Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
1.
Hum Pathol ; 83: 212-223, 2019 01.
Article in English | MEDLINE | ID: mdl-30121370

ABSTRACT

Papillary renal cell carcinoma (PRCC) is currently divided in 2 subtypes. We reviewed a large cohort of PRCC and correlated subtype, morphological features and diagnostic marker expression with overall survival (OS) to uncover differences between the 2 subtypes. Three hundred seventy-six renal tumors initially diagnosed as PRCC with clinical and survival data were collected from the participating centers. Two hundred forty-six tumors were classified as PRCC1 (65.4%) and 130 as PRCC2 (34.6%) and graded according to the 2016 World Health Organization/International Society of Urological Pathology grading system. Morphological features (abundant cytoplasm, necrosis, fibrous stroma, foamy macrophages and psammoma bodies) were noted. Immunohistochemical stains (MIB1, p53, Racemase, EMA, CK7, CK20, E-Cadherin) were performed using tissue microarrays. χ2-Tests, log-rank tests and uni- and multivariate Cox regression analysis were performed. Both subtypes displayed different morphological features and immunohistochemical profiles: abundant cytoplasm was more frequent in PRCC2, while foamy macrophages were more common in PRCC1. Abundant cytoplasm and presence of psammoma bodies were associated with poorer OS. PRCC1 showed more frequent CK7 expression, PRCC2 more frequent E-Cadherin, p53 and higher MIB1 expression (>15%). Expression of Racemase and CK7 was associated with better OS, while high MIB1 (>15%) was associated with poorer OS. In multivariate analysis, the only independent predictors of OS were proliferation (MIB1), tumor stage, metastasis and age at surgery. Subtype was not an independent prognostic factor. Therefore, PRCC subtype on its own is not suitable for estimating survival. More data focusing on PRCC tumor biology is needed to define prognostic subgroups, especially in PRCC2.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/classification , Carcinoma, Renal Cell/mortality , Cell Proliferation/physiology , Child , Female , Humans , Kidney Neoplasms/classification , Kidney Neoplasms/mortality , Male , Middle Aged , Neoplasm Staging , Prognosis , Young Adult
2.
PLoS One ; 12(9): e0183917, 2017.
Article in English | MEDLINE | ID: mdl-28877230

ABSTRACT

BACKGROUND: Adjuvant therapy decisions in early breast cancer are based on accurate risk assessment. Urokinase plasminogen activator (uPA) and plaminogen activator inhibitor-1 (PAI-1) have been the first biomarkers in hormone receptor (HR) positive breast cancer to reach highest level of evidence. The EndoPredict test (EPclin) combines gene expression information with nodal status and tumor size. The aim of this prospective study was to compare uPA/PAI-1 and EPclin as prognostic biomarkers with regard to feasibility, risk stratification and impact on adjuvant therapy recommendation. MATERIALS AND METHOD: 395 patients with HR positive, HER2 negative, intermediate risk breast cancer were enrolled. Relations and concordance of histologic grading as well as EPclin and uPA/PAI-1 values were assessed by Spearman's rank correlation coefficient and Cohen's Kappa. To compare decision impact of EPclin and uPA/PAI-1 three independent case discussions were held: One with known uPA/PAI-1 and EPclin results, one blinded to EPclin alone and another one blinded to both EPclin and uPA/PAI-1. RESULTS: EPclin could be determined in all 395 (100%), uPA/PAI-1 in 190 (48%) of the tumor samples. EPclin allocated 250 patients (63%) to the low-risk group and 145 patients (37%) to the high-risk group, whereas uPA/PAI-1 allocated 88 patients (46%) to the low-risk group and 102 patients (54%) to the high-risk group. In 59% of cases, both tests showed concordant results. EPclin resulted more frequently in a change of therapy recommendation than the uPA/PAI-1 test (46% vs 24%). Recommendation of adjuvant chemotherapy (CTX) was abandoned twice as often by EPclin (45%) compared to uPA/PAI-1 (22%). CONCLUSION: In this first prospective comparison of EPclin and uPA/PAI-1 we found, that EPclin is superior to uPA/PAI-1 with respect to feasibility and decision impact. This leads to substantial avoidance of adjuvant CTX in endocrine-sensitive, HER2-negative breast cancer. Data collection for patients´ clinical outcome is ongoing.


