Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Eur J Cancer ; 156: 175-186, 2021 10.
Article in English | MEDLINE | ID: mdl-34461420

ABSTRACT

BACKGROUND: Recent phase II-III trials of immuno(chemo)therapy before resection in locally advanced resectable non-small cell lung cancer (NSCLC) report high rates of pathological response and promising survival. However, primarily, patients who did not undergo resection were excluded from these studies. Moreover, there are no data on chemoradiotherapy (CRT) after immuno(chemo)therapy in patients who are primarily not amenable to CRT. We hypothesised that induction immuno(chemo)therapy may enable patients with NSCLC with a potentially curative stage (III-IVA), for whom primary curative treatment (either resection or CRT) is not possible for anatomical or functional reasons, to receive curative treatment. PATIENTS AND METHODS: We enrolled 35 patients with NSCLC with aforementioned characteristics into a prospective real-world trial of induction immuno(chemo)therapy followed by morphologic and metabolic reassessment and multidisciplinary board-guided curative treatment (resection [preferred] or CRT) or palliative therapy. The primary end-point was the proportion of patients receiving curative treatment. RESULTS: Thirty-two patients (91%) received curative treatment (11 resections and 21 CRT). 73% and 64% of patients who underwent resection had a major or complete pathological response, respectively. There were 14 recurrences: 2 (18%) in patients who underwent resection, 9 (43%) in patients who received CRT and 3 (100%) in patients who received palliative therapy (median follow-up 17 months). Eight tumour-related deaths occurred: 5 (24%) in patients who received CRT; and 3 (100%) in patients who received palliative therapy. There were no treatment-related deaths. CONCLUSIONS: In locally advanced or oligometastatic NSCLC without a primary curative option, induction immuno(chemo)therapy results in a high rate of curative treatment with promising early survival data. patients who underwent resection achieved a high rate of prognostically favourable pathological response.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Chemoradiotherapy , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Neoadjuvant Therapy , Nivolumab/therapeutic use , Palliative Care , Pneumonectomy , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/secondary , Chemoradiotherapy/adverse effects , Chemoradiotherapy/mortality , Chemotherapy, Adjuvant , Female , Germany , Humans , Immune Checkpoint Inhibitors/adverse effects , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Neoadjuvant Therapy/adverse effects , Neoadjuvant Therapy/mortality , Nivolumab/adverse effects , Pneumonectomy/adverse effects , Pneumonectomy/mortality , Prospective Studies , Time Factors , Treatment Outcome
2.
J Cancer Res Clin Oncol ; 144(10): 2059-2066, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30062488

ABSTRACT

BACKGROUND: Patients with non-small cell lung cancer (NSCLC) and a prior or synchronous second malignancy are generally excluded from clinical trials. Therefore, little is known on prevalence and prognosis of these patients. METHODS: 1252 patients diagnosed with NSCLC in our center from 2006 to 2017 were studied. Overall survival (OS) of patients with a prior or synchronous malignancy was compared to controls including case-control analysis. RESULTS: 158 patients (12.6%) had a prior malignancy. The most common sites were prostate (17%), breast (16%), gastrointestinal tract (12%), head and neck (11%), bladder (10%), and lung (8%). Compared to controls, patients with prior malignancy were older (71.3 vs. 67.5 years), but had otherwise better prognostic characteristics (stage I-III 63 vs. 53%). Survival was identical compared to controls [hazard ratio (HR) 1.017, CI 0.776-1.333]. A further 3.5% of patients had a synchronous malignancy including 34% prior lung cancer. Patients with a synchronous malignancy had an earlier stage (I-III 84%), and had longer median OS in unselected patients (38.6 vs. 16.2 months, p = 0.021). However, the case-control analysis showed similar OS [hazard ratio (HR) 0.899, CI 0.497-1.621]. CONCLUSIONS: Prior or synchronous second malignancies are common at diagnosis of NSCLC. The sites reflect the high proportion of smokers in the population. The earlier stage of NSCLC with a second malignancy might be attributed to chance finding of NSCLC during follow-up. The second malignancy does not affect OS of NSCLC. Therefore, the exclusion of patients with second malignancies from NSCLC trials should be reconsidered.


