Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cancer Treat Rev ; 123: 102674, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38176220

ABSTRACT

The Cancer Drug Development Forum (CDDF)'s 'Histology independent drug development - is this the future for cancer drugs?' workshop was set up to explore the current landscape of histology independent drug development, review the current regulatory landscape and propose recommendations for improving the conduct of future trials. The first session considered lessons learnt from previous trials, including innovative solutions for reimbursement. The session explored why overall survival represents the most valuable endpoint, and the importance of duration of response, which can be captured with swimmer and spider plots. The second session on biomarker development and treatment optimisation considered current regulations for companion diagnostics, FDA guidance on histology independent drug development in oncology, and the need to establish cut-offs for the biomarker of tumour mutational burden to identify the patients most likely to benefit from PDL1 treatment. The third session reviewed novel trial designs, including basket, umbrella and platform trials, and statistical approaches of hierarchical modelling where homogeneity between study cohorts enables information to be borrowed between cohorts. The discussion highlighted the need to agree 'common assessment standards' to facilitate pooling of data across studies. In the fourth session, the sharing of data sets was recognised as a key step for improving equity of access to precision medicines across Europe. The session considered how the European Health Data Space (EHDS) could streamline access to medical records, emphasizing the importance of introducing greater accountability into the digital space. In conclusion the workshop proposed 11 recommendations to facilitate histology agnostic drug development.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Drug Development , Medical Oncology , Biomarkers, Tumor
2.
Clin Pharmacol Ther ; 114(2): 316-324, 2023 08.
Article in English | MEDLINE | ID: mdl-37132507

ABSTRACT

The new In Vitro Diagnostic Regulation (EU) 2017/746 (IVDR) introduces important changes in the EU legal framework for companion diagnostics (CDx), including a new risk-based classification system for in vitro diagnostic tests (IVDs), a first legal definition for CDx and enhanced involvement of notified bodies in the conformity assessment and certification process of CDx. The IVDR also establishes an important link between the assessment of a CDx and the corresponding medicinal product by requiring the notified body to seek a scientific opinion from the medicines regulator on the suitability of the CDx for use with the concerned medicinal product(s) before issuing an IVD certificate. Whereas the IVDR aims at establishing a robust regulatory framework for IVDs, it is also associated with several challenges, such as insufficient capacity of notified bodies and readiness of manufacturers. To ensure timely access for patients to essential IVDs, a progressive roll-out for this new legislation has been introduced. In addition, the new consultation process for CDx requires increased collaboration and alignment of assessments performed by the different stakeholders involved in this process. The European Medicines Agency (EMA) and notified bodies are currently building experience based on the first CDx consultation procedures that have been submitted from January 2022 onward. In the current article, we describe the new European regulatory framework for certification of CDx and highlight several challenges for medicine and CDx co-development. In addition, we briefly touch upon the interplay between the Clinical Trial Regulation (EU) No. 536/2014 (CTR) and the IVDR.


Subject(s)
Precision Medicine , Humans , European Union , Precision Medicine/methods , Biomarkers
3.
Article in German | MEDLINE | ID: mdl-33001218

ABSTRACT

In recent years, a breakthrough in tumor therapy was achieved with the development of checkpoint inhibitors. Checkpoint inhibitors activate the immune defense against tumors by overcoming the inhibitory effect of specific cell surface proteins acting as control points, the so-called checkpoints. This article provides an overview of the mode of action of approved checkpoint inhibitors and the status of current clinical development.The previously approved checkpoint inhibitors, monoclonal antibodies directed against the checkpoints CTLA­4 and PD-1/PD-L1, are used in various tumor entities (including lung, kidney, and urothelial carcinoma; head and neck cancer; melanoma; and Hodgkin lymphoma). For the first time, long-term survival has been achieved in some of these patients with advanced tumors. Unfortunately, this efficacy can be observed only in a small proportion of the treated patients, depending on the tumor indication. Improved efficacy is envisioned by patient selection via predictive biomarkers and the development of combination therapies. Mandatory testing of the expression level of the predictive PD-L1 biomarker is already required in some indications to select patients with an enhanced benefit/risk relationship.


