Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 18(5): 709-722, 2020 05.
Article in English | MEDLINE | ID: mdl-32079743

ABSTRACT

High-constitutive activity of the DNA damage response protein checkpoint kinase 1 (CHK1) has been shown in glioblastoma (GBM) cell lines and in tissue sections. However, whether constitutive activation and overexpression of CHK1 in GBM plays a functional role in tumorigenesis or has prognostic significance is not known. We interrogated multiple glioma patient cohorts for expression levels of CHK1 and the oncogene cancerous inhibitor of protein phosphatase 2A (CIP2A), a known target of high-CHK1 activity, and examined the relationship between these two proteins in GBM. Expression levels of CHK1 and CIP2A were independent predictors for reduced overall survival across multiple glioma patient cohorts. Using siRNA and pharmacologic inhibitors we evaluated the impact of their depletion using both in vitro and in vivo models and sought a mechanistic explanation for high CIP2A in the presence of high-CHK1 levels in GBM and show that; (i) CHK1 and pSTAT3 positively regulate CIP2A gene expression; (ii) pSTAT3 and CIP2A form a recursively wired transcriptional circuit; and (iii) perturbing CIP2A expression induces GBM cell senescence and retards tumor growth in vitro and in vivo. Taken together, we have identified an oncogenic transcriptional circuit in GBM that can be destabilized by targeting CIP2A. IMPLICATIONS: High expression of CIP2A in gliomas is maintained by a CHK1-dependent pSTAT3-CIP2A recursive loop; interrupting CIP2A induces cell senescence and slows GBM growth adding impetus to the development of CIP2A as an anticancer drug target.


Subject(s)
Autoantigens/metabolism , Biomarkers, Tumor/metabolism , Checkpoint Kinase 1/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , Apoptosis , Autoantigens/genetics , Biomarkers, Tumor/genetics , Cell Movement , Cell Proliferation , Checkpoint Kinase 1/genetics , Female , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Phosphorylation , Prognosis , STAT3 Transcription Factor/genetics , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Int J Cancer ; 138(3): 525-32, 2016 Feb 01.
Article in English | MEDLINE | ID: mdl-25628223

ABSTRACT

Cancerous Inhibitor of Protein Phosphatase 2A (CIP2A), a recently identified oncogene, has emerged as a potential drug target for a range of different tumor types. High CIP2A expression has been reported in almost all solid organ cancers and in some hematological tumors and is associated with high grade and poor prognosis. Notably, high CIP2A expression is determined in over 70% of tumor patient samples in the majority of human cancers. High expression of CIP2A has also been proposed as a useful biomarker that predicts therapeutic response to chemotherapeutics such as Bortezomib, Erlotinib, Checkpoint Kinase 1 inhibitors and pro-senescence based therapies. In this review, we highlight, critically evaluate and discuss the ambiguity in CIP2A's prognostic role in different human cancers and its role in modulating response and resistance to chemotherapeutics.


Subject(s)
Autoantigens/analysis , Biomarkers, Tumor/analysis , Membrane Proteins/analysis , Autoantigens/physiology , Checkpoint Kinase 1 , Genes, myc , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/physiology , Neoplasms/drug therapy , Neoplasms/pathology , Prognosis , Protein Kinases/physiology
3.
Oncotarget ; 6(23): 19661-70, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-25965834

ABSTRACT

Residual androgen receptor (AR)-signaling and presence of cancer stem-like cells (SCs) are the two emerging paradigms for clinically challenging castration-resistant prostate cancer (CRPC). Therefore, identification of AR-target proteins that are also overexpressed in the cancer SC population would be an attractive therapeutic approach.Our analysis of over three hundred clinical samples and patient-derived prostate epithelial cultures (PPECs), revealed Cancerous inhibitor of protein phosphatase 2A (CIP2A) as one such target. CIP2A is significantly overexpressed in both hormone-naïve prostate cancer (HN-PC) and CRPC patients . CIP2A is also overexpressed, by 3- and 30-fold, in HN-PC and CRPC SCs respectively. In vivo binding of the AR to the intronic region of CIP2A and its functionality in the AR-moderate and AR-high expressing LNCaP cell-model systems is also demonstrated. Further, we show that AR positively regulates CIP2A expression, both at the mRNA and protein level. Finally, CIP2A depletion reduced cell viability and colony forming efficiency of AR-independent PPECs as well as AR-responsive LNCaP cells, in which anchorage-independent growth is also impaired.These findings identify CIP2A as a common denominator for AR-signaling and cancer SC functionality, highlighting its potential therapeutic significance in the most clinically challenging prostate pathology: castration-resistant prostate cancer.


