Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
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
2.
J Pathol ; 228(4): 459-70, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22430872

ABSTRACT

Regional lymph node metastasis negatively affects prognosis in colon cancer patients. The molecular processes leading to regional lymph node metastasis are only partially understood and proteomic markers for metastasis are still scarce. Therefore, a tissue-based proteomic approach was undertaken for identifying proteins associated with regional lymph node metastasis. Two complementary tissue-based proteomic methods have been employed. MALDI imaging was used for identifying small proteins (≤25 kDa) in situ and label-free quantitative proteomics was used for identifying larger proteins. A tissue cohort comprising primary colon tumours without metastasis (UICC II, pN0, n = 21) and with lymph node metastasis (UICC III, pN2, n = 33) was analysed. Subsequent validation of identified proteins was done by immunohistochemical staining on an independent tissue cohort consisting of primary colon tumour specimens (n = 168). MALDI imaging yielded ten discriminating m/z species, and label-free quantitative proteomics 28 proteins. Two MALDI imaging-derived candidate proteins (FXYD3 and S100A11) and one from the label-free quantitative proteomics (GSTM3) were validated on the independent tissue cohort. All three markers correlated significantly with regional lymph node metastasis: FXYD3 (p = 0.0110), S100A11 (p = 0.0071), and GSTM3 (p = 0.0173). FXYD3 and S100A11 were more highly expressed in UICC II patient tumour tissues. GSTM3 was more highly expressed in UICC III patient tumour tissues. By our tissue-based proteomic approach, we could identify a large panel of proteins which are associated with regional lymph node metastasis and which have not been described so far. Here we show that novel markers for regional lymph metastasis can be identified by MALDI imaging or label-free quantitative proteomics and subsequently validated on an independent tissue cohort.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/secondary , Glutathione Transferase/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Proteomics , S100 Proteins/metabolism , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Female , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged , Prognosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
3.
J Proteomics ; 75(15): 4693-704, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22365974

ABSTRACT

To characterize proteomic changes found in Barrett's adenocarcinoma and its premalignant stages, the proteomic profiles of histologically defined precursor and invasive carcinoma lesions were analyzed by MALDI imaging MS. For a primary proteomic screening, a discovery cohort of 38 fresh frozen Barrett's adenocarcinoma patient tissue samples was used. The goal was to find proteins that might be used as markers for monitoring cancer development as well as for predicting regional lymph node metastasis and disease outcome. Using mass spectrometry for protein identification and validating the results by immunohistochemistry on an independent validation set, we could identify two of 60 differentially expressed m/z species between Barrett's adenocarcinoma and the precursor lesion: COX7A2 and S100-A10. Furthermore, among 22 m/z species that are differentially expressed in Barrett's adenocarcinoma cases with and without regional lymph node metastasis, one was identified as TAGLN2. In the validation set, we found a correlation of the expression levels of COX7A2 and TAGLN2 with a poor prognosis while S100-A10 was confirmed by multivariate analysis as a novel independent prognostic factor in Barrett's adenocarcinoma. Our results underscore the high potential of MALDI imaging for revealing new biologically significant molecular details from cancer tissues which might have potential for clinical application. This article is part of a Special Issue entitled: Translational Proteomics.


Subject(s)
Adenocarcinoma/metabolism , Annexin A2/biosynthesis , Biomarkers, Tumor/biosynthesis , Electron Transport Complex IV/biosynthesis , Gene Expression Regulation, Neoplastic , Microfilament Proteins/biosynthesis , Muscle Proteins/biosynthesis , S100 Proteins/biosynthesis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Female , Humans , Immunohistochemistry/methods , Male , Neoplasm Invasiveness , Prognosis , Proteomics/methods
4.
J Proteome Res ; 11(3): 1996-2003, 2012 Mar 02.
Article in English | MEDLINE | ID: mdl-22224404