Subject(s)
Breast Neoplasms/diagnosis , Plasminogen Activator Inhibitor 1/analysis , Urokinase-Type Plasminogen Activator/analysis , Adult , Aged , Aged, 80 and over , Biomarkers/analysis , Breast Neoplasms/chemistry , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Chemotherapy, Adjuvant , Feasibility Studies , Female , Humans , Middle Aged , Practice Guidelines as Topic , Prognosis , Prospective Studies , Risk Assessment
3.
Mol Oncol ; 9(1): 282-94, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25241146

ABSTRACT

The human epidermal growth factor receptor 2 (HER2) and the protein tyrosine kinase 6 (PTK6) are often co- and over-expressed in invasive breast cancers. At early diagnosis, only distinct groups, such as HER2-or hormone receptor-positive benefit from a targeted therapy. However, a part of these tumours develops resistance within a year of administration of the drug but the majority of the patients depends on general therapies with severe side effects. A PTK6-directed approach does not yet exist. In our present study, we successfully demonstrate, in vitro and in vivo, a significantly additive reduction of tumourigenesis of breast cancer cells simultaneously depleted of both HER2 and PTK6. In comparison with single RNAi approaches, the combined RNAi (co-RNAi) led to a stronger reduced phosphorylation of tumour-promoting proteins. Moreover, the co-RNAi additively decreased cell migration as well as two and three dimensional cell proliferation in vitro. The in vivo experiments showed an additive reduction (p < 0.00001) in the growth of xenografts due to the co-RNAi compared with HER2 or PTK6 RNAi alone. Interestingly, the complexes of HER2 or PTK6 with tumour-relevant interaction partners, such as HER3 or the insulin-like growth factor receptor 1 (IGF-1R), respectively, were also reduced in xenografts although their protein expression levels were not affected following the co-RNAi of HER2 and PTK6. Our present study reveals the potential of using combined HER2- and PTK6- knockdown as a powerful strategy for the treatment of breast cancers. Therefore, the combined inhibition of these proteins may represent an attractive tool for efficient therapy of breast cancers.


Subject(s)
Breast Neoplasms/metabolism , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , RNA Interference , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/metabolism , Receptors, Somatomedin/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Cell Line, Tumor , Female , Humans , Mice , Mice, Nude , Neoplasm Proteins/genetics , Protein-Tyrosine Kinases/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Receptor, IGF Type 1 , Receptors, Somatomedin/genetics , Xenograft Model Antitumor Assays
4.
J Pathol ; 235(1): 3-13, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25201776

ABSTRACT

An essential and so far unresolved factor influencing the evolution of cancer and the clinical management of patients is intratumour clonal and phenotypic heterogeneity. However, the de novo identification of tumour subpopulations is so far both a challenging and an unresolved task. Here we present the first systematic approach for the de novo discovery of clinically detrimental molecular tumour subpopulations. In this proof-of-principle study, spatially resolved, tumour-specific mass spectra were acquired, using matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry, from tissues of 63 gastric carcinoma and 32 breast carcinoma patients. The mass spectra, representing the proteomic heterogeneity within tumour areas, were grouped by a corroborated statistical clustering algorithm in order to obtain segmentation maps of molecularly distinct regions. These regions were presumed to represent different phenotypic tumour subpopulations. This was confirmed by linking the presence of these tumour subpopulations to the patients' clinical data. This revealed several of the detected tumour subpopulations to be associated with a different overall survival of the gastric cancer patients (p = 0.025) and the presence of locoregional metastases in patients with breast cancer (p = 0.036). The procedure presented is generic and opens novel options in cancer research, as it reveals microscopically indistinct tumour subpopulations that have an adverse impact on clinical outcome. This enables their further molecular characterization for deeper insights into the biological processes of cancer, which may finally lead to new targeted therapies.


Subject(s)
Breast Neoplasms/pathology , Gastrointestinal Stromal Tumors/pathology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Algorithms , Breast Neoplasms/mortality , Cluster Analysis , Female , Gastrointestinal Stromal Tumors/mortality , Humans , Male , Phenotype , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
5.
J Thorac Oncol ; 9(11): 1685-92, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25436802

ABSTRACT

INTRODUCTION: Detection of anaplastic lymphoma kinase (ALK)-gene rearrangements in non-small-cell lung cancer (NSCLC) is mainly performed by fluorescence in-situ hybridization (FISH). The question was raised if FISH might be replaced by immunohistochemistry (IHC) in a reliable and reproducible manner across different laboratories. METHODS: After calibration of the staining instruments and training of the observers to binary interpretation (positive versus negative), 15 NSCLC were independently tested for ALK protein expression by IHC only in a multicenter setting (16 institutes). Each laboratory utilized the VENTANA ALK-D5F3 IHC assay. As demonstrated by FISH the samples displayed unequivocal ALK break-positivity (6×) and negativity (7×), as well as ALK positive-"borderline" character (2×), which is challenging for FISH diagnosis and thus was RT-PCR-confirmed. RESULTS: All seven ALK FISH-negative cases were homogenously scored as ALK-IHC negative. All 16 participants scored the two ALK positive-"borderline" samples as unequivocally positive according to their protein expression. Concordant IHC interpretation was also noticed in four of six unequivocal ALK break positive cases. In two of six some observers described a weak/heterogeneous ALK-IHC staining. This would have resulted in a subsequent ALK-testing (FISH/PCR) in a routine diagnostic setting. CONCLUSIONS: This so-called "ALK-Harmonization-Study" shows for the first time that predictive semiquantitative IHC reveals reliable and reproducible results across several labs when methodology and interpretation are strictly defined and the pathologists are uniquely trained. The application of validated ALK IHC assays and its comparison to ALK-FISH is highly needed in future clinical trials. This might answer the question if ALK-IHC cannot only serve as a prescreening tool, but as a stand-alone test at least in cases displaying an unequivocally staining pattern as well as an alternative predictive test in samples with reduced FISH interpretability.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/enzymology , Receptor Protein-Tyrosine Kinases/analysis , Anaplastic Lymphoma Kinase , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Gene Rearrangement , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism
6.
Pathol Res Pract ; 210(11): 705-12, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25305148