Subject(s)
Immunotherapy , Melanoma , Antibodies, Monoclonal/therapeutic use , Combined Modality Therapy , Germany , Humans
4.
Oncotarget ; 7(15): 20945-52, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26967391

ABSTRACT

BACKGROUND: Tenascin-C is overexpressed in the stroma of most solid malignancies and may function as a diagnostic tumor marker. This study was conducted to evaluate the potential significance of Tenascin-C as a predictive marker for tumor progression in the sera of non-small cell lung cancer (NSCLC) patients. RESULTS: Serum concentration of Tenascin-C is significantly elevated in NSCLC patients compared to healthy controls (p=0.013). The sensitivity of Tenascin-C in detecting NSCLC was 74% at a specificity of 57%. Elevated Tenascin-C serum values are associated with larger tumor size and lymph node involvement (p=0.022 and p=0.036, respectively). The Kaplan-Meyer-curves showed a significant association of Tenascin-C with the patient's overall survival (p=0.004), but not with the recurrence-free survival (p=0.328). METHODS: We quantified Tenascin-C in the sera of 103 NSCLC patients and 76 healthy blood donors by enzyme-linked immune-absorbance assay tests. Prognostic significance was determined by area under the curve analysis and Youden-index. The results were correlated with clinical, histopathological, and patient survival data (Chi-square test, Kaplan-Meier analysis, log-rank test, multivariate Cox-regression analysis). CONCLUSION: Although significantly elevated in patients with NSCLC, the sensitivity and specificity of the Tenascin-C serum quantification test was low. However, although failing to be an independent prognosticator in multivariate analysis, the results implicate Tenascin-C as a predictive prognostic marker for NSCLC patients. The data must be further validated in future prospective trials with larger patient cohorts.


Subject(s)
Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/secondary , Lung Neoplasms/pathology , Tenascin/blood , Adult , Aged , Aged, 80 and over , Carcinoma, Non-Small-Cell Lung/blood , Case-Control Studies , Female , Follow-Up Studies , Humans , Lung Neoplasms/blood , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Survival Rate
5.
Stem Cells ; 32(6): 1429-36, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24501004

ABSTRACT

Recently, the activated leukocyte cell adhesion molecule (CD166) was identified as an "inert" cancer stem cell (CSC) marker for non-small cell lung cancer (NSCLC). Few data exist regarding the clinical relevance of CD166 expression in NSCLC. We evaluated the expression of CD166 using immunohistochemistry in a large cohort of NSCLC patients (n = 1,910) on a tissue microarray basis. Expression was inversely associated with tumor size and lymph node status. Grading slightly failed to be significantly inversely associated, and survival analysis revealed no significant survival benefit of CD166-positive patients. Due to the results of this study, the theory of CD166 as a CSC marker for NSCLC must be questioned. The association of CD166 with smaller tumors and no nodal metastases does not make it a typical CSC marker. Further studies are required to investigate the functional role of CD166 in NSCLC.


Subject(s)
Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion Molecules, Neuronal/metabolism , Fetal Proteins/metabolism , Lung Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Adult , Aged , Aged, 80 and over , Antigens, CD/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Cell Adhesion Molecules, Neuronal/genetics , Cell Line, Tumor , Female , Fetal Proteins/genetics , Flow Cytometry , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Middle Aged , Multivariate Analysis , Prognosis , Proportional Hazards Models , Regression Analysis
6.
J Transl Med ; 11: 238, 2013 Sep 30.
Article in English | MEDLINE | ID: mdl-24074251

ABSTRACT

BACKGROUND: The chemokine CXCL12 and its receptor CXCR4 play a major role in tumor invasion, proliferation and metastasis in different malignant diseases, including esophageal carcinoma, amongst others. CXCR7 was recently identified as a novel alternate receptor for CXCL12. The aim of this study was to evaluate the prognostic impact of expression of chemokine receptor CXCR7 in patients with esophageal carcinoma (EC). METHODS: Expression of CXCR7 in primary tumors, lymph nodes and distant metastases of 299 patients with EC was evaluated by immunohistochemistry on a tissue microarray and compared with clinical and histopathological data. RESULTS: In esophageal cancer sections, CXCR7-specific reactivity was apparent in 45% of the squamous cell carcinomas (ESCC), but only occasionally in adenocarcinomas. No correlation between CXCR4 and CXCR7 expression was evident. We correlated expression with clinical and histopathological characteristics, but could not find any association. CONCLUSIONS: Contrary to the other known CXCL12 receptor, CXCR4, CXCR7 is expressed in ESCC only, underlining the divergent mechanisms and backgrounds of EAC and ESCC. The results of the study do not indicate a significant functional role for CXCR7 in EAC or ESCC of the esophagus. However, its variable expression in the main two main types of EC needs to be further investigated.


