and second PSC. The nomogram was calibrated for OS2, and recursive partitioning analysis (RPA) was performed for risk stratification.ResultsA total of 617 patients were identified. Several independent prognostic factors were identified and integrated into the nomogram for OS2, including gender, age (2nd), nodal status (1st), node metastasis (2nd), and extrapulmonary metastasis (2nd). The calibration curves showed optimal agreement between the predictions and actual observations, and the c-index was 0.678. Surgery was associated with longer survival for msPSC patients. The prognosis of sublobectomy was comparable and inferior to that of lobectomy in the low- and moderate-risk groups, respectively. Radiotherapy was associated with better outcomes in patients who did not undergo surgery.ConclusionsThe RPA-based clinical nomogram appears to be suitable for the prognostic prediction and risk stratification of OS2 in msPSC. This practical system may help clinicians make decisions and design clinical studies.  相似文献   

15.
Purpose: obligatory cutaneous paraneoplastic disorders comprising acanthosis nigricans maligna, erythema gyratum repens, paraneoplastic pemphigus, hypertrichosis lanuginosa acquisita, erythema necrolyticum migrans and acrokeratosis paraneoplastica are rare. However, as markers of an underlying internal malignancy they are of utmost importance for the patient. Acrokeratosis paraneoplastica (first described by Gougerot and Rupp in 1922) was named after Bazex who had then reported several cases in a French dermatological journal since 1965 (Bazex et al. in Bull Soc Fr Dermatol Syphiligr 72:182, 1965; Bazex and Griffiths in Br J Dermatol 102:301–306, 1980). Method: the study is a clinical case of a patient with acrokeratosis paraneoplastica. Results: the patient was later diagnosed with a cervical lymph node metastasis and thereafter with a primary squamous cell carcinoma of the left upper lobe and upon treatment responded with the clearing of the skin changes. Conclusion: identification of a paraneoplastic syndrome may enhance the earlier diagnosis of the associated tumor and may thus enable curative treatment.  相似文献   

16.
The empirical criteria for defining a clinical subtype of lung cancer are gradually transiting from histopathology to genetic variations in driver genes. Targeting these driver mutations, such as sensitizing epidermal growth factor receptor (EGFR) mutations, has dramatically improved the prognosis of advanced non–small cell lung cancer (NSCLC). However, the clinical benefit of molecularly targeted therapy on NSCLC appears to be different between lung adenocarcinomas and squamous cell carcinomas (SqCCs). We report here that the resistance of lung SqCC harboring EGFR mutations to EGFR tyrosine kinase inhibitors (EGFR-TKIs) was due to the activation of BMP-BMPR-Smad1/5-p70S6K. The combined treatment of these tumor cells with EGFR-TKI, together with inhibitors specific to BMPR or downstream mTOR, effectively reversed the resistance to EGFR-TKI. Moreover, blocking the whole PI3K-AKT-mTOR pathway with the PI3K/mTOR dual inhibitor BEZ235 also showed efficacy in treating this subtype of lung SqCC. This study details the empirical basis for a feasible clinical solution for squamous cell carcinomas with EGFR mutations.Traditionally, the classification of lung cancer has been based primarily on histology and morphology (1, 2). With the identification of mutated driver oncogenes, methods from molecular pathology are gradually transforming the definition of the various types of lung cancers (3). Targeting gene aberrances such as epidermal growth factor receptor (EGFR) mutations (48) and anaplastic lymphoma kinase (ALK) fusion (9, 10) has significantly improved the prognosis of advanced non–small cell lung cancer (NSCLC). However, the clinical benefits brought by targeted therapies are mainly limited to nonsquamous NSCLC (11), while chemotherapy remains the major therapeutic choice for squamous cell carcinomas (SqCCs).Recent studies assessing somatic mutations and copy number alteration (CNA) profiles in SqCC performed by the Cancer Genome Atlas project and other investigators have disclosed specific gene mutations, including GRM8, BAI3, ERBB4, RUNX1T1, KEAP1, and FBXW7, and CNAs in 3q26, 24, 27, 32–34, and 8p12.35 in lung SqCC (12, 13). About half of all patients with lung SqCC carry multiple gene aberrances, indicating that complex genomic characterizations are more common in lung SqCC than in adenocarcinoma (ADC). Thus, successful therapeutic strategies that target a single driver gene in lung ADC might not be feasible for lung SqCC patients.Targeting EGFR mutations by EGFR tyrosine kinase inhibitors (TKIs) is one of the successful strategies in treating lung ADC. EGFR-TKIs obtained median progression-free survival (PFS) of 10–13 mo in EGFR-mutated lung ADC, but only ∼3 mo in lung SqCC with EGFR mutations (11, 14). Moreover, whether EGFR mutations exist in lung SqCC still remains controversial. There is a belief that EGFR mutations might not even occur in pure pulmonary SqCC, and that the occasional detection of these mutations in samples diagnosed as SqCC was due to mixed adenosquamous carcinoma and poorly differentiated ADC (15, 16). In current clinical practice, the utilization of EGFR-TKIs and the assessment of EGFR mutations are still routinely performed in lung SqCC, especially in nonsmokers. Therefore, it is critical to identify the subgroups of lung SqCC patients that are suitable for EGFR-TKI treatment. Exploring the mechanism of resistance to EGFR-TKIs in lung SqCC will deepen our understanding of the differences in tumorigenic profiling between lung SqCC and ADC, and should contribute to guiding clinical therapeutic decisions.In the present study, we demonstrated that lung SqCC patients with EGFR mutations indeed represent a subset of NSCLC. We further showed that lung SqCC cell lines were resistant to EGFR-TKIs both in vitro and in vivo. We also investigated the mechanisms of resistance to EGFR-TKI in this subset of patients and provided potential strategies for overcoming TKI resistance in these patients.  相似文献   