ABSTRACT

Clinical studies and preclinical investigations are essential in order to test new therapies and diagnostic techniques aimed at sustainable improvements in the treatment of patients. Fortunately, the number of clinical studies is continuously increasing and pathology and tissue-based research are more frequently involved. Pathologists are essential in this process and committed to support it by joining forces with our clinical partners. The investigative diagnostic technologies we apply to human cell and tissue samples and our specific expertise are essential contributions to the quality and success of preclinical investigations, clinical studies, and the implementation of results into clinical diagnostic pathology. In order to support this process, the German Society of Pathology has formulated a statement on the participation in and support of clinical studies and other scientific investigations with a special focus on tissue-based research.


Subject(s)
Biomedical Research , Pathology , Clinical Trials as Topic , Germany , Humans , Societies, Medical
7.
Virchows Arch ; 465(5): 509-19, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25085759

ABSTRACT

The aim of our study was to evaluate the quality of histo- and cytomorphological features of PAXgene-fixed specimens and their suitability for histomorphological classification in comparison to standard formalin fixation. Fifteen colon cancer tissues were collected, divided into two mirrored samples and either formalin fixed (FFPE) or PAXgene fixed (PFPE) before paraffin embedding. HE- and PAS-stained sections were scanned and evaluated in a blinded, randomised ring trial by 20 pathologists from Europe and the USA using virtual microscopy. The pathologists evaluated histological grading, histological subtype, presence of adenoma, presence of lymphovascular invasion, quality of histomorphology and quality of nuclear features. Statistical analysis revealed that the reproducibility with regard to grading between both fixation methods was rather satisfactory (weighted kappa statistic (k w) = 0.73 (95 % confidence interval (CI), 0.41-0.94)), with a higher agreement between the reference evaluation and the PFPE samples (k w = 0.86 (95 % CI, 0.67-1.00)). Independent from preservation method, inter-observer reproducibility was not completely satisfactory (k w = 0.60). Histomorphological quality parameters were scored equal or better for PFPE than for FFPE samples. For example, overall quality and nuclear features, especially the detection of mitosis, were judged significantly better for PFPE cases. By contrast, significant retraction artefacts were observed more frequently in PFPE samples. In conclusion, our findings suggest that the PAXgene Tissue System leads to excellent preservation of histomorphology and nuclear features of colon cancer tissue and allows routine morphological diagnosis.


Subject(s)
Colonic Neoplasms/pathology , Tissue Fixation/methods , Adenocarcinoma, Mucinous/pathology , Formaldehyde , Humans , Observer Variation , Paraffin Embedding , Reagent Kits, Diagnostic , Reproducibility of Results , User-Computer Interface
8.
Ann Surg Oncol ; 21(3): 915-21, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24281419

ABSTRACT

BACKGROUND: For esophageal adenocarcinoma treated with neoadjuvant chemotherapy, postoperative staging classifications initially developed for non-pretreated tumors may not accurately predict prognosis. We tested whether a multifactorial TNM-based histopathologic prognostic score (PRSC), which additionally applies to tumor regression, may improve estimation of prognosis compared with the current Union for International Cancer Control/American Joint Committee on Cancer (UICC) staging system. PATIENTS AND METHODS: We evaluated esophageal adenocarcinoma specimens following cis/oxaliplatin-based therapy from two separate centers (center 1: n = 280; and center 2: n = 80). For the PRSC, each factor was assigned a value from 1 to 2 (ypT0-2 = 1 point; ypT3-4 = 2 points; ypN0 = 1 point; ypN1-3 = 2 points; ≤ 50 % residual tumor/tumor bed = 1 point; >50 % residual tumor/tumor bed = 2 points). The three-tiered PRSC was based on the sum value of these factors (group A: 3; group B: 4-5; group C: 6) and was correlated with patients' overall survival (OS). RESULTS: The PRSC groups showed significant differences with respect to OS (p < 0.0001; hazard ratio [HR] 2.2 [95 % CI 1.7-2.8]), which could also be demonstrated in both cohorts separately (center 1 p < 0.0001; HR 2.48 [95 % CI 1.8-3.3] and center 2 p = 0.015; HR 1.7 [95 % CI 1.1-2.6]). Moreover, the PRSC showed a more accurate prognostic discrimination than the current UICC staging system (p < 0.0001; HR 1.15 [95 % CI 1.1-1.2]), and assessment of two goodness-of-fit criteria (Akaike Information Criterion and Schwarz Bayesian Information Criterion) clearly supported the superiority of PRSC over the UICC staging. CONCLUSION: The proposed PRSC clearly identifies three subgroups with different outcomes and may be more helpful for guiding further therapeutic decisions than the UICC staging system.