Subject(s)
Adenocarcinoma/mortality , Carcinoma, Large Cell/mortality , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/mortality , Lung Neoplasms/mortality , Neoplasms, Second Primary/epidemiology , Neoplasms, Second Primary/mortality , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Aged , Carcinoma, Large Cell/pathology , Carcinoma, Large Cell/therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/therapy , Combined Modality Therapy , Female , Follow-Up Studies , Humans , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Prognosis , Retrospective Studies , Survival Rate
3.
Oncotarget ; 9(29): 20781-20794, 2018 Apr 17.
Article in English | MEDLINE | ID: mdl-29755689

ABSTRACT

Malignant pleural mesothelioma (MPM) is a neoplasm with inferior prognosis and notorious chemotherapeutic resistance. Targeting aberrantly overexpressed kinases to cure MPM is a promising therapeutic strategy. Here, we examined ALK, MET and mTOR as potential therapeutic targets and determined the combinatorial efficacy of ALK and mTOR targeting on tumor cell growth in vivo. First, ALK overexpression, rearrangement and mutation were studied in primary MPM by qRT-PCR, FISH, immunohistochemistry and sequence analysis; mTOR and MET expression by qRT-PCR and immunohistochemistry. Overexpression of full-length ALK transcripts was observed in 25 (19.5%) of 128 primary MPM, of which ten expressed ALK protein. ALK overexpression was not associated with gene rearrangement, amplification or kinase-domain mutation. mTOR protein was detected in 28.7% MPM, co-expressed with ALK or MET in 5% and 15% MPM, respectively. The ALK/MET inhibitor crizotinib enhanced the anti-tumor effect of the mTOR-inhibitor rapamycin in a patient-derived MPM xenograft with co-activated ALK/mTOR: combined therapy achieved tumor shrinkage in 4/5 tumors and growth stagnation in one tumor. Treatment effects on proliferation, apoptosis, autophagy and pathway signaling were assessed using Ki-67 immunohistochemistry, TUNEL assay, LC3B immunofluorescence, and immunoblotting. Co-treatment significantly suppressed cell proliferation and induced autophagy and caspase-independent, necrotic cell death. Rapamycin/crizotinib simultaneously inhibited mTORC1 (evidenced by S6 kinase and RPS6 dephosphorylation) and ALK signaling (ALK, AKT, STAT3 dephosphorylation), and crizotinib suppressed the adverse AKT activation induced by rapamycin. In conclusion, co-treatment with rapamycin and crizotinib is effective in suppressing MPM tumor growth and should be further explored as a therapeutic alternative in mesothelioma.

4.
Oncotarget ; 8(44): 77897-77914, 2017 Sep 29.
Article in English | MEDLINE | ID: mdl-29100434

ABSTRACT

INTRODUCTION: Oncogenic driver mutations activating EGFR, ALK, or BRAF in NSCLC predict sensitivity to specific tyrosine-kinase inhibitors (TKIs). We provide data on prevalence, treatment and survival of driver-mutation positive NSCLC in a predominantly Caucasian population in routine clinical practice. PATIENTS AND METHODS: NSCLC patients diagnosed from 2006-2015 with an EGFR-test result were included (n=265). Testing for EGFR, ALK, or BRAF was performed if specific TKI therapy was considered. Case-control analyses of overall survival (OS) comparing driver-mutation positive and negative patients were performed. RESULTS: 44 sensitizing EGFR mutations (17%), 8 ALK translocations (7%, n=111) and 3 BRAF mutations (8%, n=39) were detected in adenocarcinoma or adenosquamous carcinoma. We did not find mutations in tumors without an adenocarcinoma-component. More than 90% of inoperable driver-mutation positive patients received TKI-therapy. Case-control analysis revealed improved OS of driver-mutation positive patients (39.6 vs. 19.4 months, HR 0.51). OS was improved in stage IV patients but not in stage I-III patients.OS of EGFR-TKI treated patients was similar for 1st and 2nd-line EGFR-TKI treatment. Patients not treated with EGFR-TKI had no benefit in OS. Re-biopsies obtained at progression revealed an EGFR-T790M mutation in 73% (n=11). These patients responded to the 3rd-generation EGFR-TKI osimertinib. DISCUSSION: Testing guided by predictive clinical parameters resulted in twice as high rates of mutation-positive patients than expected, and TKI treatment resulted in a strong long-term OS advantage. CONCLUSION: Testing for driver mutations is feasible in routine clinical practice, and identifies patients who benefit from TKI-therapy. OS compares favorably with OS in clinical studies.

