Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Microsc Res Tech ; 84(10): 2421-2433, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33929071

ABSTRACT

Our purpose was to employ microscopy images of amplified in breast cancer 1 (AIB1)-stained biopsy material of patients with colorectal cancer (CRC) to: (a) find statistically significant differences (SSDs) in the texture and color of the epithelial gland tissue, between 5-year survivors and non-survivors after the first diagnosis and (b) employ machine learning (ML) methods for predicting the CRC-patient 5-year survival. We collected biopsy material from 54 patients with diagnosed CRC from the archives of the University Hospital of Patras, Greece. Twenty-six of the patients had survived 5 years after the first diagnosis. We selected regions of interest containing the epithelial gland at different microscope lens magnifications. We computed 69 textural and color features. Furthermore, we identified features with SSDs between the two groups of patients and we designed a supervised ML system for predicting the CRC-patient 5-year survival. Additionally, we employed the VGG16 pretrained convolution neural network to extract deep learning (DL) features, the support vector machines classifier, and the bootstrap cross-validation method for boosting the accuracy of predicting 5-year survival. Fourteen features sustained SSDs between the two groups of patients. The supervised ML system achieved 87% accuracy in predicting 5-year survival. In comparison, the DL system, using images from all magnifications, gave 97% classification accuracy. Glandular texture in 5-year non-survivors appeared to be of lower contrast, coarseness, roughness, local pixel correlation, and lower AIB1 variation, all indicating loss of textural definition. The supervised ML system revealed useful information regarding features that discriminate between 5-year survivors and non-survivors while the DL system displayed superior accuracy by employing DL features.


Subject(s)
Colorectal Neoplasms , Microscopy , Biopsy , Humans , Machine Learning , Neural Networks, Computer
2.
Curr Mol Pharmacol ; 14(6): 1013-1027, 2021.
Article in English | MEDLINE | ID: mdl-32778046

ABSTRACT

Ovarian cancer is an aggressive disease, and only a few cases are diagnosed at early stages due to the absence of symptoms. Τhe majority of malignant ovarian tumors (>90%) are of epithelial origin and are subdivided into five histological sub types according to different molecular pathogenesis and clinical behavior. High-grade serous ovarian cancer is the most common subtype (70%). However, the different histotypes of ovarian cancer should be viewed as separate diseases both clinically and in biomarker studies. At present, surgical debulking and platinum/taxane - based chemotherapy is the standard of care for epithelial ovarian cancer. Most patients show an initial response to this therapeutic approach, but the majority of them experience disease recurrence at which point cure is no longer possible, due to acquired resistance in those chemotherapeutic regimens. Nevertheless, the current treatment model is still a "one-sizefits- all" approach. Epigenetic modifications represent heritable modifications in gene expression without alteration of the DNA sequence. DNA methylation is the best-studied epigenetic mechanism, and in epithelial ovarian cancer, the methylenome is widely altered. In addition, patterns of DNA methylation may represent potential diagnostic and prognostic markers as well as markers predictive of chemoresistance and potential therapeutic targets. This article systematically reviews the complex area of DNA methylation in ovarian carcinoma and summarizes the current implications and future perspectives of its use as a screening, diagnostic, prognostic and predictive tool as well as in personalized cancer therapy.


Subject(s)
DNA Methylation , Ovarian Neoplasms , Carcinoma, Ovarian Epithelial/complications , Carcinoma, Ovarian Epithelial/genetics , Epigenesis, Genetic , Humans , Neoplasm Recurrence, Local/complications , Neoplasm Recurrence, Local/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology
3.
Transl Oncol ; 14(1): 100912, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33074124

ABSTRACT

The Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) family of transcription factors plays an important role in immune responses and cancer development and progression. We have focused on NF-κB2 and RELB of the alternative pathway of NF-κB, which remains largely underexplored in colorectal cancer (CRC). We found that NF-κB2 and RELB protein levels were upregulated in tumour and surrounding stromal tissue compared to distant non-neoplastic tissue (NN) and associated stroma (p<0.001 in all associations). Moreover, low RELB protein expression was associated with decreased overall survival (p = 0.032). Lower RELB gene expression levels were observed in tumour compared to NN tissue (p = 0.003) and were associated with shorter time to progression (TTP) (p = 0.025). NF-κB2 gene expression levels were similar in tumour and NN tissue, but higher tumour levels were prognostic for improved survival (p = 0.038) and TTP (p<0.001). We also assessed the significance of two NF-κB2 genetic polymorphisms, rs12769316 and rs7897947. Both polymorphisms were associated with lymph node infiltration (p = 0.045 and p = 0.009, respectively). In addition, rs12769316 AA homozygotes relapsed less often compared to G allele carriers (p = 0.029). Moreover, rs7897947 allele frequencies differed significantly between CRC patients and healthy controls (p<0.001) and the minor allele (G) was associated with reduced risk for developing CRC (p<0.001, OR: 0.527, 95% CI: 0.387-0.717). In conclusion, the alternative NF-κB pathway appears deregulated in CRC. Moreover, NF-κB2 and RELB expression levels seem to be significant for the clinical outcome of CRC patients and rs7897947 appears to be a risk factor for CRC development.