Subject(s)
Autoantigens/metabolism , Membrane Proteins/metabolism , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Prostatic Neoplasms, Castration-Resistant/therapy , Androgen Antagonists/pharmacology , Antineoplastic Agents, Hormonal/pharmacology , Autoantigens/genetics , Binding Sites , Cell Line, Tumor , Cell Proliferation , Cell Survival , Gene Expression Regulation, Neoplastic , Genetic Therapy/methods , Humans , Intracellular Signaling Peptides and Proteins , Introns , Male , Membrane Proteins/genetics , Molecular Targeted Therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Binding , RNA Interference , RNA, Messenger/metabolism , Receptors, Androgen/drug effects , Receptors, Androgen/metabolism , Signal Transduction , Time Factors , Transcription, Genetic , Transfection , Tumor Cells, Cultured , Up-Regulation
4.
Cancer Res ; 75(11): 2133-8, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25931285

ABSTRACT

Millions of DNA-damaging lesions occur every day in each cell of our bodies due to various stresses. The failure to detect and accurately repair these lesions can give rise to cells with high levels of endogenous DNA damage, deleterious mutations, or genomic aberrations. Such genomic instability can lead to the activation of specific signaling pathways, including the DNA damage response (DDR) pathway. Constitutive activation of DDR proteins has been observed in human tumor specimens from different cancer stages, including precancerous and metastatic cancers, although not in normal tissues. The tumor-suppressive role of DDR activity during the premalignant stage has been studied, and strong evidence is emerging for an oncogenic role for DDR proteins such as DNA-PK and CHK1 during the later stages of tumor development. However, the majority of current cancer therapies induce DNA damage, potentially exacerbating protumorigenic genomic instability and enabling the development of resistance. Therefore, elucidating the molecular basis of DNA damage-mediated genomic instability and its role in tumorigenesis is critical. Finally, I discuss the potential existence of distinct DNA damage thresholds at various stages of tumorigenesis and what the ramifications of such thresholds would be, including the ambiguous role of the DDR pathway in human cancers, therapy-induced malignancies, and enhanced therapies.


Subject(s)
Carcinogenesis/genetics , DNA Damage/genetics , Neoplasms, Radiation-Induced/genetics , Neoplasms/genetics , Carcinogenesis/drug effects , Carcinogenesis/radiation effects , DNA Repair/genetics , Drug Therapy , Drug-Related Side Effects and Adverse Reactions/genetics , Drug-Related Side Effects and Adverse Reactions/pathology , Genomic Instability , Humans , Neoplasms/chemically induced , Neoplasms/pathology , Neoplasms/radiotherapy , Neoplasms, Radiation-Induced/pathology , Signal Transduction/genetics
5.
Mol Cell Biol ; 35(12): 2165-72, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25870111

ABSTRACT

The bone morphogenetic protein (BMP)/SMAD signaling pathway is a critical regulator of angiogenic sprouting and is involved in vascular development in the embryo. SMAD1 and SMAD5, the core mediators of BMP signaling, are vital for this activity, yet little is known about their transcriptional regulation in endothelial cells. Here, we have integrated multispecies sequence conservation, tissue-specific chromatin, in vitro reporter assay, and in vivo transgenic data to identify and validate Smad1+63 and the Smad5 promoter as tissue-specific cis-regulatory elements that are active in the developing endothelium. The activity of these elements in the endothelium was dependent on highly conserved ETS, GATA, and E-box motifs, and chromatin immunoprecipitation showed high levels of enrichment of FLI1, GATA2, and SCL at these sites in endothelial cell lines and E11 dorsal aortas in vivo. Knockdown of FLI1 and GATA2 but not SCL reduced the expression of SMAD1 and SMAD5 in endothelial cells in vitro. In contrast, CD31(+) cKit(-) endothelial cells harvested from embryonic day 9 (E9) aorta-gonad-mesonephros (AGM) regions of GATA2 null embryos showed reduced Smad1 but not Smad5 transcript levels. This is suggestive of a degree of in vivo selection where, in the case of reduced SMAD1 levels, endothelial cells with more robust SMAD5 expression have a selective advantage.