Subject(s)
Esophageal Neoplasms/metabolism , Receptors, CXCR/metabolism , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Esophageal Neoplasms/pathology , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Male , Middle Aged
7.
PLoS One ; 7(6): e39018, 2012.
Article in English | MEDLINE | ID: mdl-22745698

ABSTRACT

BACKGROUND: This study was conducted to evaluate the expression of the activated leukocyte cell adhesion molecule (ALCAM) in pancreatic cancer (PAC) and to determine whether or not the ectodomain shedding of ALCAM (s-ALCAM) could serve as a biomarker in the peripheral blood of PAC patients. MATERIAL AND METHODS: Tissue specimens and blood sera of patients with PAC (n = 264 and n = 116, respectively) and the sera of 115 patients with chronic pancreatitis (CP) were analyzed via ALCAM immunohistochemistry and s-ALCAM ELISA tests. Results were correlated with clinical, histopathological, and patient survival data (Chi-square test, Kaplan-Meier analysis, log-rank test, respectively). RESULTS: ALCAM was expressed in the majority of PAC lesions. Immunohistochemistry and serum ELISA tests revealed no association between ALCAM expression in primary tumors or s-ALCAM and clinical or histopathological data. Neither ALCAM nor s-ALCAM showed a significant impact regarding overall survival (p = 0.261 and p = 0.660, respectively). S-ALCAM serum levels were significantly elevated compared to the sera of CP patients (p<0.001). The sensitivity of s-ALCAM in detecting PAC was 58.6% at a specificity of 73.9% (AUC = 0.69). CONCLUSIONS: ALCAM is expressed in the majority of PAC lesions, but statistical analysis revealed no association with clinical or pathological data. Although significantly elevated in patients with PAC, the sensitivity and specificity of the s-ALCAM serum quantification test was low. Therefore, its potential as a novel diagnostic marker for PAC remains elusive and further investigations are required.


Subject(s)
Antigens, CD/blood , Antigens, CD/metabolism , Cell Adhesion Molecules, Neuronal/blood , Cell Adhesion Molecules, Neuronal/metabolism , Fetal Proteins/blood , Fetal Proteins/metabolism , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , In Vitro Techniques , Male , Middle Aged , Pancreatitis, Chronic/blood , Pancreatitis, Chronic/metabolism
8.
Oncology ; 82(6): 305-12, 2012.
Article in English | MEDLINE | ID: mdl-22555284

ABSTRACT

OBJECTIVE: Conflicting results have been reported about activated leukocyte cell adhesion molecule (ALCAM) expression in breast cancer and its prognostic value. Little is known about the role of ALCAM levels in the serum of breast cancer patients. METHODS: We analyzed soluble ALCAM (sALCAM) levels in the serum of 157 primary breast cancer patients and 48 healthy women by ELISA. In addition, we determined ALCAM protein expression by Western blot analysis (n = 120) and mRNA expression by cDNA microarray analysis (n = 115) in the tumor tissue of corresponding patients. RESULTS: sALCAM levels differed between patients and healthy controls (median 24.2 vs. 18.9 ng/ml, p < 0.001). We observed no correlation between serum levels and protein or mRNA expression in corresponding tumors (r < 0.1, p = n.s.). sALCAM levels were not correlated with histological type, grading, tumor stage, or patient age, but elevated sALCAM levels were associated with shorter disease-free survival (HR = 1.97, 95% CI 1.01-3.2, p = 0.043). CONCLUSIONS: Our results indicate that sALCAM can be detected in the serum of patients with primary breast cancer. Elevated serum levels might indicate more aggressive tumor behavior as they might be an independent factor for a worse prognosis in breast cancer patients.


Subject(s)
Antigens, CD/blood , Breast Neoplasms/blood , Cell Adhesion Molecules, Neuronal/blood , Fetal Proteins/blood , Adult , Aged , Aged, 80 and over , Antigens, CD/biosynthesis , Antigens, CD/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Adhesion Molecules, Neuronal/biosynthesis , Cell Adhesion Molecules, Neuronal/genetics , Disease-Free Survival , Female , Fetal Proteins/biosynthesis , Fetal Proteins/genetics , Humans , Middle Aged , Neoplasm Grading , Prognosis , RNA, Messenger/genetics
9.
BMC Cancer ; 12: 140, 2012 Apr 04.
Article in English | MEDLINE | ID: mdl-22475274