4.
Cancers (Basel) ; 12(4)2020 Apr 07.
Article in English | MEDLINE | ID: mdl-32272654

ABSTRACT

: Deregulation of the transcribed ultra-conserved regions (T-UCRs) Uc160, Uc283, and Uc346 has been reported in colorectal cancer (CRC) recently. Here, we investigated promoter methylation of these T-UCRs during the adenoma-carcinoma sequence and their clinical significance in CRC patients. Methylation levels were assessed in CRC, adenomas, infiltrated lymph nodes, and metastatic tissue specimens. In situ hybridization was performed in representative tissue specimens. T-UCRs expression levels were also evaluated in HT-29 colon cancer cells before and after the acquired resistance to 5-fluorouracil (5-FU) and oxaliplatin. A gradual increase in T-UCRs methylation levels from hyperplastic polyps to adenomas and to in situ carcinomas (ISC) and a gradual decrease from ISC to infiltrative and metastatic carcinomas was observed (p < 0.001 for Uc160 and Uc283, p = 0.018 for Uc346). Uc160 and Uc283 methylation was associated with the grade of dysplasia in adenoma specimens (p = 0.034 and p = 0.019, respectively). Furthermore, higher Uc160 methylation, mainly in stage III and IV patients, was related to improved overall survival (OS) in univariate (p = 0.009; HR, 0.366) and multivariate analysis (p = 0.005; HR, 0.240). Similarly, higher methylation of Uc283 was associated with longer OS (p = 0.030). Finally, T-UCRs expression was significantly reduced in HT-29 cells after resistance to chemotherapy. This study suggests that promoter methylation of Uc160, Uc283, and Uc346 is altered during CRC development and that Uc160 and Uc283 methylation may have prognostic significance for CRC patients.

5.
J Clin Med ; 8(5)2019 May 24.
Article in English | MEDLINE | ID: mdl-31137630

ABSTRACT

An increasing number of studies implicates the NF-κB (Nuclear Factor of kappa light chain gene enhancer in B cells) alternative pathway in non-small-cell lung cancer (NSCLC). We assessed the clinical significance of CD40 (Tumor necrosis factor receptor superfamily member 5, TNFRSF5), BAFFR (B-cell activating factor receptor), RANK (Receptor activator of NF-κB) and LTßR (lymphotoxin ß receptor) receptors, which activate the alternative pathway of NF-κB, in NSCLC. Evaluation of CD40, BAFFR, RANK and LTßR expression was performed based on the Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) datasets, while protein expression was assessed by immunohistochemistry in specimens from 119 operated NSCLC patients. CD40 gene overexpression was correlated with improved five-year overall survival (OS) (p < 0.001), while increased BAFFR and LTßR mRNA levels were associated with worse OS in patients with adenocarcinomas (p < 0.001 and p < 0.001, respectively). Similarly, patients with adenocarcinomas exhibited a negative correlation between membranous BAFFR protein expression in carcinoma cells and three- and five-year survival (p = 0.021; HR, 4.977 and p = 0.030; HR, 3.358, respectively) as well as between BAFFR protein overexpression in cancer-associated fibroblasts (CAFs) and two-year survival (p = 0.036; HR, 1.983). Patients with increased LTßR nuclear protein staining or stage II patients with lower cytoplasmic LTßR protein expression had worse five-year OS (p = 0.039 and p = 0.008, respectively). Moreover, CD40 protein expression in tumor infiltrating lymphocytes (TILs) and CAFs was positively associated with metastatic spread while BAFFR protein expression in CAFs was negatively associated with bone metastasis (p = 0.041). Our data suggests that CD40, BAFFR, RANK and LTßR play an important role in NSCLC and further supports the role of NF-κB alternative pathway in NSCLC.