5.
Int J Cancer ; 127(4): 873-80, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20013803

ABSTRACT

Apoptosis protease activating factor-1 (APAF-1), caspase-8 and caspase-9 are important factors in the execution of death signals. To study their prognostic influence in colon carcinoma, expression of APAF-1, caspase-8 and caspase-9 was determined by immunohistochemistry in normal colon mucosa (n = 8) and R0-resected stage II/III colon carcinomas (n >or= 124) using a semiquantitative score. Staining results were correlated with disease-free survival by Kaplan-Meier estimates, and multivariate Cox analyses were performed. In normal colon, APAF-1 and caspase-8 are most strongly expressed in the luminal surface epithelium, whereas caspase-9 is expressed all along the crypt axis. In colon carcinomas, there is considerable variability in the expression of these proapoptotic factors, although complete loss of caspase-8 and caspase-9 is rare. APAF-1 expression did not correlate with disease-free survival. Instead, both expression of caspase-9 and high-level expression of caspase-8 in a majority of tumor cells were significantly associated with adverse prognosis (p = 0.004 and p = 0.029, respectively). The influence of caspase-8 expression was mainly seen in patients with stage III colon carcinoma (p = 0.011), whereas the prognostic influence of caspase-9 expression was significant in stage II cases (p = 0.037) and just failed to be significant in stage III tumors (p = 0.0581). After adjusting for confounding factors in a multivariate Cox analysis, the effect of caspase-9 in predicting disease-free survival was confirmed (p = 0.003). Our data suggest that, in colon carcinomas, expression of caspase-8 and caspase-9 is significantly associated with poor survival. Caspase-9 may be an independent prognosticator in colon carcinoma.


Subject(s)
Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma/metabolism , Apoptotic Protease-Activating Factor 1/metabolism , Biomarkers, Tumor/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Colonic Neoplasms/metabolism , Adenocarcinoma/pathology , Adenocarcinoma, Mucinous/pathology , Aged , Colon/metabolism , Colonic Neoplasms/pathology , Female , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Staging , Prognosis , Prospective Studies , Survival Rate
6.
Dtsch Med Wochenschr ; 134(49): 2514, 2009 Dec.
Article in German | MEDLINE | ID: mdl-19941234

ABSTRACT

BACKGROUND: Hypothyroidism is a common endocrine disorder which is easily treatable by an appropriate thyroid hormone replacement therapy in the majority of patients. In some patients, hypothyroidism is refractory to oral levothyroxine substitution. Common causes of lack of response to levothyroxine replacement comprise non-compliance and impaired absorption. HISTORY: We report on a 32-year-old women who presented with persistent clinical and biochemical signs of hypothyroidism after thyroid surgery for Graves' disease despite high doses of levothyroxine replacement therapy. TREATMENT AND COURSE: An extensive evaluation for malabsorption syndromes proofed negative. Supervised absorption tests of two different levothyroxine preparations were normal. Pseudomalabsorption was presumed, though the patient continued to deny noncompliance. Supervised once weekly oral levothyroxine was advised. CONCLUSION: Non-compliance with medical therapy should be considered in patients with treatment-refractory hypothyroidism prior to initiation of an extensive evaluation for malabsorption syndromes. Supervised levothyroxine absorption test is a useful tool in the diagnostic workup for supposed pseudomalabsorption. In non-compliant patients, supervised once weekly levothyroxine replacement appears to be a safe and well-tolerated treatment option.


Subject(s)
Graves Disease/surgery , Hypothyroidism/etiology , Malabsorption Syndromes/diagnosis , Medication Adherence , Postoperative Complications/etiology , Thyroidectomy , Thyroxine/administration & dosage , Administration, Oral , Adult , Diagnosis, Differential , Dose-Response Relationship, Drug , Female , Humans , Hypothyroidism/blood , Hypothyroidism/drug therapy , Malabsorption Syndromes/blood , Postoperative Complications/blood , Postoperative Complications/drug therapy , Thyroxine/blood
7.
BMC Cancer ; 7: 58, 2007 Apr 02.
Article in English | MEDLINE | ID: mdl-17407575