ABSTRACT

BACKGROUND: An altered expression of the activated leukocyte cell adhesion molecule (ALCAM) is associated with cancer progression in various cancer types. In some cancers ALCAM has a prognostic value or is predictive for the benefit of therapeutic interventions. To date there are no data on the role of ALCAM in cervical cancer available. METHODS: In this study, ALCAM expression was analysed by immunohistochemistry (IHC) in tissue samples of 233 patients with cervical cancer, among them 178 with complete follow-up information. In addition, soluble (s-)ALCAM was measured in sera of a subset of the included patients (n = 55) by enzyme-linked immunosorbent assay (ELISA). RESULTS: ALCAM overexpression was detected (immunoreactive score (IRS) 2-12) in 58.4% of the cervical cancer samples. The normal ectocervical or endocervical epithelium showed no ALCAM reactivity. In untreated patients, ALCAM overexpression in tumor tissue tended to be associated with shorter cancer-specific survival (CSS) and disease-free survival (DFS). Patients, whose tumor samples showed ALCAM overexpression receiving a cytotoxic therapy like radiotherapy or chemoradiation, however, had a favourable prognosis compared to those patients, whose cancers showed no or minimal ALCAM staining. This effect was particularly apparent in patients receiving chemoradiation where the CSS was significantly longer in patients with ALCAM-positive tumors (p = 0.038; cumulative incidence rates at 96 months 8%, 95% CI 0%-23%, and 26%, CI 3%-43% in ALCAM-positive and ALCAM-negative cases, respectively).Median preoperative s-ALCAM concentration in sera from tumor patients was 27.6 ng/ml (range 17.5-55.1 ng/ml, mean 28.9 ng/ml), serum levels did not correlate with intratumoral ALCAM expression. CONCLUSIONS: The data of our retrospective study suggest that the prognostic value of ALCAM expression in cervical carcinoma might be therapy-dependent, and that ALCAM might function as a predictive marker for the response to chemoradiation. This should be confirmed in further, prospective studies.


Subject(s)
Antigens, CD/metabolism , Carcinoma/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Fetal Proteins/metabolism , Neoplasm Proteins/metabolism , Uterine Cervical Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Antigens, CD/blood , Carcinoma/blood , Cell Adhesion Molecules, Neuronal/blood , Cohort Studies , Enzyme-Linked Immunosorbent Assay , Female , Fetal Proteins/blood , Humans , Immunohistochemistry , Middle Aged , Neoplasm Proteins/blood , Retrospective Studies , Uterine Cervical Neoplasms/blood , Young Adult
10.
J Surg Res ; 177(1): e15-20, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22482754

ABSTRACT

BACKGROUND: The activated leukocyte cell adhesion molecule (ALCAM, CD166) has been reported to be involved in tumorigenesis of colorectal cancer (CRC) and to function as a cancer stem cell marker. Controversial data exist regarding the prognostic power of ALCAM expression in CRC. Here, we evaluate the expression of ALCAM in a cohort of CRC patients and its usage as a prognostic marker for survival. MATERIALS AND METHODS: Tissue specimens from 299 patients with CRC treated between 1993 and 2006 were analyzed via ALCAM immunohistochemistry (clone MOG/07) using a tissue microarray. Results were correlated with clinical, histopathological, and patient survival data (Chi-square test, Kaplan-Meier analysis, and log-rank test, respectively). Multivariate analysis also was performed (Cox regression). RESULTS: ALCAM is expressed in most primary (76%) and secondary (62%) CRC lesions (P = 0.014). Immunohistochemistry revealed an inverse association with tumor grading (P = 0.002) but not with any other clinical or histopathological data. Kaplan-Meier survival analysis revealed a significant overall survival benefit in the group of ALCAM-positive patients (P = 0.019). Multivariate analysis showed that ALCAM is an independent positive prognostic marker for overall survival (P = 0.023). CONCLUSIONS: ALCAM expression is a positive prognostic marker for overall survival of CRC patients, and its detection might help to optimize the existing prognostic staging system. Elevated expression in higher differentiated tumors might indicate a potential role in the early steps of tumorigenesis, and its loss might be associated with reduced cellular adhesion, resulting in a higher metastatic potential of the tumor. Further studies must be conducted investigating these hypotheses.


Subject(s)
Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Colorectal Neoplasms/metabolism , Fetal Proteins/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Colon/pathology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Female , Humans , Kaplan-Meier Estimate , Liver/pathology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/secondary , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Middle Aged , Multivariate Analysis , Neoplasm Metastasis , Prognosis , Young Adult
11.
J Surg Res ; 173(2): 314-9, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21195422