6.
Appl Immunohistochem Mol Morphol ; 27(10): 749-757, 2019.
Article in English | MEDLINE | ID: mdl-30095464

ABSTRACT

OBJECTIVE: The objective of this study was to study the textural and color changes occurring in the epithelial gland tissue with advancing colorectal cancer (CRC), utilizing immunohistochemical stain for AIB1 expression biopsy material. MATERIAL AND METHODS: Clinical material comprised biopsy specimens of 67 patients with a diagnosis of CRC. Two experienced pathologists used H&E-stained material for grading CRC lesions and immunohistochemical (IHC) stain for AIB1 expression. Twenty six patients were diagnosed with grade I, 28 with grade II, and 13 with grade III CRC. Guided by pathologists, we selected the regions of interest from AIB1-digitized images of each patient, encompassing the epithelial gland, and we computed 69 features, quantifying textural and color properties of the AIB1-stained lesions. We evaluated the statistical differences between grades by means of the Wilcoxon statistical test for each feature, and we assessed changes in feature values with advancing tumor grade by means of the Point Biserial Correlation. RESULTS: Statistical analysis revealed 14 single features, quantifying textural and color properties of the epithelial gland, which sustained statistically significant differences between LG-CRC and HG-CRC cases. These features were drawn from the gray-level image histogram, the cooccurrence matrix, the run length matrix, the discrete wavelet transform, the Tamura method, and the L*a*b color transform. CONCLUSIONS: A systematic statistical analysis of AIB1-stained biopsy material showed that high-grade CRC lesions contain higher intensity levels, appear coarser, are more homogeneous with smooth variation across the image, have lower contrast that is slowly varying across the image, have lower AIB1 staining, and have lower edges. A combination of textural and color attributes, evaluating image gray-tone distribution, textural roughness, inhomogeneity, AIB1 staining, and image coarseness should be considered in evaluating AIB1-stained CRC lesions.


Subject(s)
Colonic Neoplasms/diagnosis , Colorectal Neoplasms/diagnosis , Epithelial Cells/metabolism , Immunohistochemistry/methods , Nuclear Receptor Coactivator 3/metabolism , Adult , Aged , Aged, 80 and over , Biopsy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Epithelial Cells/pathology , Female , Humans , Image Processing, Computer-Assisted , Male , Middle Aged , Neoplasm Grading
7.
J Cutan Pathol ; 45(5): 337-347, 2018 May.
Article in English | MEDLINE | ID: mdl-29419888

ABSTRACT

BACKGROUND: Tumor infiltrating lymphocytes (TILs) represent important regulators of carcinogenesis. Cutaneous invasive squamous cell carcinoma (inSCC) develops through precursor lesions, namely in situ squamous cell carcinoma (isSCC) and actinic keratosis (AK), representing a natural model of carcinogenesis. The study evaluates TIL subpopulations in inSCC and its precursors by comparing 2 semiquantitative scoring systems, and assesses the presence of regulatory T-cells (Tregs) in these lesions. METHODS: Paraffin sections from 33 cases of AK, 19 isSCCs and 34 inSCCs with adjacent precursor lesions or normal skin (NS) were immunostained for CD3, CD4, CD8 and Foxp3. TIL subgroups were evaluated by the semiquantitative Klintrup-Mäkinen (K-M) score, and by a more detailed modification of this system. Treg counts were assessed by image analysis quantification. RESULTS: An increase of all TIL subpolulations from precursor lesions toward inSCC was shown by both scoring systems. Treg counts progressively increased from NS to AK and isSCC, but decreased in inSCC. Tregs were more numerous in pT2 and around indolent inSCCs compared to T1 and aggressive subtypes. CONCLUSIONS: T-cells and cytotoxic T-cells progressively increase in cutaneous squamous cell carcinogenesis, while Treg counts diminish in inSCC. The K-M score is an appropriate, easily applicable TIL scoring system in cutaneous inSCC.


Subject(s)
Carcinoma in Situ/immunology , Carcinoma, Squamous Cell/immunology , Keratosis, Actinic/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Skin Neoplasms/immunology , T-Lymphocyte Subsets/immunology , Aged , Aged, 80 and over , Carcinoma in Situ/pathology , Carcinoma, Squamous Cell/pathology , Female , Humans , Keratosis, Actinic/pathology , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Precancerous Conditions/immunology , Precancerous Conditions/pathology , Retrospective Studies , Skin Neoplasms/pathology , T-Lymphocyte Subsets/pathology
8.
Epigenetics ; 11(3): 184-93, 2016 03 03.
Article in English | MEDLINE | ID: mdl-26890396

ABSTRACT

Small cell prostate carcinoma (SCPC) morphology is rare at initial diagnosis but often emerges during prostate cancer progression and portends a dismal prognosis. It does not express androgen receptor (AR) or respond to hormonal therapies. Clinically applicable markers for its early detection and treatment with effective chemotherapy are needed. Our studies in patient tumor-derived xenografts (PDX) revealed that AR-negative SCPC (AR(-)SCPC) expresses neural development genes instead of the prostate luminal epithelial genes characteristic of AR-positive castration-resistant adenocarcinomas (AR(+)ADENO). We hypothesized that the differences in cellular lineage programs are reflected in distinct epigenetic profiles. To address this hypothesis, we compared the DNA methylation profiles of AR(-) and AR(+) PDX using methylated CpG island amplification and microarray (MCAM) analysis and identified a set of differentially methylated promoters, validated in PDX and corresponding donor patient samples. We used the Illumina 450K platform to examine additional regions of the genome and the correlation between the DNA methylation profiles of the PDX and their corresponding patient tumors. Struck by the low frequency of AR promoter methylation in the AR(-)SCPC, we investigated this region's specific histone modification patterns by chromatin immunoprecipitation. We found that the AR promoter was enriched in silencing histone modifications (H3K27me3 and H3K9me2) and that EZH2 inhibition with 3-deazaneplanocin A (DZNep) resulted in AR expression and growth inhibition in AR(-)SCPC cell lines. We conclude that the epigenome of AR(-) is distinct from that of AR(+) castration-resistant prostate carcinomas, and that the AR(-) phenotype can be reversed with epigenetic drugs.