Subject(s)
Endothelium/embryology , GATA2 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , Proto-Oncogene Protein c-fli-1/metabolism , Smad1 Protein/genetics , Smad5 Protein/genetics , Animals , Base Sequence , Cell Line , Endothelium/metabolism , GATA2 Transcription Factor/genetics , Gene Knockdown Techniques , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Proto-Oncogene Protein c-fli-1/genetics
6.
J Clin Diagn Res ; 8(1): 124-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24596742

ABSTRACT

BACKGROUND: Incidence of prostate cancer in India is relatively low compared to the western countries. Nevertheless, an increase by 1% yearly has been recorded in the last three years, thereby making early diagnosis of prostate cancer crucial for controlling its incidence. Differentiating between benign and malignant lesions has been a diagnostic dilemma, especially in prostate pathology. This is compounded by unavailability of modern tests in certain regions of developing nations. METHODS: A cohort of one hundred seventy six prostatomegaly patients used in the current study was obtained both retrospectively and prospectively at the Jawaharlal Nehru Medical College, Sawangi, Wardha, Maharashtra, India. Details of the patients were recorded which included their age. The samples were then cut into 5 sections, each of 5micron thickness. One section was preserved and the other 4 sections were subjected to Hematoxylin and Eosin (H and E), Periodic Acid-Schiff (PAS), Alcian Blue and AgNOR stains. Degree of differentiation was estimated and correlated with the Gleason score and the outcome of the stainings. RESULTS: Majority of benign prostatic hyperplasia and all primary carcinoma patients were in their sixth to eighth decade of life. While all the benign lesions were negative, 6 out of 9 primary prostate carcinomas were positive for Alcian Blue stain. Majority of both benign and malignant lesions were positive for Periodic Acid Schiff (PAS) stain. In terms of Argyrophilic Nucleolar Organiser Region (AgNOR) count per nucleus, the value in benign lesions was observed to be half the count observed in malignant lesions per nucleus. CONCLUSION: Although the potential use of the orthodox stains individually may not serve the purpose to differentiate between benign and malignant lesions, together they may have the potential to identify relatively more malignant cases. This may be helpful especially in low socio-economic countries and rural areas where molecular based tests may not yet be available.

7.
Cancer Res ; 73(22): 6548-53, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24204027

ABSTRACT

Protein phosphatase 2A (PP2A) complexes function as tumor suppressors by inhibiting the activity of several critical oncogenic signaling pathways. Consequently, inhibition of the PP2A phosphatase activity is one of many prerequisites for the transformation of normal human cells into cancerous cells. However, mechanisms for PP2A inactivation in human cancers are poorly understood. The aberrant expression of cancerous inhibitor of protein phosphatase 2A (CIP2A), a recently identified endogenous PP2A inhibitor in malignant cells, is one such mechanism. Various independent studies have validated CIP2A's role in promoting tumor growth and resistance to apoptosis and senescence-inducing therapies. Notably, high CIP2A expression predicts poor patient prognosis in several human cancer types. Among the oncogenic proteins dephosphorylated by PP2A, the MYC oncoprotein, which is phosphorylated at serine 62, has surfaced as a marker for the oncogenic activity of CIP2A. The positive-feedback loop between CIP2A and MYC augments the activity of MYC in cancer cells. In addition, CIP2A promotes the phosphorylation and activity of additional oncoproteins, including E2F1 and AKT. However, CIP2A is not essential for normal mouse growth and development. These findings indicate that CIP2A is a novel anticancer target based on PP2A reactivation and inhibition of the oncogenic activity of its downstream effectors. The potential approaches and feasibility of targeting CIP2A are discussed here.


Subject(s)
Autoantigens/physiology , Membrane Proteins/physiology , Molecular Targeted Therapy/methods , Neoplasms/genetics , Neoplasms/therapy , Oncogene Proteins/physiology , Animals , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/antagonists & inhibitors , Mice , Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/metabolism
8.
Cancer Res ; 73(22): 6757-69, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-24072747