Subject(s)
Adenocarcinoma/secondary , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Esophageal Neoplasms/pathology , Esophagectomy , Neoadjuvant Therapy , Adenocarcinoma/mortality , Adenocarcinoma/therapy , Adult , Aged , Aged, 80 and over , Chemotherapy, Adjuvant , Combined Modality Therapy , Esophageal Neoplasms/mortality , Esophageal Neoplasms/therapy , Female , Follow-Up Studies , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Prognosis , Survival Rate
9.
Ann Surg ; 259(1): 96-101, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24096772

ABSTRACT

OBJECTIVE: To determine the prevalence and localization of lymph node metastases in patients with pT1 carcinoma of the esophagus, esophagogastric junction, and stomach. BACKGROUND: Retrospective analysis and topographic description. METHODS: We included 793 consecutive patients with pT1 carcinomas who underwent primary surgery for squamous cell carcinoma (SCC) of the esophagus, adenocarcinomas of the esophagogastric junction (AEG), or gastric cancer (GC). Clinical records and pathology reports were reviewed, and the prevalence and topography of lymph node metastases were identified. RESULTS: The prevalence of lymph node metastases in SCC, AEG, and GC was 7%, 0%, and 5% for pT1a tumors and 24%, 18%, and 14% for pT1b tumors, respectively. Positive lymph node status was associated with worse overall survival (P<0.001). Not only infiltration of the submucosa (P=0.002) but also lymphatic vessel invasion (P<0.001), multifocal tumor growth (P=0.001), lower patient age (P=0.001), and poor tumor differentiation (P=0.05) were associated with nodal disease. These 5 parameters allowed the compilation of a nomogram to estimate the individual risk of lymph node metastases. In SCC, lymph node metastases were found from the neck to the celiac axis. In AEG, nodal disease was limited to the lower mediastinum and the D1 compartment. In GC, lymphatic spread exceeded the D1 compartment in 7% of node positive patients. CONCLUSIONS: Risk estimation for lymph node metastases should not be based on depth of tumor infiltration alone but additional clinicopathological parameters should also be considered. The extent of lymphadenectomy in surgical procedures should respect the presented topography of lymph node metastases.


Subject(s)
Adenocarcinoma/pathology , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Esophagogastric Junction/pathology , Lymph Nodes/pathology , Stomach Neoplasms/pathology , Adenocarcinoma/surgery , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/surgery , Esophageal Neoplasms/surgery , Esophagectomy , Esophagogastric Junction/surgery , Female , Humans , Lymph Node Excision , Lymphatic Metastasis , Male , Middle Aged , Prevalence , Retrospective Studies , Stomach Neoplasms/surgery , Survival Analysis , Young Adult
11.
Mod Pathol ; 27(1): 73-86, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23828315

ABSTRACT

Intraductal papillary neoplasms of the bile duct are still poorly characterized regarding (1) their molecular alterations during the development to invasive carcinomas, (2) their subtype stratification and (3) their biological behavior. We performed a multicenter study that analyzed these issues in a large European cohort. Intraductal papillary neoplasms of the bile duct from 45 patients were graded and subtyped using mucin markers and CDX2. In addition, tumors were analyzed for common oncogenic pathways, and the findings were correlated with subtype and grade. Data were compared with those from 22 extra- and intrahepatic cholangiocarcinomas. Intraductal papillary neoplasms showed a development from preinvasive low- to high-grade intraepithelial neoplasia to invasive carcinoma. Molecular and immunohistochemical analysis revealed mutated KRAS, overexpression of TP53 and loss of p16 in low-grade intraepithelial neoplasia, whereas loss of SMAD4 was found in late phases of tumor development. Alterations of HER2, EGFR, ß-catenin and GNAS were rare events. Among the subtypes, pancreato-biliary (36%) and intestinal (29%) were the most common, followed by gastric (18%) and oncocytic (13%) subtypes. Patients with intraductal papillary neoplasm of the bile duct showed a slightly better overall survival than patients with cholangiocarcinoma (hazard ratio (cholangiocarcinoma versus intraductal papillary neoplasm of the bile duct): 1.40; 95% confidence interval: 0.46-4.30; P=0.552). The development of biliary intraductal papillary neoplasms of the bile duct follows an adenoma-carcinoma sequence that correlates with the stepwise activation of common oncogenic pathways. Further large trials are needed to investigate and verify the finding of a better prognosis of intraductal papillary neoplasms compared with conventional cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms/chemistry , Bile Duct Neoplasms/genetics , Biomarkers, Tumor , Carcinoma in Situ/chemistry , Carcinoma in Situ/genetics , Carcinoma, Intraductal, Noninfiltrating/chemistry , Carcinoma, Intraductal, Noninfiltrating/genetics , Papilloma/chemistry , Papilloma/genetics , Adult , Aged , Aged, 80 and over , Bile Duct Neoplasms/mortality , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/chemistry , Bile Ducts, Intrahepatic/pathology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biopsy , Carcinoma in Situ/mortality , Carcinoma in Situ/pathology , Carcinoma, Intraductal, Noninfiltrating/mortality , Carcinoma, Intraductal, Noninfiltrating/pathology , Cholangiocarcinoma/chemistry , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Cyclin-Dependent Kinase Inhibitor p16/analysis , DNA Mutational Analysis , Disease Progression , Europe , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Mutation , Neoplasm Grading , Neoplasm Invasiveness , Papilloma/mortality , Papilloma/pathology , Prognosis , Proportional Hazards Models , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Signal Transduction , Time Factors , Tumor Suppressor Protein p53/analysis , ras Proteins/genetics
12.
PLoS One ; 8(10): e77825, 2013.
Article in English | MEDLINE | ID: mdl-24204986