Subject(s)
Carcinoma, Small Cell/genetics , DNA Methylation/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Adenosine/administration & dosage , Adenosine/analogs & derivatives , Animals , Carcinoma, Small Cell/pathology , Cell Line, Tumor , Cell Lineage/genetics , CpG Islands/genetics , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Promoter Regions, Genetic , Prostatic Neoplasms, Castration-Resistant/pathology , Receptors, Androgen/biosynthesis , Xenograft Model Antitumor Assays
9.
Oncology (Williston Park) ; 28(10): 831-8, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25323607

ABSTRACT

Atypical clinical features in men with prostate cancer-such as clinical evidence of disease progression in the absence of a proportional increase in serum prostate-specific antigen level, bulky symptomatic tumor masses, exclusive visceral metastases, or a predominance of lytic bone metastases-should alert the clinician that an aggressive prostate cancer variant is present or emerging. Aggressive variants of prostate cancer often take the form of neuroendocrine or small-cell carcinomas, which frequently lack androgen receptor expression and respond poorly to hormonal therapies. Indeed, the finding of neuroendocrine or small-cell prostate carcinoma indicates the need for multimodality treatments that incorporate early combination chemotherapy and locoregional control of bulky tumor deposits, including untreated or recurrent primaries. As we learn to recognize this prostate cancer variant more often, we are reminded that not all prostate cancers share the same biology and that the androgen receptor is not the sole driver of this disease.


Subject(s)
Carcinoma, Small Cell/pathology , Neuroendocrine Tumors/pathology , Prostatic Neoplasms/pathology , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Small Cell/therapy , Cell Differentiation , Combined Modality Therapy , Disease Progression , Fatal Outcome , Humans , Male , Neuroendocrine Tumors/therapy , Prostatic Neoplasms/therapy
10.
In Vivo ; 28(3): 315-26, 2014.
Article in English | MEDLINE | ID: mdl-24815833

ABSTRACT

BACKGROUND: Occludin and claudins are integral constituents of tight junction proteins and are de-regulated in various malignancies, including hepatocellular carcinoma (HCC). This study investigated whether expression of claudins 1, 4, 5, 7 and occludin may be used as prognostic markers for overall and disease-free survival in patients with HCC after hepatectomy. PATIENTS AND METHODS: The study included 67 hepatectomy specimens obtained from an equal number of patients with HCC who underwent partial hepatectomy at the Patras University Hospital for therapeutic reasons. Ten normal liver tissues were used as controls. Expression of claudins 1, 4, 5, 7 and occludin in liver tissues was assessed by immunochemistry. Clinicopathological features were also available for each case. RESULTS: Expression of claudins 1, 4, 5, 7 and occludin was significantly increased in HCC specimens compared to non-neoplastic liver tissues and normal controls (p<0.001 in each case) Moreover, there was a statistically significant association between low level of claudin-4 and advanced tumor grade (p=0.03). Down-regulation of claudin-1 was associated with low overall survival in univariate survival analysis (p=0.049) and Kaplan-Meier analysis (p=0.04). Multivariate analysis showed that the claudin-4 level was an independent factor for survival prognosis (p=0.01). In addition, down-regulation of claudin-4 expression was associated with increased recurrence rate and low disease-free survival rate in univariate analysis (p=0.038), Kaplan-Meier plot (p=0.013) and multivariate analysis (p=0.013). A low level of claudin-5 and high level of claudin-7 levels were independent negative prognostic factors according to multivariate analysis (p=0.015 and 0.009, respectively). CONCLUSION: The present study demonstrates that high expression of claudins 1, 4, 5 and down-regulation of claudin-7 are positive prognostic markers and are associated with good outcome and increased survival rates. Moreover, an increase in claudin-4 expression may serve as an independent positive prognostic factor for low recurrence rate after hepatectomy.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Claudins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Occludin/metabolism , Aged , Carcinoma, Hepatocellular/mortality , Claudin-1/genetics , Claudin-1/metabolism , Claudin-4/genetics , Claudin-4/metabolism , Claudin-5/genetics , Claudin-5/metabolism , Claudins/genetics , Female , Gene Expression , Humans , Immunohistochemistry , Liver Neoplasms/mortality , Male , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local , Neoplasm Staging , Occludin/genetics , Prognosis , Tumor Burden
12.
Prostate ; 72(15): 1638-47, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22457212