ABSTRACT

BACKGROUND: The identification of genomic signatures of colorectal cancer for risk stratification requires the study of large series of cancer patients with an extensive clinical follow-up. Multicentric clinical studies represent an ideal source of well documented archived material for this type of analyses. METHODS: To verify if this material is technically suitable to perform matrix-CGH, we performed a pilot study using macrodissected 29 formalin-fixed, paraffin-embedded tissue samples collected within the framework of the EORTC-GI/PETACC-2 trial for colorectal cancer. The scientific aim was to identify prognostic genomic signatures differentiating locally restricted (UICC stages II-III) from systemically advanced (UICC stage IV) colorectal tumours. RESULTS: The majority of archived tissue samples collected in the different centers was suitable to perform matrix-CGH. 5/7 advanced tumours displayed 13q-gain and 18q-loss. In locally restricted tumours, only 6/12 tumours showed a gain on 13q and 7/12 tumours showed a loss on 18q. Interphase-FISH and high-resolution array-mapping of the gain on 13q confirmed the validity of the array-data and narrowed the chromosomal interval containing potential oncogenes. CONCLUSION: Archival, paraffin-embedded tissue samples collected in multicentric clinical trials are suitable for matrix-CGH analyses and allow the identification of prognostic signatures and aberrations harbouring potential new oncogenes.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Profiling , Nucleic Acid Hybridization , Clinical Trials as Topic , Dissection , Fixatives , Formaldehyde , Humans , In Situ Hybridization, Fluorescence , Paraffin Embedding , Pilot Projects , Prognosis , Risk Assessment , Specimen Handling
8.
Intensive Care Med ; 33(4): 694-702, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17237934

ABSTRACT

OBJECTIVE: We previously reported in healthy volunteers that a cantaloupe melon extract chemically combined with wheat gliadin (melon extract/gliadin) and containing SOD, catalase and residual glutathione peroxidase (GPx), protected against DNA strand-break damage induced by hyperbaric oxygen (HBO), a well-established model of DNA damage resulting from oxidative stress. Aortic cross-clamping is a typical example of ischemia/reperfusion injury-related oxidative stress, and therefore we investigated whether this melon extract/gliadin would also reduce DNA damage after aortic cross-clamping and reperfusion. DESIGN: Prospective, randomized, controlled experimental study. SETTING: Animal laboratory. PATIENTS AND PARTICIPANTS: 18 anesthetized, mechanically ventilated and instrumented pigs. INTERVENTIONS: After 14 days of oral administration of 1250 mg of the melon extract/gliadin (n=9) or vehicle (n=9), animals underwent 30 min of thoracic aortic cross-clamping and 4 h of reperfusion. MEASUREMENTS AND RESULTS: Before clamping, immediately before declamping, and at 2 and 4 h of reperfusion, we measured blood isoprostane (immunoassay) and malondialdehyde concentrations (fluorimetric thiobarbituric acid test), SOD, catalase and GPx activities (spectrophotometric kits), NO formation (nitrate+nitrite; chemoluminescence), DNA damage in whole blood samples and isolated lymphocytes exposed to hyperbaric oxygen (comet assay). Organ function was also evaluated. Kidney and spinal cord specimen were analysed for apoptosis (TUNEL assay). The melon extract/gliadin blunted the DNA damage, reduced spinal cord apoptosis and attenuated NO release, however, without any effect on lipid peroxidation and organ function. CONCLUSIONS: Pre-treatment with the oral melon extract/gliadin may be a therapeutic option to reduce oxidative cell injury affiliated with aortic cross-clamping.


Subject(s)
Apoptosis , DNA Damage , Gliadin/therapeutic use , Oxidative Stress/drug effects , Plant Extracts/therapeutic use , Reperfusion Injury/prevention & control , Animals , Comet Assay , Cucumis melo , Female , Hyperbaric Oxygenation/adverse effects , In Situ Nick-End Labeling , Male , Reperfusion Injury/etiology , Swine
9.
Verh Dtsch Ges Pathol ; 90: 159-67, 2006.
Article in German | MEDLINE | ID: mdl-17867593