17.
18.
Insulin and insulin-like growth factor 1 (IGF-1) receptors share many downstream signaling pathways but have unique biological effects. To define the molecular signals contributing to these distinct activities, we performed global phosphoproteomics on cells expressing either insulin receptor (IR), IGF-1 receptor (IGF1R), or chimeric IR-IGF1R receptors. We show that IR preferentially stimulates phosphorylations associated with mammalian target of rapamycin complex 1 (mTORC1) and Akt pathways, whereas IGF1R preferentially stimulates phosphorylations on proteins associated with the Ras homolog family of guanosine triphosphate hydrolases (Rho GTPases), and cell cycle progression. There were also major differences in the phosphoproteome between cells expressing IR versus IGF1R in the unstimulated state, including phosphorylation of proteins involved in membrane trafficking, chromatin remodeling, and cell cycle. In cells expressing chimeric IR-IGF1R receptors, these differences in signaling could be mapped to contributions of both the extra- and intracellular domains of these receptors. Thus, despite their high homology, IR and IGF1R preferentially regulate distinct networks of phosphorylation in both the basal and stimulated states, allowing for the unique effects of these hormones on organismal function.

Insulin and insulin-like growth factor 1 (IGF-1) act through their cognate receptors to regulate a wide variety of biological processes. The insulin and IGF-1 receptors (IR and IGF1R) are highly homologous and share many overlapping signaling pathways (1). Activation of both receptors results in stimulation of two major canonical pathways of signaling: the PI 3-kinase/Akt pathway, which is linked to most metabolic actions of these hormones, and the Ras/MAP kinase pathway, which is linked to regulation of cell and organismal growth and differentiation (25). Perturbations in these pathways can lead to insulin resistance, as manifested by glucose intolerance, dyslipidemia, and increased risk of cardiovascular disease or alterations in growth at both the cellular and organismal level (1, 3). Due to the high degree of homology between these receptors and shared downstream signaling, defining differential molecular signatures has been challenging. This is further complicated by the fact that although insulin and IGF-1 preferentially bind to their own receptors, both ligands can also bind to the alternate receptor with lower affinity (6, 7). Many studies have tried to identify the differences between IR and IGF1R which account for their differences in vivo (2, 8). Some of the differential effects between insulin and IGF-1 have been ascribed to differences in the relative concentration of the hormones, the presence of IGF-1 binding proteins, the relative expression level of receptors in different tissues and the affinity of the two ligands for the two different receptors (1, 9).Some insight has come from the use of chimeric receptors. For example, activation of chimeric receptors containing the extracellular domains (ECDs) of the neurotrophin receptor (TrkC) fused to the intracellular domains (ICDs) of the insulin or IGF-1 receptors have suggested that the IGF1R intracellular domain (IGF1R-ICD) is more important for mitogenic response, while the IR-ICD is more strongly related to metabolic effects (10, 11). Similar conclusions have been reached using chemical and antibody-based inhibitors of these receptors (12). More recently, using chimeric receptors containing the ECD of IR fused to the ICD of IGF1R and vice versa, we showed that both the intracellular and extracellular domains of IR and IGF1R contribute to their effects on gene expression and that this is in part due to differences in their intracellular juxtamembrane regions and ability to engage IRS proteins versus Shc as substrates of the receptor (2).In addition to these ligand-stimulated and tyrosine-kinase-dependent events, previous studies from our laboratory have demonstrated a class of ligand and tyrosine-kinase-independent effects mediated by the unoccupied IR and IGF1R. Thus, cells lacking both IR and IGF1R are resistant to apoptosis (13) and show altered expression of a network of imprinted genes and miRNAs associated with changes in DNA methylation (14). These findings suggest that these receptors have receptor-dependent but ligand-independent signaling events, but how these are mediated is unknown.Intracellular signal transduction by membrane receptors such as IR and IGF1R is primarily mediated by the reversible phosphorylation of networks of signaling molecules. While IR and IGF1R are tyrosine kinases with a limited number of primary substrates, these in turn activate downstream kinases that lead to much broader networks of phosphorylation on serine and threonine residues. The resultant phosphorylated proteins may have altered activity levels, tertiary structure, and/or subcellular localization. To define the molecular differences in signaling between IR and IGF1R that create their unique physiological actions and to determine to what extent these differences are contributed by the ECDs vs. ICDs of these receptors, we performed global phosphoproteomic analysis of preadipocytes in which both endogenous receptors had been genetically inactivated to create double knock out (DKO) cells after which the cells were reconstituted with IR, IGF1R, and chimeric receptors consisting of the ECD of IR and the ICD of IGF1R (named hereafter as IR_IGF1R) and vice versa (i.e., IGF1R_IR) and studied both with and without ligand stimulation to understand signaling by the unoccupied receptor. We show that despite many similarities in signaling, there are major differences in the phosphoproteome mediated by activated IR versus IGF1R which contribute to their specific patterns of action. In addition, there are important and previously unrecognized differences in the basal phosphoproteome in cells expressing IR versus IGF1R. Using chimeric receptors we show that these differences in both the basal and stimulated states are contributed to by both the ICD and ECD of these receptors. Together, these unique signaling networks lead to the important differential effects of insulin and IGF-1 at the postreceptor level.  相似文献   

19.
20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号