ABSTRACT

BACKGROUND: L1 cell adhesion molecule (CD171) has been detected in different malignant tumors and is associated with unfavorable outcome. It thus represents a target for tumor diagnosis and therapy. In this study, we assessed L1 expression in more than 8000 normal human tissues and different types of tumors, both malignant and non-malignant, and neural and non-neural. MATERIALS AND METHODS: Tissue micro-arrays, including a multi-tumor-array of 128 different tumor types, up to 50 samples of each type (approximately 5500 different samples), arrays with approximately 3000 different prostate and 600 mesenchymal tumor samples, and a normal human tissue-array were analyzed by immunohistochemistry with a monoclonal antibody using immunoperoxidase staining. RESULTS: L1 expression was detected in tumors of neural and neural crest origin and other types of non-neural tumors, but not in those of epithelial origin. In normal human tissues, L1 was detected in skin basal cells and small blood vessels, most notably in the mature placenta and peripheral nerves. CONCLUSION: This first comprehensive study of the importance of L1 expression in human demonstrates strong L1 overexpression in tumors of neuroectodermal and neural crest origin and an expression in only very few normal human tissues. L1 thus is a potentially important therapeutic target, particularly with respect to malignant melanoma, gastrointestinal stromal tumor, neuroblastoma, and certain subtypes of non-neural tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Nervous System Neoplasms/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Humans , Tissue Array Analysis
12.
Int J Cancer ; 131(2): 396-405, 2012 Jul 15.
Article in English | MEDLINE | ID: mdl-21858815

ABSTRACT

The expression of the activated leukocyte cell adhesion molecule (ALCAM and CD166) is increased in various types of cancer. We aimed to evaluate its role as a prognostic marker for esophageal cancer (EC). We retrospectively analyzed ALCAM expression in 299 primary lesions, 147 lymph node and 46 distant metastases from EC patients, on a tissue microarray using immunohistochemistry. Bone marrow samples from representative cancer patients (n = 16), taken before primary surgery, were stained by double-immunofluorescence for ALCAM and cytokeratins (CK). Blood serum samples from 236 cancer patients and 127 controls were analyzed for serum ALCAM (s-ALCAM) by ELISA. The immunohistochemical analysis showed increased ALCAM expression in the majority of lesions (primary tumor 71%, lymph node 76% and distant metastases 80%). ALCAM expression was not associated with histopathological parameters except for tumor grading (p = 0.015). ALCAM-positive patients had significantly worse recurrence-free and overall survival (OS; p = 0.002). Disseminated tumor cells (DTC) in bone marrow showed two phenotypes, ALCAM+/CK+ (36%) and ALCAM-/CK+ (64%). Multivariate analysis revealed that ALCAM expression and elevated s-ALCAM serum values are powerful prognostic variables for OS in patients with EC (hazard ratio [HR] 3.987, 95% confidence interval [95%CI] 1.906-8.340, p < 0.001 and HR 1.915, 95%CI 1.021-3.592, p = 0.043). The results of our study provide preliminary evidence for the potential clinical utility of ALCAM as a prognostic biomarker for EC, which might be a basis for future clinical application. In addition, ALCAM expression in a subset of DTC of the bone marrow indicates a potential function in the metastatic cascade of EC.


Subject(s)
Activated-Leukocyte Cell Adhesion Molecule/blood , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/mortality , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Biomarkers, Tumor/metabolism , Esophageal Neoplasms/pathology , Female , Humans , Immunohistochemistry , Keratins/analysis , Male , Middle Aged , Neoplasm Grading , Prognosis , Protein Array Analysis , Retrospective Studies
13.
J Surg Res ; 170(2): 226-32, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21816425

ABSTRACT

BACKGROUND: Activated leukocyte cell adhesion molecule (ALCAM, CD166) is a cell membrane protein that is aberrantly expressed in different tumors, including pancreatic neuroendocrine tumors (PNET). The aim of this study was to determine the expression of ALCAM in PNET to learn more about the prevalence and clinical significance of ALCAM expression in PNET. METHODS: Primary tumors (n = 38) and corresponding lymph node (n = 5) and liver metastases (n = 9) of patients with PNET, treated at the University Medical Center Hamburg-Eppendorf between 1993 and 2006, were analyzed via ALCAM immunohistochemistry in a tissue microarray format. The results were correlated with clinical and histopathologic data, including the WHO classification of PNET. RESULTS: The majority of primary (74%) and secondary (50%) lesions of PNET showed strong ALCAM expression. Immunohistochemistry of primary tumors revealed an association between high ALCAM expression and the hormone production status of the tumor (P = 0.037), and an inverse correlation with metastasis status (P = 0.041) and tumor size (cut off level 2 cm, P = 0.013). Elevated ALCAM expression was a significant positive prognostic factor for recurrence-free (P = 0.002) and disease-specific survival (P = 0.009) in Kaplan-Meier survival analysis (log rank test). CONCLUSIONS: ALCAM is abundantly expressed in PNET and decreased expression is significantly associated with poor prognosis. ALCAM may be a potential marker for risk prediction in patients diagnosed with PNET. Further studies with larger patient collectives are required to validate the results of this study and investigate the functional role of ALCAM in PNET.