ABSTRACT

In clinical diagnostics, it is of outmost importance to correctly identify the source of a metastatic tumor, especially if no apparent primary tumor is present. Tissue-based proteomics might allow correct tumor classification. As a result, we performed MALDI imaging to generate proteomic signatures for different tumors. These signatures were used to classify common cancer types. At first, a cohort comprised of tissue samples from six adenocarcinoma entities located at different organ sites (esophagus, breast, colon, liver, stomach, thyroid gland, n = 171) was classified using two algorithms for a training and test set. For the test set, Support Vector Machine and Random Forest yielded overall accuracies of 82.74 and 81.18%, respectively. Then, colon cancer liver metastasis samples (n = 19) were introduced into the classification. The liver metastasis samples could be discriminated with high accuracy from primary tumors of colon cancer and hepatocellular carcinoma. Additionally, colon cancer liver metastasis samples could be successfully classified by using colon cancer primary tumor samples for the training of the classifier. These findings demonstrate that MALDI imaging-derived proteomic classifiers can discriminate between different tumor types at different organ sites and in the same site.


Subject(s)
Adenocarcinoma/secondary , Neoplasms/metabolism , Proteome/metabolism , Adenocarcinoma/metabolism , Algorithms , Humans , Neoplasms/diagnosis , Neoplasms/pathology , Proteomics , Sensitivity and Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Support Vector Machine
5.
J Mol Med (Berl) ; 90(2): 163-74, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21938494

ABSTRACT

In papillary thyroid carcinoma (PTC), metastasis is a feature of an aggressive tumor phenotype. To identify protein biomarkers that distinguish patients with an aggressive tumor behavior, proteomic signatures in metastatic and non-metastatic tumors were investigated comparatively. In particular, matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry (IMS) was used to analyze primary tumor samples. We investigated a tumor cohort of PTC (n = 118) that were matched for age, tumor stage, and gender. Proteomic screening by MALDI-IMS was performed for a discovery set (n = 29). Proteins related to the discriminating mass peaks were identified by 1D-gel electrophoresis followed by mass spectrometry. The candidate proteins were subsequently validated by immunohistochemistry (IHC) using a tissue microarray for an independent PTC validation set (n = 89). In this study, we found 36 mass-to-charge-ratio (m/z) species that specifically distinguished metastatic from non-metastatic tumors, among which m/z 11,608 was identified as thioredoxin, m/z 11,184 as S100-A10, and m/z 10,094 as S100-A6. Furthermore, using IHC on the validation set, we showed that the overexpression of these three proteins was highly associated with lymph node metastasis in PTC (p < 0.005). For functional analysis of the metastasis-specific proteins, we performed an Ingenuity Pathway Analysis and discovered a strong relationship of all candidates with the TGF-ß-dependent EMT pathway. Our results demonstrated the potential application of the MALDI-IMS proteomic approach in identifying protein markers of metastasis in PTC. The novel protein markers identified in this study may be used for risk stratification regarding metastatic potential in PTC.


Subject(s)
Biomarkers, Tumor/metabolism , S100 Proteins/metabolism , Thioredoxins/metabolism , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/metabolism , Adult , Biomarkers, Tumor/genetics , Carcinoma , Carcinoma, Papillary , Female , Gene Expression Regulation, Neoplastic , Humans , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , S100 Proteins/genetics , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Thioredoxins/genetics , Thyroid Cancer, Papillary , Thyroid Neoplasms/pathology
6.
Oncotarget ; 2(12): 970-5, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22202598

ABSTRACT

Predicting the clinical course of osteosarcoma patients is a crucial prerequisite for a better treatment stratification in these highly aggressive neoplasms of bone. In search of new and reliable biomarkers we recently identified cysteine-rich intestinal protein 1 (CRIP1) to have significant prognostic impact in gastric cancer and therefore decided to investigate its role also in osteosarcoma. For this purpose we analyzed 223 pretherapeutic and well characterized osteosarcoma samples for their immunohistochemical expression of CRIP1 and correlated our findings with clinico-pathological parameters including follow­up, systemic spread and response to chemotherapy. Interestingly and contrarily to gastric cancer, we found CRIP1 expression more frequently in patients with long­term survival (10-year survival 73% in positive vs. 54% in negative cases, p = 0.0433) and without metastases (p = 0.0108) indicating a favorable prognostic effect. CRIP1 therefore seems to represent a promising new biomarker in osteosarcoma patients which should be considered for a prospective validation.