ABSTRACT

Although the expression of cell signaling proteins is used as prognostic and predictive biomarker, variability of protein levels within tumors is not well studied. We assessed intratumoral heterogeneity of protein expression within primary ovarian cancer. Full-length proteins were extracted from 88 formalin-fixed and paraffin-embedded tissue samples of 13 primary high-grade serous ovarian carcinomas with 5-9 samples each. In addition, 14 samples of normal fallopian tube epithelium served as reference. Quantitative reverse phase protein arrays were used to analyze the expression of 36 cell signaling proteins including HER2, EGFR, PI3K/Akt, and angiogenic pathways as well as 15 activated (phosphorylated) proteins. We found considerable intratumoral heterogeneity in the expression of proteins with a mean coefficient of variation of 25% (range 17-53%). The extent of intratumoral heterogeneity differed between proteins (p<0.005). Interestingly, there were no significant differences in the extent of heterogeneity between phosphorylated and non-phosphorylated proteins. In comparison, we assessed the variation of protein levels amongst tumors from different patients, which revealed a similar mean coefficient of variation of 21% (range 12-48%). Based on hierarchical clustering, samples from the same patient clustered more closely together compared to samples from different patients. However, a clear separation of tumor versus normal tissue by clustering was only achieved when mean expression values of all individual samples per tumor were analyzed. While differential expression of some proteins was detected independently of the sampling method used, the majority of proteins only demonstrated differential expression when mean expression values of multiple samples per tumor were analyzed. Our data indicate that assessment of established and novel cell signaling proteins as diagnostic or prognostic markers may require sampling of serous ovarian cancers at several distinct locations to avoid sampling bias.


Subject(s)
Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/metabolism , Fallopian Tubes/metabolism , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/metabolism , Selection Bias , Carcinoma, Ovarian Epithelial , Case-Control Studies , Cohort Studies , Cystadenocarcinoma, Serous/pathology , ErbB Receptors/metabolism , Fallopian Tubes/pathology , Female , Humans , Neoplasm Grading , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , Protein Array Analysis , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction
13.
J Clin Invest ; 123(8): 3272-91, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23863708

ABSTRACT

Aging is a major risk factor for a large number of disorders and functional impairments. Therapeutic targeting of the aging process may therefore represent an innovative strategy in the quest for novel and broadly effective treatments against age-related diseases. The recent report of lifespan extension in mice treated with the FDA-approved mTOR inhibitor rapamycin represented the first demonstration of pharmacological extension of maximal lifespan in mammals. Longevity effects of rapamycin may, however, be due to rapamycin's effects on specific life-limiting pathologies, such as cancers, and it remains unclear if this compound actually slows the rate of aging in mammals. Here, we present results from a comprehensive, large-scale assessment of a wide range of structural and functional aging phenotypes, which we performed to determine whether rapamycin slows the rate of aging in male C57BL/6J mice. While rapamycin did extend lifespan, it ameliorated few studied aging phenotypes. A subset of aging traits appeared to be rescued by rapamycin. Rapamycin, however, had similar effects on many of these traits in young animals, indicating that these effects were not due to a modulation of aging, but rather related to aging-independent drug effects. Therefore, our data largely dissociate rapamycin's longevity effects from effects on aging itself.