Subject(s)
Antigens, CD/metabolism , Biomarkers, Tumor/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Fetal Proteins/metabolism , Neuroendocrine Tumors/metabolism , Neuroendocrine Tumors/secondary , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Adult , Aged , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Liver Neoplasms/metabolism , Liver Neoplasms/secondary , Lymphatic Metastasis , Male , Middle Aged , Prognosis , Young Adult
14.
Cancer Res ; 71(14): 5009-19, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21632553

ABSTRACT

The incidence of pancreatic ductal adenocarcinoma (PDAC) nearly equals its mortality rate, partly because most PDACs are intrinsically chemoresistant and thus largely untreatable. It was found recently that chemoresistant PDAC cells overexpress the Notch-2 receptor and have undergone epithelial-mesenchymal transition (EMT). In this study, we show that these two phenotypes are interrelated by expression of Midkine (MK), a heparin-binding growth factor that is widely overexpressed in chemoresistant PDAC. Gemcitabine, the front-line chemotherapy used in PDAC treatment, induced MK expression in a dose-dependent manner, and its RNAi-mediated depletion was associated with sensitization to gemcitabine treatment. We identified an interaction between the Notch-2 receptor and MK in PDAC cells. MK-Notch-2 interaction activated Notch signaling, induced EMT, upregulated NF-κB, and increased chemoresistance. Taken together, our findings define an important pathway of chemoresistance in PDAC and suggest novel strategies for its clinical attack.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/metabolism , Cytokines/biosynthesis , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism , Receptor, Notch2/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cytokines/genetics , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Epithelial-Mesenchymal Transition , Fluorouracil/pharmacology , Humans , Midkine , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , RNA Interference , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Signal Transduction , Tumor Cells, Cultured , Gemcitabine
15.
BMC Cancer ; 11: 189:1-7, 2011 May 22.
Article in English | MEDLINE | ID: mdl-21600041

ABSTRACT

BACKGROUND: L1 cell adhesion molecule (CD171) is expressed in many malignant tumors and its expression correlates with unfavourable outcome. It thus represents a target for tumor diagnosis and therapy. An earlier study conducted by our group identified L1 expression levels in primary gastrointestinal stromal tumors (GIST) as a prognostic marker. The aim of the current study was to compare L1 serum levels of GIST patients with those of healthy controls and to determine whether levels of soluble L1 in sera could serve as a prognostic marker. METHODS: Using a sensitive enzyme-linked immunosorbent assay (ELISA), soluble L1 was measured in sera of 93 GIST patients und 151 healthy controls. Soluble L1 levels were then correlated with clinicopathological data. RESULTS: Median levels of soluble L1 were significantly higher (p < 0.001; Mann-Whitney U test) in sera of GIST patients compared to healthy individuals. Median soluble L1 levels were particularly elevated in patients with recurrence and relapse (p < 0.05; Mann Whitney U test). CONCLUSION: These results suggest that high soluble L1 levels predict poor prognosis and may thus be a promising tumor marker that can contribute to individualise therapy.


Subject(s)
Biomarkers, Tumor/blood , Gastrointestinal Stromal Tumors/diagnosis , Neural Cell Adhesion Molecule L1/blood , Adult , Aged , Aged, 80 and over , Case-Control Studies , Female , Gastrointestinal Stromal Tumors/blood , Gastrointestinal Stromal Tumors/mortality , Humans , Male , Middle Aged , Prognosis , Recurrence , Retrospective Studies , Survival Analysis
16.
J Surg Oncol ; 104(2): 140-5, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21520098

ABSTRACT

BACKGROUND: Chemokines and their receptors are known to play important roles in the tumorigenesis of many malignancies. The aim of this study was to evaluate the prognostic impact of the expression of the chemokine receptors CXCR4 and CXCR7 in patients with pancreatic adenocarcinoma (PAC). METHODS: Expression of CXCR4 and CXCR7 in specimens from 249 patients with PAC was evaluated by immunohistochemistry on a tissue microarray and matched with clinicopathological parameters and overall survival. RESULTS: Expression of CXCR4 was detected in 215 patients (86.4%) and CXCR7 in 47 patients (18.9%). No association between CXCR4 and CXCR7 expression was evident, although all the CXCR7 positive tumors were also CXCR4 positive. pT1/2 tumors showed a higher frequency of CXCR7 expression than pT3/4 tumors (P = 0.018), while more dedifferentiated tumors had elevated CXCR7 expression (P = 0.036). Overall and disease-free survival revealed no association with either CXCR4 or CXCR7 expression. CONCLUSION: CXCR7 is associated with tumor grade and inversely associated with tumor size and may play a potential role in tumor progression and differentiation. In contrast to previously reported data our results revealed no significant association between CXCR4 expression and clinical or pathological data.