ABSTRACT

BACKGROUND: Hedgehog signaling is a stromal-mesenchymal pathway central to the development and homeostasis of both the prostate and the bone. Aberrant Hedgehog signaling activation has been associated with prostate cancer aggressiveness. We hypothesize that Hedgehog pathway is a candidate therapeutic target in advanced prostate cancer. We confirm increased Hedgehog signaling in advanced and bone metastatic castrate resistant prostate cancer and examine the pharmacodynamic effect of Smoothened inhibition by the novel reagent GDC-0449 in an experimental prostate cancer model. METHODS: Hedgehog signaling component expression was assessed in tissue microarrays of high grade locally advanced and bone metastatic disease. Male SCID mice subcutaneously injected with the bone forming xenograft MDA PCa 118b were treated with GDC-0449. Hedgehog signaling in the tumor microenvironment was assessed by proteomic and species specific RNA expression and compared between GDC-0449 treated and untreated animals. RESULTS: We observe Hedgehog signaling in high grade locally advanced and bone marrow infiltrating disease. Evidence of paracrine activation of Hedgehog signaling in the tumor xenograft, was provided by increased Sonic Hedgehog expression in human tumor epithelial cells, coupled with increased Gli1 and Patched1 expression in the murine stromal compartment, while normal murine stroma did not exhibit Hh signaling expression. GDC-0449 treatment attenuated Hh signaling as evidenced by reduced expression of Gli1 and Ptch1. Reduction in proliferation (Ki67) was observed with no change in tumor volume. CONCLUSIONS: GDC-0449 treatment is pharmacodynamically effective as evidenced by paracrine Hedgehog signaling inhibition and results in tumor cell proliferation reduction. Understanding these observations will inform the clinical development of therapy based on Hedgehog signaling inhibition.


Subject(s)
Adenocarcinoma/drug therapy , Anilides/pharmacology , Antineoplastic Agents/pharmacology , Hedgehog Proteins/antagonists & inhibitors , Osteogenesis/drug effects , Prostatic Neoplasms/drug therapy , Pyridines/pharmacology , Signal Transduction/drug effects , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Line, Tumor , Humans , Male , Mice , Mice, SCID , Neoplasm Transplantation , Ossification, Heterotopic/chemically induced , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tissue Array Analysis , Transplantation, Heterologous , Xenograft Model Antitumor Assays
13.
Clin Cancer Res ; 18(3): 726-36, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22298898

ABSTRACT

PURPOSE: To study Wnt/ß-catenin in castrate-resistant prostate cancer (CRPC) and understand its function independently of the ß-catenin-androgen receptor (AR) interaction. EXPERIMENTAL DESIGN: We carried out ß-catenin immunocytochemical analysis, evaluated TOP-flash reporter activity (a reporter of ß-catenin-mediated transcription), and sequenced the ß-catenin gene in MDA prostate cancer 118a, MDA prostate cancer 118b, MDA prostate cancer 2b, and PC-3 prostate cancer cells. We knocked down ß-catenin in AR-negative MDA prostate cancer 118b cells and carried out comparative gene-array analysis. We also immunohistochemically analyzed ß-catenin and AR in 27 bone metastases of human CRPCs. RESULTS: ß-Catenin nuclear accumulation and TOP-flash reporter activity were high in MDA prostate cancer 118b but not in MDA prostate cancer 2b or PC-3 cells. MDA prostate cancer 118a and MDA prostate cancer 118b cells carry a mutated ß-catenin at codon 32 (D32G). Ten genes were expressed differently (false discovery rate, 0.05) in MDA prostate cancer 118b cells with downregulated ß-catenin. One such gene, hyaluronan synthase 2 (HAS2), synthesizes hyaluronan, a core component of the extracellular matrix. We confirmed HAS2 upregulation in PC-3 cells transfected with D32G-mutant ß-catenin. Finally, we found nuclear localization of ß-catenin in 10 of 27 human tissue specimens; this localization was inversely associated with AR expression (P = 0.056, Fisher's exact test), suggesting that reduced AR expression enables Wnt/ß-catenin signaling. CONCLUSION: We identified a previously unknown downstream target of ß-catenin, HAS2, in prostate cancer, and found that high ß-catenin nuclear localization and low or no AR expression may define a subpopulation of men with bone metastatic prostate cancer. These findings may guide physicians in managing these patients.