ABSTRACT

Checkpoint kinase Chk1 is constitutively active in many cancer cell types and new generation Chk1 inhibitors show marked antitumor activity as single agents. Here we present a hitherto unrecognized mechanism that contributes to the response of cancer cells to Chk1-targeted therapy. Inhibiting chronic Chk1 activity in cancer cells induced the tumor suppressor activity of protein phosphatase protein phosphatase 2A (PP2A), which by dephosphorylating MYC serine 62, inhibited MYC activity and impaired cancer cell survival. Mechanistic investigations revealed that Chk1 inhibition activated PP2A by decreasing the transcription of cancerous inhibitor of PP2A (CIP2A), a chief inhibitor of PP2A activity. Inhibition of cancer cell clonogenicity by Chk1 inhibition could be rescued in vitro either by exogenous expression of CIP2A or by blocking the CIP2A-regulated PP2A complex. Chk1-mediated CIP2A regulation was extended in tumor models dependent on either Chk1 or CIP2A. The clinical relevance of CIP2A as a Chk1 effector protein was validated in several human cancer types, including neuroblastoma, where CIP2A was identified as an NMYC-independent prognostic factor. Because the Chk1-CIP2A-PP2A pathway is driven by DNA-PK activity, functioning regardless of p53 or ATM/ATR status, our results offer explanative power for understanding how Chk1 inhibitors mediate single-agent anticancer efficacy. Furthermore, they define CIP2A-PP2A status in cancer cells as a pharmacodynamic marker for their response to Chk1-targeted therapy.


Subject(s)
Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Protein Phosphatase 2/metabolism , Autoantigens/genetics , Autoantigens/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Checkpoint Kinase 1 , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Membrane Proteins/metabolism , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/pathology , Tumor Cells, Cultured , Tumor Suppressor Proteins/metabolism
9.
Cancer Discov ; 3(2): 182-97, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23306062

ABSTRACT

UNLABELLED: Senescence induction contributes to cancer therapy responses and is crucial for p53-mediated tumor suppression. However, whether p53 inactivation actively suppresses senescence induction has been unclear. Here, we show that E2F1 overexpression, due to p53 or p21 inactivation, promotes expression of human oncoprotein CIP2A, which in turn, by inhibiting PP2A activity, increases stabilizing serine 364 phosphorylation of E2F1. Several lines of evidence show that increased activity of E2F1-CIP2A feedback renders breast cancer cells resistant to senescence induction. Importantly, mammary tumorigenesis is impaired in a CIP2A-deficient mouse model, and CIP2A-deficient tumors display markers of senescence induction. Moreover, high CIP2A expression predicts for poor prognosis in a subgroup of patients with breast cancer treated with senescence-inducing chemotherapy. Together, these results implicate the E2F1-CIP2A feedback loop as a key determinant of breast cancer cell sensitivity to senescence induction. This feedback loop also constitutes a promising prosenescence target for therapy of cancers with an inactivated p53-p21 pathway. SIGNIFICANCE: It has been recently realized that most currently used chemotherapies exert their therapeutic effect at least partly by induction of terminal cell arrest, senescence. However, the mechanisms by which cell-intrinsic senescence sensitivity is determined are poorly understood. Results of this study identify the E2F1-CIP2A positive feedback loop as a key determinant of breast cancer cell sensitivity to senescence and growth arrest induction. Our data also indicate that this newly characterized interplay between 2 frequently overexpressed oncoproteins constitutes a promising prosenescence target for therapy of cancers with inactivated p53 and p21. Finally, these results may also facilitate novel stratification strategies for selection of patients to receive senescence-inducing cancer therapies.


Subject(s)
Autoantigens/genetics , Breast Neoplasms/genetics , Cellular Senescence , E2F1 Transcription Factor/genetics , Feedback, Physiological , Membrane Proteins/genetics , Animals , Antinematodal Agents/pharmacology , Autoantigens/metabolism , Blotting, Western , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Docetaxel , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , E2F1 Transcription Factor/metabolism , Embryo, Mammalian/cytology , Female , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , HCT116 Cells , Humans , Intracellular Signaling Peptides and Proteins , MCF-7 Cells , Mammary Neoplasms, Animal/drug therapy , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction , Taxoids/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vinblastine/analogs & derivatives , Vinblastine/pharmacology , Vinorelbine
10.
PLoS One ; 6(3): e17979, 2011 Mar 22.
Article in English | MEDLINE | ID: mdl-21445343