Subject(s)
Adenocarcinoma/metabolism , Pancreatic Neoplasms/metabolism , Receptors, CXCR4/biosynthesis , Receptors, CXCR/biosynthesis , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry , Male , Pancreatic Neoplasms/mortality , Prognosis , Tissue Array Analysis
17.
Ann Surg Oncol ; 18(2): 559-65, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20589434

ABSTRACT

BACKGROUND: A previous study identified midkine (MK) expression in primary gastrointestinal stromal tumor (GIST) as a prognostic marker. The aim of the current study was to compare serum midkine (S-MK) concentrations of GIST patients with those of healthy controls and to determine if MK can serve as a prognostic serum marker for these patients. MATERIALS AND METHODS: S-MK concentrations were measured by enzyme-linked immunosorbent assay in GIST patients (n = 96) and healthy controls (n = 148). S-MK levels were then correlated with clinicopathological data and the administration of imatinib therapy. In addition, MK expression was evaluated in 39 surgically resected GIST and in 17 leiomyoma specimens on a tissue microarray. RESULTS: S-MK concentrations in GIST patients were significantly higher than in healthy controls: median (25th and 75th percentiles) S-MK concentration was 235 (139 and 376) pg/ml in the GIST patients and 99 (33 and 198) pg/ml in the controls (P < 0.001; Mann-Whitney U test). Significantly higher median S-MK concentrations were found in GIST with recurrence compared with those without (295 vs 230; P = 0.009). GIST patients with S-MK levels higher than 400 pg/ml showed a significantly worse recurrence-free survival (P = 0.026; log-rank test). Patients receiving imatinib therapy had decreased median S-MK concentrations compared with those who were not treated with imatinib (331 vs 201; P < 0.001). CONCLUSIONS: S-MK concentration is a potential marker for evaluating the progression and prognosis of GIST, especially during imatinib therapy. Further studies could focus on the role of midkine in the tumorigenesis of GIST and responsiveness toward imatinib therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/blood , Gastrointestinal Stromal Tumors/blood , Leiomyoma/blood , Neoplasm Recurrence, Local/blood , Nerve Growth Factors/blood , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Adult , Aged , Aged, 80 and over , Benzamides , Case-Control Studies , Disease Progression , Enzyme-Linked Immunosorbent Assay , Female , Gastrointestinal Stromal Tumors/drug therapy , Gastrointestinal Stromal Tumors/pathology , Humans , Imatinib Mesylate , Immunoenzyme Techniques , Leiomyoma/drug therapy , Leiomyoma/pathology , Male , Middle Aged , Midkine , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Prognosis , Survival Rate , Tissue Array Analysis
18.
J Mol Recognit ; 20(3): 185-96, 2007.
Article in English | MEDLINE | ID: mdl-17486692

ABSTRACT

Nogo-A is a potent inhibitor of axonal outgrowth in the central nervous system of adult mammals, where it is expressed as a membrane protein on oligodendrocytes and in myelin. Here we describe an attempt to identify linear peptide epitopes in its sequence that are responsible for the interaction either with the Nogo receptor (NgR) or with the neutralizing monoclonal antibody IN-1. Analysis of an array of immobilized overlapping 15 mer peptides covering the entire amino acid sequence of human Nogo-A (1192 residues) revealed a single epitope with prominent binding activity both towards the recombinant NgR and the IN-1 F(ab) fragment. Further truncation and substitution analysis yielded the minimal epitope sequence 'IKxLRRL' (x not equal to P), which occurs within the so-called Nogo66 region (residues 1054-1120) of Nogo-A. The bacterially produced Nogo66 fragment exhibited binding activity both for the recombinant NgR and for the IN-1 F(ab) fragment on the Western blot as well as in ELISA. Unexpectedly, the synthetic epitope peptide and the recombinant Nogo66 showed cross-reactivity with the 8-18C5 F(ab) fragment, which is directed against myelin oligodendrocyte glycoprotein (MOG) as a structurally unrelated target. On the other hand, the recombinant N-terminal domain of Nogo-A (residues 334-966) was shown to specifically interact on the Western blot and in an ELISA with the IN-1 F(ab) fragment but not with the recombinant NgR, which is in agreement with previous results. Hence, our data suggest that there is a distinct binding site for the Nogo receptor in the Nogo66 region of Nogo-A, whereas its interaction with NgR is less specific than anticipated before. Although there probably exists a non-linear epitope for the neutralizing antibody IN-1 in the N-terminal region of Nogo-A, which is likely to be accessible from outside the cell, a previously postulated second binding site for NgR in this region (called Nogo-A-24) remains elusive.