Subject(s)
Glucuronosyltransferase/genetics , Prostatic Neoplasms/genetics , Signal Transduction/physiology , beta Catenin/genetics , Animals , Blotting, Western , Bone Neoplasms/secondary , Gene Expression Profiling , Glucuronosyltransferase/metabolism , Humans , Hyaluronan Synthases , Immunohistochemistry , Male , Mice , Mice, SCID , Mutation , Prostatic Neoplasms/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Androgen/biosynthesis , Receptors, Androgen/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , beta Catenin/metabolism
14.
Am J Pathol ; 180(3): 895-903, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22245216

ABSTRACT

The identification of new and effective therapeutic targets for the lethal, castration-resistant stage of prostate cancer (CRPC) has been challenging because of both the paucity of adequate frozen tissues and a lack of integrated molecular analysis. Therefore, in this study, we performed a genome-wide analysis of DNA copy number alterations from 34 unique surgical CRPC specimens and 5 xenografts, with matched transcriptomic profiling of 25 specimens. An integrated analysis of these data revealed that the asparagine synthetase (ASNS) gene showed a gain in copy number and was overexpressed at the transcript level. The overexpression of ASNS was validated by analyzing other public CRPC data sets. ASNS protein expression, as detected by reverse-phase protein lysate array, was tightly correlated with gene copy number. In addition, ASNS protein expression, as determined by IHC analysis, was associated with progression to a therapy-resistant disease state in TMAs that included 77 castration-resistant and 40 untreated prostate cancer patient samples. Knockdown of ASNS by small-interfering RNAs in asparagine-deprived media led to growth inhibition in both androgen-responsive (ie, LNCaP) and castration-resistant (ie, C4-2B) prostate cancer cell lines and in cells isolated from a CRPC xenograft (ie, MDA PCa 180-30). Together, our results suggest that ASNS is up-regulated in cases of CRPC and that depletion of asparagine using ASNS inhibitors will be a novel strategy for targeting CRPC cells.


Subject(s)
Aspartate-Ammonia Ligase/genetics , Prostatic Neoplasms/enzymology , Animals , Aspartate-Ammonia Ligase/metabolism , DNA/genetics , DNA Copy Number Variations/genetics , Gene Expression Profiling , Gene Knockdown Techniques , Genome-Wide Association Study , Male , Mice , Mice, Inbred C57BL , Mice, SCID , Neoplasms, Hormone-Dependent/enzymology , Neoplasms, Hormone-Dependent/genetics , Orchiectomy , Prostatic Neoplasms/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/pharmacology , Transplantation, Heterologous , Tumor Cells, Cultured
15.
J Neurooncol ; 106(1): 23-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21735116

ABSTRACT

Estrogen receptors alpha (ERα) and beta (ERß) and their co-regulatory proteins are key components of complex signaling networks that specifically regulate the growth and development of various tissues and tumors. Still, their protein expression profiles and possible role in the pathogenesis of astrocytic tumors remain largely unknown. The purpose of the present study is to evaluate the differential protein expression of ΕRα, ERß, and their co-activators, AIB1, TIF2, and PELP1 in astrocytic tumors of World Health Organization (WHO) grade II-IV, using immunohistochemistry. Potential correlations with clinicopathological parameters and patient prognosis were also explored. ERα protein expression was undetectable while ERß levels were significantly decreased with progression of tumor grade (P < 0.001). High expression of ERß was an independent favorable prognostic factor on multivariate analysis (P = 0.003). Expression of AIB1, TIF2, and PELP1 was not correlated with ERß expression and followed an opposite trend, with increasing levels in high-grade relative to low-grade tumors (P < 0.001). Univariate survival analysis revealed that high AIB1, TIF2, and PELP1 expression was associated with worse prognosis (P = 0.049, P = 0.033, and P = 0.020, respectively). ERß and ER co-activators AIB1, TIF2, and PELP1 appear to play an important role in the pathogenesis and progression of astrocytic tumors and might have prognostic significance. The mechanisms underlying their involvement in astrocytic tumorigenesis, as well as their utility for prognostic and therapeutic purposes merit further investigation.


Subject(s)
Astrocytoma/metabolism , Biomarkers, Tumor/analysis , Brain Neoplasms/metabolism , Co-Repressor Proteins/analysis , Nuclear Receptor Coactivator 2/analysis , Nuclear Receptor Coactivator 3/analysis , Transcription Factors/analysis , Adult , Analysis of Variance , Astrocytoma/diagnosis , Astrocytoma/pathology , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Estrogen Receptor beta/metabolism , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Paraffin Embedding , Prognosis , Survival Analysis , Tissue Fixation
16.
J Clin Oncol ; 30(6): 637-43, 2012 Feb 20.
Article in English | MEDLINE | ID: mdl-22184395