ABSTRACT

In chronic pancreatitis (CP), fibrous replacement of exocrine tissue spares islets. There is local production of IFNgamma and death ligands by inflammatory cells as well as TGFbeta and TRAIL by pancreatic stellate cells (PSCs), along with functional death receptor neo-expression and apoptosis in exocrine but not in endocrine cells. Moreover, islets are strongly induced for TRAIL-receptor(R)-4 lacking a functional death domain. TRAIL-R4 signalling in T-cells induces NFkappaB transcription factors which activate anti-apoptotic programs. Whether TRAIL elicits this response in endocrine cells, we tested human insulinoma cell line CM and determined NFkappaB subunits transcripts and NFkappaB dependent inhibitor of apoptosis proteins (IAPs) in normal pancreas (NP) and CP. We treated CM with cytokines, determined TRAIL-R expression by flow cytometry, graded degree of fibrosis in CP specimens, microdissected epithelial compartments, performed real time PCRs for NFkappaB subunits transcripts, and immunohistochemistry for IKK-gamma, IkappaB-alpha, RelA, survivin, and cIAP1. In CM, TGFbeta/IFNgamma/TRAIL induced TRAIL-R4 surface expression. TRAIL/ IFNgamma, upregulated NFkappaB subunits and survivin while down-modulating 1kappaBalpha. NP epithelia had low RNA levels of NFkappaB subunits. These were increased in parenchymal areas of CP with severe fibrosis and most intensely in islets. The NFkappaB regulated proteins IkappaBalpha, survivin, and cIAP1 were found in corresponding sites, again, at highest levels in islets surrounded by fibrosis. In CP, islets not only evade immune attack by non-exposure of functional death receptors in presence of TRAIL-R4. They also neo-express NFkappaB subunits, survivin, and cIAP1. This apoptosis-inhibitory security program might be enforced by PSC-derived TRAIL.


Subject(s)
Genes, Intracisternal A-Particle/genetics , Islets of Langerhans/pathology , NF-kappa B/genetics , Pancreatitis/pathology , Chronic Disease , Epithelial Cells/cytology , Epithelial Cells/pathology , Gene Expression Regulation , Humans , Inhibitor of Apoptosis Proteins/genetics , TNF-Related Apoptosis-Inducing Ligand/genetics
10.
Shock ; 24(5): 476-81, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16247335

ABSTRACT

The aim of this study was to investigate the effects of intrarenal administration of the cyclooxygenase-2 inhibitor parecoxib during suprarenal aortic cross-clamping. In a prospective, controlled, blinded, randomized manner, 16 anesthetized and mechanically ventilated pigs were instrumented to measure systemic and right kidney hemodynamics, oxygen exchange, and metabolism. During 45 min of suprarenal aortic cross-clamping, animals received 40 mg of parecoxib (n = 8) or vehicle (n = 8) infused continuously into the right renal artery. Hemodynamic and metabolic data, right kidney venous blood, as well as urine samples were obtained before clamping, as well as before and 75 and 195 min after declamping. Clamping transiently increased mean arterial pressure in both groups. Systemic and renal blood flow did not differ between the pre- and postclamping measurements or between groups. Parecoxib attenuated the otherwise significant fall in right kidney creatinine clearance (controls: from 45 [7;111] to 17 [9;22] mL/min; parecoxib: from 39 [3;59] to 27 [11;45] mL/min, P = 0.039 and P = 0.297, respectively versus before clamping, P = 0.021 versus controls at 195 min) and prevented the impairment of renal lactate balance observed in the control group (controls: from 0.5 [-0.8;3.5] to 0.2 [-0.2;0.6] mumol/kg/min; parecoxib: from 0.6 [-1.0;2.0] to 0.4 [-1.2;0.6] mumol/kg/min, P = 0.038 and P = 0.285, respectively, versus before clamping). In conclusion, intrarenal parecoxib infusion beneficially influenced kidney function in this clinically relevant model of suprarenal aortic cross-clamping.


Subject(s)
Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Isoxazoles/pharmacology , Renal Circulation/drug effects , Angiography , Animals , Aorta/pathology , Aortic Aneurysm, Abdominal/surgery , Arachidonic Acid/metabolism , Creatinine/metabolism , Female , Hemodynamics , Isoxazoles/administration & dosage , Kidney/blood supply , Kidney/metabolism , Kidney/pathology , Male , Prospective Studies , Regional Blood Flow , Reperfusion Injury , Swine , Time Factors
11.
Lab Invest ; 85(10): 1263-75, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16127431