Subject(s)
Antibodies, Monoclonal/immunology , Combinatorial Chemistry Techniques , Epitope Mapping , Immunoglobulin Fab Fragments/immunology , Myelin Proteins/immunology , Receptors, Cell Surface/immunology , Binding Sites, Antibody/genetics , Binding Sites, Antibody/immunology , Blotting, Western , Cloning, Molecular , Enzyme-Linked Immunosorbent Assay , Epitopes , Fluorescence , GPI-Linked Proteins , Humans , Myelin Proteins/metabolism , Nogo Proteins , Nogo Receptor 1 , Peptide Fragments/immunology , Peptide Fragments/metabolism , Plasmids , Protein Engineering , Receptors, Cell Surface/metabolism
19.
FEBS J ; 274(10): 2603-13, 2007 May.
Article in English | MEDLINE | ID: mdl-17437522

ABSTRACT

Nogo-A is a physiologically relevant inhibitor of neuronal growth and regeneration in the myelin of the adult human central nervous system and has attracted considerable attention as a molecular target for the treatment of spinal cord injuries. To gain insight into the structural and functional properties of the large extramembrane region that is characteristic for the Nogo-A splice form of this member of the Reticulon family of membrane proteins, we cloned and expressed the region comprising residues 334-966 as a soluble homogeneous protein in the periplasm of Escherichia coli. SDS/PAGE, under nonreducing conditions, and a systematic substitution analysis of all six Cys residues of Nogo-A indicated that this domain forms two structural disulfide bonds among Cys residues 424, 464, 559 and 597, whereas the Cys residues at positions 699 and 912 seem to be dispensable for folding. The occurrence of a hot spot for host cell proteases and a limited proteolysis experiment suggest that the N-terminal region of Nogo-A up to residue 373 is structurally disordered. Although analytical gel permeation chromatography revealed a large apparent molecular size for the recombinant Nogo-A fragment, indicating oligomer formation, data from analytical ultracentrifugation and dynamic light scattering support a stable monomeric quaternary structure. Notably, the CD spectrum is indicative of a high content of random coil, such that Nogo-A exhibits--at least in part--features of a natively unfolded protein. Nevertheless, the protein fragment identified in this study, as well as its biochemical analysis, provide a promising starting point for future investigations to track down globular subdomains and functionally important regions as well as putative receptor-binding sites therein.


Subject(s)
Myelin Proteins/chemistry , Circular Dichroism , Cloning, Molecular , Disulfides/analysis , Electrophoresis, Polyacrylamide Gel , Escherichia coli/metabolism , Humans , Nogo Proteins , Protein Structure, Quaternary , Protein Structure, Tertiary , Recombinant Proteins/chemistry
20.
Proc Natl Acad Sci U S A ; 100(16): 9446-51, 2003 Aug 05.
Article in English | MEDLINE | ID: mdl-12874380

ABSTRACT

Multiple sclerosis is a chronic disease of the central nervous system (CNS) characterized by inflammation, demyelination, and axonal loss. The immunopathogenesis of demyelination in multiple sclerosis involves an autoantibody response to myelin oligodendrocyte glycoprotein (MOG), a type I transmembrane protein located at the surface of CNS myelin. Here we present the crystal structures of the extracellular domain of MOG (MOGIgd) at 1.45-A resolution and the complex of MOGIgd with the antigen-binding fragment (Fab) of the MOG-specific demyelinating monoclonal antibody 8-18C5 at 3.0-A resolution. MOGIgd adopts an IgV like fold with the A'GFCC'C" sheet harboring a cavity similar to the one used by the costimulatory molecule B7-2 to bind its ligand CTLA4. The antibody 8-18C5 binds to three loops located at the membrane-distal side of MOG with a surprisingly dominant contribution made by MOG residues 101-108 containing a strained loop that forms the upper edge of the putative ligand binding site. The sequence R101DHSYQEE108 is unique for MOG, whereas large parts of the remaining sequence are conserved in potentially tolerogenic MOG homologues expressed outside the immuno-privileged environment of the CNS. Strikingly, the only sequence identical to DHSYQEE was found in a Chlamydia trachomatis protein of unknown function, raising the possibility that Chlamydia infections may play a role in the MOG-specific autoimmune response in man. Our data provide the structural basis for the development of diagnostic and therapeutic strategies targeting the pathogenic autoantibody response to MOG.


Subject(s)
Antigens/chemistry , Myelin-Associated Glycoprotein/chemistry , Myelin-Associated Glycoprotein/immunology , Amino Acid Sequence , Animals , Binding Sites , Cell Membrane/metabolism , Chlamydia trachomatis/metabolism , Crystallography, X-Ray , Escherichia coli/metabolism , Ligands , Models, Molecular , Molecular Sequence Data , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Protein Conformation , Protein Folding , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...