ABSTRACT

EGFR-MEK-ERK signaling pathway has an established role in promoting malignant growth and disease progression in human cancers. Therefore identification of transcriptional targets mediating the oncogenic effects of the EGFR-MEK-ERK pathway would be highly relevant. Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently characterized human oncoprotein. CIP2A promotes malignant cell growth and is over expressed at high frequency (40-80%) in most of the human cancer types. However, the mechanisms inducing its expression in cancer still remain largely unexplored. Here we present systematic analysis of contribution of potential gene regulatory mechanisms for high CIP2A expression in cancer. Our data shows that evolutionary conserved CpG islands at the proximal CIP2A promoter are not methylated both in normal and cancer cells. Furthermore, sequencing of the active CIP2A promoter region from altogether seven normal and malignant cell types did not reveal any sequence alterations that would increase CIP2A expression specifically in cancer cells. However, treatment of cancer cells with various signaling pathway inhibitors revealed that CIP2A mRNA expression was sensitive to inhibition of EGFR activity as well as inhibition or activation of MEK-ERK pathway. Moreover, MEK1/2-specific siRNAs decreased CIP2A protein expression. Series of CIP2A promoter-luciferase constructs were created to identify proximal -27 to -107 promoter region responsible for MEK-dependent stimulation of CIP2A expression. Additional mutagenesis and chromatin immunoprecipitation experiments revealed ETS1 as the transcription factor mediating stimulation of CIP2A expression through EGFR-MEK pathway. Thus, ETS1 is probably mediating high CIP2A expression in human cancers with increased EGFR-MEK1/2-ERK pathway activity. These results also suggest that in addition to its established role in invasion and angiogenesis, ETS1 may support malignant cellular growth via regulation of CIP2A expression and protein phosphatase 2A inhibition.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , MAP Kinase Kinase 1/metabolism , MAP Kinase Kinase 2/metabolism , Proto-Oncogene Protein c-ets-1/physiology , Steroid Hydroxylases/antagonists & inhibitors , Aryl Hydrocarbon Hydroxylases/genetics , Base Sequence , Cell Line, Tumor , DNA , DNA Methylation , Humans , Molecular Sequence Data , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Steroid Hydroxylases/genetics
11.
J Natl Cancer Inst ; 101(11): 793-805, 2009 Jun 03.
Article in English | MEDLINE | ID: mdl-19470954

ABSTRACT

BACKGROUND: Cancerous inhibitor of protein phosphatase 2A (CIP2A) is a recently identified human oncoprotein that stabilizes the c-Myc (MYC) protein. However, the clinical relevance of CIP2A to human cancers had not been demonstrated, but the mechanism of its regulation and its clinical role in cancer were completely unknown. METHODS: Tissue microarrays consisting of 223 gastric adenocarcinoma specimens were evaluated for the presence of CIP2A using immunohistochemistry, and the association of CIP2A expression with survival was assessed using Kaplan-Meier analysis. The effects of MYC and CIP2A on each other's expression and on cell proliferation were investigated in several gastric cancer cell lines using small interfering RNAs to CIP2A and MYC and immunoblotting. To further evaluate the role of MYC in CIP2A regulation, an inhibitor of MYC dimerization, 10058-F4, and an inducible MycER model were used. RESULTS: Expression of CIP2A protein was associated with reduced overall survival for gastric cancer patients with tumors 5 cm or smaller, with a 10-year overall survival in the CIP2A-immunopositive group of 8.1% as compared with 37.6% in the CIP2A-negative group (difference = 29.5%, 95% confidence interval = 12.5% to 46.5%, P = .001). In gastric cancer cell lines, CIP2A depletion led to decreased proliferation and anchorage-independent growth of the cells, as well as to reduced stability and expression of MYC protein. Interestingly, MYC depletion led to reduced expression of CIP2A mRNA and protein. Moreover, experiments with an MYC inhibitor and activator suggested that MYC directly promotes CIP2A gene expression. Finally, CIP2A and MYC immunopositivities were associated in gastric cancer specimens (P = .021). CONCLUSIONS: CIP2A immunopositivity is a predictor of survival for some subgroups of gastric cancer patients. CIP2A and MYC appear to be regulated in a positive feedback loop, wherein they promote each other's expression and gastric cancer cell proliferation.


Subject(s)
Adenocarcinoma/chemistry , Autoantigens/analysis , Biomarkers, Tumor/analysis , Feedback, Physiological , Membrane Proteins/analysis , Proto-Oncogene Proteins c-myc/metabolism , Stomach Neoplasms/chemistry , Adenocarcinoma/mortality , Autoantigens/genetics , Autoantigens/immunology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/immunology , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Down-Regulation , Finland , Gene Expression Regulation, Neoplastic , Humans , Immunoblotting , Immunohistochemistry , Intracellular Signaling Peptides and Proteins , Kaplan-Meier Estimate , Membrane Proteins/genetics , Membrane Proteins/immunology , Predictive Value of Tests , Prognosis , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/immunology , RNA, Messenger/analysis , RNA, Small Interfering/analysis , Registries , Retrospective Studies , Stomach Neoplasms/mortality , Tissue Array Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...