Subject(s)
Bone Neoplasms/genetics , Bone Neoplasms/pathology , Carrier Proteins/metabolism , LIM Domain Proteins/metabolism , Osteosarcoma/genetics , Osteosarcoma/pathology , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor , Child , Child, Preschool , Female , Humans , Male , Middle Aged , Neoplasm Metastasis/genetics , Prognosis , Retrospective Studies , Survival Analysis , Young Adult
7.
Am J Pathol ; 179(6): 2720-9, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22015459

ABSTRACT

Proteomics-based approaches allow us to investigate the biology of cancer beyond genomic initiatives. We used histology-based matrix-assisted laser desorption/ionization (MALDI) imaging mass spectrometry to identify proteins that predict disease outcome in gastric cancer after surgical resection. A total of 181 intestinal-type primary resected gastric cancer tissues from two independent patient cohorts were analyzed. Protein profiles of the discovery cohort (n = 63) were directly obtained from tumor tissue sections by MALDI imaging. A seven-protein signature was associated with an unfavorable overall survival independent of major clinical covariates. The prognostic significance of three individual proteins identified (CRIP1, HNP-1, and S100-A6) was validated immunohistochemically on tissue microarrays of an independent validation cohort (n = 118). Whereas HNP-1 and S100-A6 were found to further subdivide early-stage (Union Internationale Contre le Cancer [UICC]-I) and late-stage (UICC II and III) cancer patients into different prognostic groups, CRIP1, a protein previously unknown in gastric cancer, was confirmed as a novel and independent prognostic factor for all patients in the validation cohort. The protein pattern described here serves as a new independent indicator of patient survival complementing the previously known clinical parameters in terms of prognostic relevance. These results show that this tissue-based proteomic approach may provide clinically relevant information that might be beneficial in improving risk stratification for gastric cancer patients.


Subject(s)
Biomarkers, Tumor/metabolism , Carrier Proteins/metabolism , LIM Domain Proteins/metabolism , Neoplasm Proteins/metabolism , S100 Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Stomach Neoplasms/mortality , alpha-Defensins/metabolism , Adult , Aged , Aged, 80 and over , Female , Frozen Sections , Humans , Immunohistochemistry , Male , Middle Aged , Neoplasm Metastasis , Prognosis , Sensitivity and Specificity , Stomach Neoplasms/metabolism , Stomach Neoplasms/surgery
8.
J Proteome Res ; 9(12): 6317-22, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-21058730

ABSTRACT

HER2-testing in breast and gastric cancers is mandatory for the treatment with trastuzumab. We hypothesized that imaging mass spectrometry (IMS) of breast cancers may be useful for generating a classifier that may determine HER2-status in other cancer entities irrespective of primary tumor site. A total of 107 breast (n = 48) and gastric (n = 59) cryo tissue samples was analyzed by IMS (HER2 was present in 29 cases). The obtained proteomic profiles were used to create HER2 prediction models using different classification algorithms. A breast cancer proteome derived classifier, with HER2 present in 15 cases, correctly predicted HER2-status in gastric cancers with a sensitivity of 65% and a specificity of 92%. To create a universal classifier for HER2-status, breast and nonbreast cancer samples were combined, which increased sensitivity to 78%, and specificity was 88%. Our proof of principle study provides evidence that HER2-status can be identified on a proteomic level across different cancer types suggesting that HER2 overexpression may constitute a unique molecular event independent of the tumor site. Furthermore, these results indicate that IMS may be useful for the determination of potential drugable targets, as it offers a quicker, cheaper, and more objective analysis than the standard HER2-testing procedures immunohistochemistry and fluorescence in situ hybridization.


Subject(s)
Breast Neoplasms/metabolism , Proteomics/methods , Receptor, ErbB-2/analysis , Stomach Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Algorithms , Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Middle Aged , Prognosis , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Reproducibility of Results , Sensitivity and Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...