ABSTRACT

In advanced chronic pancreatitis (CP), islets are preserved even in the midst of scarring. We recently showed in CP local production of interferon (IFN)gamma, transforming growth factor (TGF)beta and death receptor ligand TRAIL (tumor necrosis factor-related apoptosis-inducing ligand), along with functional death receptor neoexpression and apoptosis in exocrine but not in endocrine cells. However, islets are strongly induced for TRAIL-receptor (R)-4 lacking the functional death domain. TRAIL-R4 signaling in T cells induces NFkappaB, which activates antiapoptotic programs. Here, we demonstrate that in insulinoma cells CM, TGFbeta/IFNgamma/TRAIL in combination induced TRAIL-R4 surface expression. TRAIL/IFNgamma upregulated NFkappaB subunits and its target gene survivin while downmodulating IkappaB alpha mRNA. RelA transcriptional activity increased upon stimulation with IFNgamma and IFNgamma/TRAIL. In situ, normal pancreatic epithelia had low mRNA levels of NFkappaB subunits. These were higher in parenchymal areas of CP with severe fibrosis and highest in islets. NFkappaB-regulated proteins IkappaB alpha, survivin and another apoptosis inhibitor, cIAP1, were found in corresponding sites, again at highest levels in islets surrounded by fibrosis. In conclusion, islets in CP not only evade immune attack by nonexposure of functional death receptors in the presence of TRAIL-R4 but also additionally neoexpress NFkappaB and its target genes, survivin and cIAP1, to protect themselves from apoptosis.


Subject(s)
Inhibitor of Apoptosis Proteins/biosynthesis , Islets of Langerhans/pathology , Microtubule-Associated Proteins/biosynthesis , NF-kappa B/biosynthesis , Neoplasm Proteins/biosynthesis , Pancreatitis, Chronic/pathology , Apoptosis , Apoptosis Regulatory Proteins/biosynthesis , Cell Line, Tumor , Epithelium/metabolism , Fibrosis , Gene Expression Regulation , Humans , I-kappa B Kinase/metabolism , Insulinoma , Interferon-gamma/physiology , Islets of Langerhans/metabolism , Membrane Glycoproteins/biosynthesis , Pancreatitis, Chronic/metabolism , Protein Subunits/biosynthesis , Receptors, Tumor Necrosis Factor/biosynthesis , Survivin , TNF-Related Apoptosis-Inducing Ligand , Transcription Factor RelA/biosynthesis , Transforming Growth Factor beta/physiology , Tumor Necrosis Factor Decoy Receptors , Tumor Necrosis Factor-alpha/biosynthesis , Up-Regulation
12.
Vitam Horm ; 67: 257-74, 2004.
Article in English | MEDLINE | ID: mdl-15110181

ABSTRACT

Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF family that can induce apoptosis when binding to either of two receptors bearing an intracellular death domain. The physiologic function of the TRAIL system, which also comprises three receptors not mediating a death signal has just begun to be elucidated. Expression of TRAIL, mostly upon stimulation by interferons, in different cytotoxic immune cells suggested it has a role as an important effector molecule in immune surveillance. In addition to its ability to induce apoptosis in transformed tumor cells, TRAIL has attracted attention for its possibly critical role in the defense against viral infection. Viruses may induce TRAIL expression in host and?or immune cells and sensitize host cells toward TRAIL-mediated apoptosis. On the other hand, viruses have evolved a variety of strategies to prevent TRAIL-mediated host cell death early in infection, which may contribute to allowing their replication and the spread of viral progeny. The knowledge of the molecular mechanisms leading to modification of TRAIL sensitivity in virus-host cell interactions may also impact upon future (virus-based) strategies to increase TRAIL sensitivity of tumor cells.


Subject(s)
Membrane Glycoproteins/physiology , Tumor Necrosis Factor-alpha/physiology , Virus Diseases , Animals , Apoptosis , Apoptosis Regulatory Proteins , Cells/virology , Humans , Immune Tolerance , Neoplasms , Receptors, Tumor Necrosis Factor , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand
13.
J Biol Chem ; 279(11): 10304-15, 2004 Mar 12.
Article in English | MEDLINE | ID: mdl-14701802