Subject(s)
Aging/drug effects , Longevity/drug effects , Sirolimus/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Cell Transformation, Neoplastic/drug effects , Drug Evaluation, Preclinical , Granuloma/prevention & control , Immunoglobulins/blood , Leukocyte Count , Liver/drug effects , Liver/pathology , Liver Cirrhosis/prevention & control , Male , Maze Learning/drug effects , Mice , Mice, Inbred C57BL , Muscle Strength/drug effects , Oxygen Consumption/drug effects , Phenotype , Platelet Count , Psychomotor Performance/drug effects , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Thyroid Gland/drug effects , Thyroid Gland/pathology
14.
Am J Pathol ; 183(2): 352-68, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23791841

ABSTRACT

Within the Munich, Germany, N-ethyl-N-nitrosourea mouse mutagenesis program, we isolated a dominant Jak1 mouse model resembling phenotypic characteristics related to autoimmune disease. Chromosomal sequencing revealed a new Jak1 (p.Ser645Pro) point mutation at the conserved serine of the pseudokinase domain, corresponding to a somatic human mutation (p.Ser646Phe) inducing a constitutive activation of the Janus kinase (JAK)/STAT pathway. Morphologically, all Jak1(S645P+/-) mice showed a progressive structural deterioration of ears starting at the age of 4 months, with mononuclear cell infiltration into the dermis. Female mutant mice, in particular, developed severe skin lesions in the neck from 7 months of age. The IHC analysis of these lesions showed an activation of Stat3 downstream to Jak1(S645P) and elevated tissue levels of IL-6. Histopathological analysis of liver revealed a nodular regenerative hyperplasia. In the spleen, the number of Russell bodies was doubled, correlating with significant increased levels of all immunoglobulin isotypes and anti-DNA antibodies in serum. Older mutant mice developed thrombocytopenia and altered microcytic red blood cell counts. Jak1(S645P+/-) mice showed phenotypes related to impaired bone metabolism as increased carboxy-terminal collagen cross-link-1 levels and alkaline phosphatase activities in plasma, hypophosphatemia, and strongly decreased bone morphometric values. Taken together, Jak1(S645P+/-) mice showed an increased activation of the IL-6-JAK-STAT pathway leading to a systemic lupus erythematosus-like phenotype and offering a new valuable tool to study the role of the JAK/STAT pathway in disease development.


Subject(s)
Autoimmune Diseases/genetics , Janus Kinase 1/genetics , Point Mutation/genetics , Animals , Autoimmune Diseases/pathology , Biomarkers/metabolism , Disease Models, Animal , Ear Diseases/genetics , Female , Hyperplasia/genetics , Hyperplasia/pathology , Hypophosphatemia/genetics , Hypophosphatemia/pathology , Interleukin-6/metabolism , Liver/pathology , Male , Megakaryocytes/pathology , Mice , Mice, Inbred Strains , Mutagenesis/genetics , Phenotype , STAT3 Transcription Factor/metabolism , Skin Diseases, Genetic/genetics , Spleen/pathology , T-Lymphocyte Subsets , Thrombocytopenia/genetics
15.
Mol Cancer ; 12: 28, 2013 Apr 09.
Article in English | MEDLINE | ID: mdl-23570421

ABSTRACT

BACKGROUND: CRIP1 (cysteine-rich intestinal protein 1) has been found in several tumor types, its prognostic impact and its role in cellular processes, particularly in breast cancer, are still unclear. METHODS: To elucidate the prognostic impact of CRIP1, we analyzed tissues from 113 primary invasive ductal breast carcinomas using immunohistochemistry. For the functional characterization of CRIP1, its endogenous expression was transiently downregulated in T47D and BT474 breast cancer cells and the effects analyzed by immunoblotting, WST-1 proliferation assay and invasion assay. RESULTS: We found a significant correlation between CRIP1 and HER2 (human epidermal growth factor receptor 2) expression levels (p = 0.016) in tumor tissues. In Kaplan Meier analyses, CRIP1 expression was significantly associated with the distant metastases-free survival of patients, revealing a better prognosis for high CRIP1 expression (p = 0.039). Moreover, in multivariate survival analyses, the expression of CRIP1 was an independent negative prognostic factor, along with the positive prognosticators nodal status and tumor size (p = 0.029). CRIP1 knockdown in the T47D and BT474 breast cancer cell lines led to the increased phosphorylation of MAPK and Akt, to the reduced phosphorylation of cdc2, and to a significantly elevated cell proliferation in vitro (p < 0.001). These results indicate that reduced CRIP1 levels may increase cell proliferation and activate cell growth. In addition, CRIP1 knockdown increased cell invasion in vitro. CONCLUSIONS: Because the lack of CRIP1 expression in breast cancer tissue is significantly associated with a worse prognosis for patients and low endogenous CRIP1 levels in vitro increased the malignant potential of breast cancer cells, we hypothesize that CRIP1 may act as a tumor suppressor in proliferation and invasion processes. Therefore, CRIP1 may be an independent prognostic marker with significant predictive power for use in breast cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Carrier Proteins/metabolism , LIM Domain Proteins/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/mortality , Carrier Proteins/genetics , Cell Line, Tumor , Cell Proliferation , Female , Gene Silencing , Humans , LIM Domain Proteins/genetics , Neoplasm Invasiveness , Prognosis , Receptor, ErbB-2/metabolism
16.
J Pathol ; 230(4): 410-9, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23592244