ABSTRACT

PURPOSE: Persistent androgen signaling is implicated in castrate-resistant prostate cancer (CRPC) progression. This study aimed to evaluate androgen signaling in bone marrow-infiltrating cancer and testosterone in blood and bone marrow and to correlate with clinical observations. PATIENTS AND METHODS: This was an open-label, observational study of 57 patients with bone-metastatic CRPC who underwent transiliac bone marrow biopsy between October 2007 and March 2010. Patients received oral abiraterone acetate (1 g) once daily and prednisone (5 mg) twice daily. Androgen receptor (AR) and CYP17 expression were assessed by immunohistochemistry, testosterone concentration by mass spectrometry, AR copy number by polymerase chain reaction, and TMPRSS2-ERG status by fluorescent in situ hybridization in available tissues. RESULTS: Median overall survival was 555 days (95% CI, 440 to 965+ days). Maximal prostate-specific antigen decline ≥ 50% occurred in 28 (50%) of 56 patients. Homogeneous, intense nuclear expression of AR, combined with ≥ 10% CYP17 tumor expression, was correlated with longer time to treatment discontinuation (> 4 months) in 25 patients with tumor-infiltrated bone marrow samples. Pretreatment CYP17 tumor expression ≥ 10% was correlated with increased bone marrow aspirate testosterone. Blood and bone marrow aspirate testosterone concentrations declined to less than picograms-per-milliliter levels and remained suppressed at progression. CONCLUSION: The observed pretreatment androgen-signaling signature is consistent with persistent androgen signaling in CRPC bone metastases. This is the first evidence that abiraterone acetate achieves sustained suppression of testosterone in both blood and bone marrow aspirate to less than picograms-per-milliliter levels. Potential admixture of blood with bone marrow aspirate limits our ability to determine the origin of measured testosterone.


Subject(s)
Androstenols/therapeutic use , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Histone Deacetylase Inhibitors/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Receptors, Androgen/metabolism , Adult , Aged , Aged, 80 and over , Androstenes , Androstenols/adverse effects , Bone Marrow/metabolism , Bone Neoplasms/metabolism , Castration , Disease-Free Survival , Drug Administration Schedule , Fatigue/chemically induced , Histone Deacetylase Inhibitors/adverse effects , Humans , Infections/chemically induced , Infusions, Intravenous , Male , Middle Aged , Nausea/chemically induced , Prostatic Neoplasms/surgery , Remission Induction , Testosterone/blood , Testosterone/metabolism , Vascular Diseases/chemically induced , Young Adult
17.
Clin Cancer Res ; 18(3): 666-77, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22156612

ABSTRACT

PURPOSE: Small-cell prostate carcinoma (SCPC) morphology predicts for a distinct clinical behavior, resistance to androgen ablation, and frequent but short responses to chemotherapy. We sought to develop model systems that reflect human SCPC and can improve our understanding of its biology. EXPERIMENTAL DESIGN: We developed a set of castration-resistant prostate carcinomas xenografts and examined their fidelity to their human tumors of origin. We compared the expression and genomic profiles of SCPC and large-cell neuroendocrine carcinoma (LCNEC) xenografts to those of typical prostate adenocarcinoma xenografts. Results were validated immunohistochemically in a panel of 60 human tumors. RESULTS: The reported SCPC and LCNEC xenografts retain high fidelity to their human tumors of origin and are characterized by a marked upregulation of UBE2C and other mitotic genes in the absence of androgen receptor (AR), retinoblastoma (RB1), and cyclin D1 (CCND1) expression. We confirmed these findings in a panel of samples of CRPC patients. In addition, array comparative genomic hybridization of the xenografts showed that the SCPC/LCNEC tumors display more copy number variations than the adenocarcinoma counterparts. Amplification of the UBE2C locus and microdeletions of RB1 were present in a subset, but none displayed AR nor CCND1 deletions. The AR, RB1, and CCND1 promoters showed no CpG methylation in the SCPC xenografts. CONCLUSION: Modeling human prostate carcinoma with xenografts allows in-depth and detailed studies of its underlying biology. The detailed clinical annotation of the donor tumors enables associations of anticipated relevance to be made. Future studies in the xenografts will address the functional significance of the findings.


Subject(s)
Carcinoma, Small Cell/genetics , Carcinoma, Small Cell/metabolism , Disease Models, Animal , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Aged , Animals , Carcinoma, Small Cell/pathology , Comparative Genomic Hybridization , Gene Expression Profiling , Humans , Immunohistochemistry , Male , Mice , Middle Aged , Prostatic Neoplasms/pathology , Tissue Array Analysis , Transplantation, Heterologous
18.
Histopathology ; 58(7): 1037-47, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21707705

ABSTRACT

AIMS: The hedgehog (Hh) signalling pathway has been implicated in the pathogenesis and aggressiveness of prostate cancer through epithelial-mesenchymal interactions. The aim of this study was to elucidate the cell-type partitioned expression of the Hh pathway biomarkers in the non-neoplastic and tumour microenvironments and to correlate it with the grade and stage of prostate cancer. METHODS AND RESULTS: Expression of the Hh pathway components (Shh, Smo, Ptch, Gli1) in the microenvironment of non-neoplastic peripheral zone (n = 119), hormone-naive primary prostate carcinoma (n = 141) and castrate-resistant bone marrow metastases (n = 53) was analysed using immunohistochemistry in tissue microarrays and bone marrow sections. Results showed that epithelial Shh, Smo and Ptch expression was up-regulated, whereas stromal Smo, Ptch, and Gli1 expression was down-regulated in prostate carcinomas compared to non-neoplastic peripheral zone tissue. Ptch expression was modulated further in high-grade and high-stage primary tumours and in bone marrow metastases. Hh signalling correlated with ki67 and vascular endothelial growth factor (VEGF) but not with CD31 expression. CONCLUSION: Our results highlight the importance of Hh-mediated epithelial-mesenchymal interactions in the non-neoplastic prostate and imply that shifting the balance from paracrine towards autocrine signalling is important in the pathogenesis and progression of prostate carcinoma.