ABSTRACT

Chemotherapeutic drugs kill cancer cells, but it is unclear why this happens in responding patients but not in non-responders. Proteomic profiles of patients with oesophageal adenocarcinoma may be helpful in predicting response and selecting more effective treatment strategies. In this study, pretherapeutic oesophageal adenocarcinoma biopsies were analysed for proteomic changes associated with response to chemotherapy by MALDI imaging mass spectrometry. Resulting candidate proteins were identified by liquid chromatography-tandem mass spectrometry (LC-MS/MS) and investigated for functional relevance in vitro. Clinical impact was validated in pretherapeutic biopsies from an independent patient cohort. Studies on the incidence of these defects in other solid tumours were included. We discovered that clinical response to cisplatin correlated with pre-existing defects in the mitochondrial respiratory chain complexes of cancer cells, caused by loss of specific cytochrome c oxidase (COX) subunits. Knockdown of a COX protein altered chemosensitivity in vitro, increasing the propensity of cancer cells to undergo cell death following cisplatin treatment. In an independent validation, patients with reduced COX protein expression prior to treatment exhibited favourable clinical outcomes to chemotherapy, whereas tumours with unchanged COX expression were chemoresistant. In conclusion, previously undiscovered pre-existing defects in mitochondrial respiratory complexes cause cancer cells to become chemosensitive: mitochondrial defects lower the cells' threshold for undergoing cell death in response to cisplatin. By contrast, cancer cells with intact mitochondrial respiratory complexes are chemoresistant and have a high threshold for cisplatin-induced cell death. This connection between mitochondrial respiration and chemosensitivity is relevant to anticancer therapeutics that target the mitochondrial electron transport chain.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Electron Transport Complex IV/metabolism , Esophageal Neoplasms/drug therapy , Mitochondria/drug effects , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Aged , Biomarkers, Tumor/genetics , Biopsy , Cell Line, Tumor , Chemotherapy, Adjuvant , Chromatography, Liquid , Cisplatin/administration & dosage , Down-Regulation , Drug Resistance, Neoplasm , Electron Transport Complex IV/genetics , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Fluorouracil/administration & dosage , Humans , Middle Aged , Mitochondria/enzymology , Mitochondria/pathology , Neoadjuvant Therapy , Precision Medicine , Proteomics/methods , RNA Interference , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Transfection , Treatment Outcome
17.
Clin Biochem ; 46(6): 539-45, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23388677

ABSTRACT

Despite the tendency to regard tumors as a simple mass of cancer cells, tumors have a high degree of complexity that is difficult to access with most analytical methods. Because the cancer tissue itself directly contains all information concerning proteomic and genetic changes, it represents the best possible sample material for any molecular research. However, an analytical method should also take advantage of morphological information contained within the cancer tissues, a feat that is not easily possible with methods based on sample homogenization such as conventional mass spectrometry. Matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry represents a method that allows the combination of mass spectrometric analyses with simultaneous histological evaluation to analyze various analytes such as proteins, peptides, lipids, or exogenous and endogenous small molecules. Spatially resolved mass spectrometric measurements are directly taken from a tissue section without destroying it. This combination allows for direct analysis of tumor samples while retaining the morphological information contained within the tissues, making it a very valuable tool in cancer research by complementing other currently used approaches. In this review, we discuss the position that MALDI imaging mass spectrometry currently occupies in the field of cancer research by showing its fields of application as well as the results and new discoveries that could be obtained using this method.


Subject(s)
Neoplasm Proteins/isolation & purification , Neoplasms/pathology , Proteomics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Gene Expression Profiling , Genetic Heterogeneity , Humans , Medical Laboratory Science , Neoplasm Proteins/classification , Neoplasm Staging , Neoplasms/diagnosis , Neoplasms/genetics , Organ Specificity
18.
Mol Cancer Res ; 11(4): 381-92, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23364537

ABSTRACT

Breast cancer is the most common malignancy in women of the Western world. One prominent feature of breast cancer is the co- and overexpression of HER2 and protein tyrosine kinase 6 (PTK6). According to the current clinical cancer therapy guidelines, HER2-overexpressing tumors are routinely treated with trastuzumab, a humanized monoclonal antibody targeting HER2. Approximately, 30% of HER2-overexpressing breast tumors at least initially respond to the anti-HER2 therapy, but a subgroup of these tumors develops resistance shortly after the administration of trastuzumab. A PTK6-targeted therapy does not yet exist. Here, we show for the first time that the simultaneous knockdown in vitro, compared with the single knockdown of HER2 and PTK6, in particular in the trastuzumab-resistant JIMT-1 cells, leads to a significantly decreased phosphorylation of crucial signaling proteins: mitogen-activated protein kinase 1/3 (MAPK 1/3, ERK 1/2) and p38 MAPK, and (phosphatase and tensin homologue deleted on chromosome ten) PTEN that are involved in tumorigenesis. In addition, dual knockdown strongly reduced the migration and invasion of the JIMT-1 cells. Moreover, the downregulation of HER2 and PTK6 led to an induction of p27, and the dual knockdown significantly diminished cell proliferation in JIMT-1 and T47D cells. In vivo experiments showed significantly reduced levels of tumor growth following HER2 or PTK6 knockdown. Our results indicate a novel strategy also for the treatment of trastuzumab resistance in tumors. Thus, the inhibition of these two signaling proteins may lead to a more effective control of breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Neoplasm Proteins/deficiency , Protein-Tyrosine Kinases/deficiency , Receptor, ErbB-2/deficiency , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Growth Processes/genetics , Cell Line, Tumor , Female , Gene Knockdown Techniques , Heterografts , Humans , Mice , Mice, Nude , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Signal Transduction , Transfection
19.
Hum Pathol ; 44(5): 829-36, 2013 May.
Article in English | MEDLINE | ID: mdl-23158210

ABSTRACT

Alterations of the epidermal growth factor receptor (EGFR) can be observed in a significant subset of esophageal adenocarcinomas (EACs), and targeted therapy against EGFR may become an interesting approach for the treatment of these tumors. Mutations of KRAS, NRAS, BRAF, and phosphatidylinositol-3-kinase catalytic subunit (PIK3CA) and deregulation of PTEN expression influence the responsiveness against anti-EGFR therapy in colorectal carcinomas. We investigated the prevalence of these events in a collection of 117 primary resected EACs, correlated the findings with EGFR expression and amplification, and determined their clinicopathologic impact. KRAS mutations were detected in 4 (3%) of 117 tumors (3× G12D and 1 G12V mutation). One tumor had a PIK3CA E545K mutation. Neither NRAS nor BRAF mutations were detected. Sixteen (14%) of 117 cases were negative for PTEN expression, determined by immunohistochemistry. Loss of PTEN was observed predominantly in advanced tumor stages (P = .004). There was no association between PTEN and EGFR status. Loss of PTEN was associated with shorter overall and disease-free survival (P < .001 each) and also an independent prognostic factor in multivariate analysis (P = .015). EGFR status had no prognostic impact in this case collection. In summary, loss of PTEN can be detected in a significant subset of EAC and is associated with an aggressive phenotype. Therefore, PTEN may be useful as a prognostic biomarker. In contrast, mutations of RAS/RAF/PIK3CA appear only very rarely, if at all, in EAC. A possible predictive role of PTEN in anti-EGFR treatment warrants further investigations, whereas determination of RAS/RAF/PIK3CA mutations may only have a minor impact in this context.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , ErbB Receptors/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , PTEN Phosphohydrolase/genetics , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Catalytic Domain , Class I Phosphatidylinositol 3-Kinases , Class Ia Phosphatidylinositol 3-Kinase/genetics , Disease-Free Survival , Esophageal Neoplasms/mortality , Female , GTP Phosphohydrolases/genetics , Humans , Male , Membrane Proteins/genetics , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras) , ras Proteins/genetics
20.
J Clin Oncol ; 31(2): 263-71, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23213098

ABSTRACT

PURPOSE: Validity of the seventh edition of the American Joint Committee on Cancer/International Union Against Cancer (AJCC/UICC) staging systems for gastric cancer has been evaluated in several studies, mostly in Asian patient populations. Only few data are available on the prognostic implications of the new classification system on a Western population. Therefore, we investigated its prognostic ability based on a German patient cohort. PATIENTS AND METHODS: Data from a single-center cohort of 1,767 consecutive patients surgically treated for gastric cancer were classified according to the seventh edition and were compared using the previous TNM/UICC classification. Kaplan-Meier analyses were performed for all TNM stages and UICC stages in a comparative manner. Additional survival receiver operating characteristic analyses and bootstrap-based goodness-of-fit comparisons via Bayesian information criterion (BIC) were performed to assess and compare prognostic performance of the competing classification systems. RESULTS: We identified the UICC pT/pN stages according to the seventh edition of the AJCC/UICC guidelines as well as resection status, age, Lauren histotype, lymph-node ratio, and tumor grade as independent prognostic factors in gastric cancer, which is consistent with data from previous Asian studies. Overall survival rates according to the new edition were significantly different for each individual's pT, pN, and UICC stage. However, BIC analysis revealed that, owing to higher complexity, the new staging system might not significantly alter predictability for overall survival compared with the old system within the analyzed cohort from a statistical point of view. CONCLUSION: The seventh edition of the AJCC/UICC classification was found to be valid with distinctive prognosis for each stage. However, the AJCC/UICC classification has become more complex without improving predictability for overall survival in a Western population. Therefore, simplification with better predictability of overall survival of patients with gastric cancer should be considered when revising the seventh edition.


Subject(s)
Stomach Neoplasms/classification , Chemotherapy, Adjuvant , Cohort Studies , Databases, Factual , Europe/epidemiology , Humans , Neoplasm Staging , Prognosis , Prospective Studies , Retrospective Studies , Stomach Neoplasms/epidemiology , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...