Subject(s)
Adenocarcinoma/metabolism , Autocrine Communication/physiology , Hedgehog Proteins/metabolism , Prostatic Neoplasms/metabolism , Adenocarcinoma/secondary , Adult , Aged , Biomarkers, Tumor/metabolism , Bone Marrow Neoplasms/metabolism , Bone Marrow Neoplasms/secondary , Humans , Male , Middle Aged , Prostatic Neoplasms/pathology , Tissue Array Analysis
19.
J Clin Oncol ; 29(18): 2574-81, 2011 Jun 20.
Article in English | MEDLINE | ID: mdl-21606419

ABSTRACT

PURPOSE: Clinicians are increasingly willing to treat prostate cancer within the primary site in the presence of regional lymph node or even limited distant metastases. However, no formal study on the merits of this approach has been reported. We used a preoperative clinical discovery platform to prioritize pathways for assessment as therapeutic targets and to test the hypothesis that the primary site harbors potentially lethal tumors after aggressive treatment. PATIENTS AND METHODS: Patients with locally advanced or lymph node-metastatic prostate cancer underwent 1 year of androgen ablation and three cycles of docetaxel therapy, followed by prostatectomy. All specimens were characterized for stage by accepted criteria. Expression of select molecular markers implicated in disease progression and therapy resistance was determined immunohistochemically and compared with that in 30 archived specimens from untreated patients with high-grade prostate cancer. Marker expression was divided into three groups: intracellular signaling pathways, stromal-epithelial interaction pathways, and angiogenesis. RESULTS: Forty patients were enrolled, 30 (75%) of whom underwent prostatectomy and two (5%) who underwent cystoprostatectomy. Twenty-nine specimens contained sufficient residual tumor for inclusion in a tissue microarray. Immunohistochemical analysis showed increased epithelial and stromal expression of CYP17, SRD5A1, and Hedgehog pathway components, and modulations of the insulin-like growth factor I pathway. CONCLUSION: A network of molecular pathways reportedly linked to prostate cancer progression is activated after 1 year of therapy; biomarker expression suggests that potentially lethal cancers persist in the primary tumor and may contribute to progression.


Subject(s)
Adenocarcinoma/pathology , Androgens , Antineoplastic Agents, Phytogenic/therapeutic use , Neoadjuvant Therapy , Neoplasm Proteins/analysis , Neoplasms, Hormone-Dependent/pathology , Neoplastic Stem Cells/chemistry , Prostatectomy , Prostatic Neoplasms/pathology , Taxoids/therapeutic use , Adenocarcinoma/chemistry , Adenocarcinoma/drug therapy , Adenocarcinoma/surgery , Adult , Aged , Androgens/metabolism , Biomarkers, Tumor , Cell Adhesion Molecules/genetics , Combined Modality Therapy , Cystectomy , Disease Progression , Docetaxel , Gene Expression Profiling , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasms, Hormone-Dependent/chemistry , Neoplasms, Hormone-Dependent/drug therapy , Neoplasms, Hormone-Dependent/surgery , Neoplastic Stem Cells/pathology , Neovascularization, Physiologic/genetics , Prospective Studies , Prostatic Neoplasms/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/surgery , Signal Transduction/genetics , Time Factors
20.
Nat Rev Urol ; 7(9): 494-509, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20818327

ABSTRACT

Solid tumors can be thought of as multicellular 'organs' that consist of a variety of cells as well as a scaffold of noncellular matrix. Stromal-epithelial crosstalk is integral to prostate cancer progression and metastasis, and androgen signaling is an important component of this crosstalk at both the primary and metastatic sites. Intratumoral production of androgen is an important mechanism of castration resistance and has been the focus of novel therapeutic approaches with promising results. Various other pathways are important for stromal-epithelial crosstalk and represent attractive candidate therapeutic targets. Hedgehog signaling has been associated with tumor progression, growth and survival, while Src family kinases have been implicated in tumor progression and in regulation of cancer cell migration. Fibroblast growth factors and transforming growth factor beta signaling regulate cell proliferation, apoptosis and angiogenesis in the prostate cancer microenvironment. Integrins mediate communication between the cell and the extracellular matrix, enhancing growth, migration, invasion and metastasis of cancer cells. The contribution of stromal-epithelial crosstalk to prostate cancer initiation and progression provides the impetus for combinatorial microenvironment-targeting strategies.


Subject(s)
Prostatic Neoplasms/drug therapy , Tumor Microenvironment , Clinical Trials as Topic , Humans , Male , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...