ABSTRACT

A tissue-protective effect of interleukin-11 (IL-11) for the intestinal mucosa has been postulated from animal models of inflammatory bowel disease (IBD). Despite the fact that the clinical usefulness of the anti-inflammatory effects of this cytokine is presently investigated in patients with IBD, there are no data available regarding the target cells of IL-11 action and the mechanisms of tissue protection within the human colonic mucosa. IL-11 responsiveness is restricted to cells that express the interleukin-11 receptor alpha-chain (IL-11Ralpha) and an additional signal-transducing subunit (gp130). In this study, we identified the target cells for IL-11 within the human colon with a new IL-11Ralpha monoclonal antibody and investigated the functional expression of the receptor and downstream effects of IL-11-induced signaling. Immunohistochemistry revealed expression of the IL-11Ralpha selectively on colonic epithelial cells. HT-29 and colonic epithelial cells (CEC) constitutively expressed IL-11Ralpha mRNA and protein. Co-expression of the signal-transducing subunit gp130 was also demonstrated. IL-11 induced signaling through triggering activation of the Jak-STAT pathway without inducing anti-inflammatory or proliferative effects in colonic epithelial cells. However, IL-11 stimulation resulted in a dose-dependent tyrosine phosphorylation of Akt, a decreased activation of caspase-9, and a reduced induction of apoptosis in cultured CEC. In HLA-B27 transgenic rats treated with IL-11, a reduction of apoptotic cell numbers was found. This study demonstrates functional expression of the IL-11Ralpha restricted on CEC within the human colonic mucosa. IL-11 induced signaling through triggering activation of the Jak-STAT pathway, without inducing anti-inflammatory or proliferative effects. The beneficial effects of IL-11 therapy are likely to be mediated by CEC via activation of the Akt-survival pathway, mediating antiapoptotic effects to support mucosal integrity.


Subject(s)
Apoptosis , Colon/cytology , Epithelial Cells/cytology , Receptors, Interleukin/biosynthesis , Receptors, Interleukin/chemistry , Animals , Animals, Genetically Modified , Antigens, CD/metabolism , Blotting, Northern , Blotting, Western , Caspase 9 , Caspases/metabolism , Cell Division , Cell Line , Cells, Cultured , Cytokine Receptor gp130 , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Interleukin-11/metabolism , Interleukin-11 Receptor alpha Subunit , Interleukin-8/metabolism , Janus Kinase 1 , Membrane Glycoproteins/metabolism , Mucous Membrane/pathology , Phosphorylation , Protein Binding , Protein-Tyrosine Kinases/metabolism , RNA, Messenger/metabolism , Rats , Receptors, Interleukin-11 , Reverse Transcriptase Polymerase Chain Reaction , STAT3 Transcription Factor , Time Factors , Trans-Activators/metabolism , Tyrosine/metabolism
14.
Shock ; 20(6): 511-6, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14625474

ABSTRACT

The cause for the high morbidity of blunt chest trauma is not fully understood. It is still unclear if and to what extent a second insult, e.g., apoptotic tissue damage initiated by the primary insult itself, may contribute to the development of serious complications. This study was done to elucidate whether a pulmonary contusion may induce programmed cell death. Sixty-four Wistar rats were evenly randomized to eight experimental groups: four sets were subjected to a standardized blast wave injury and sacrificed 6, 24, 48, and 72 h after the trauma; four groups served as controls for the same time points. Lung and liver samples were stained (H & E; TUNEL), and PMN infiltration was determined by myeloperoxidase (MPO) activity. Caspase 8 was analyzed by Western blot, and TNF-alpha plasma levels by ELISA. Postmortem examination revealed bilateral pulmonary contusion in trauma animals with higher (P < 0.05) numbers of apoptotic cells in lung but not in liver tissue as early as 6 h after the injury. This amount gradually increased and reached a maximum after 48 h: 6.8 +/- 1.1 apoptotic cells/hpf vs. 0.6 +/- 0.06 in controls. Chest trauma caused an increased expression of active caspase 8 in lung but not in liver tissue at 48 and 72 h. TNF-alpha plasma levels were not different. MPO activity in lung tissue of trauma animals increased (P < 0.05) after 6 h and peaked at 72 h. This study has provided the first evidence that apoptotic cell death in lung tissue is initiated following (experimental) pulmonary contusion. The exact mechanism remains, however, unclear and has to be elucidated further.


Subject(s)
Apoptosis , Thoracic Injuries/pathology , Animals , Blotting, Western , Caspase 8 , Caspases/metabolism , Enzyme-Linked Immunosorbent Assay , Hypoxia , In Situ Nick-End Labeling , Inflammation , Liver/metabolism , Liver/pathology , Lung/metabolism , Lung/pathology , Neutrophils/metabolism , Peroxidase/metabolism , Rats , Rats, Wistar , Time Factors , Tumor Necrosis Factor-alpha/metabolism
15.
Clin Cancer Res ; 9(11): 4116-24, 2003 Sep 15.
Article in English | MEDLINE | ID: mdl-14519634

ABSTRACT

PURPOSE: Despite adjuvant 5-fluorouracil (5-FU) therapy, approximately 30% of patients with International Union against Cancer stage II and III colorectal cancer develop recurrence. In this study, we determined the prognostic value of thymidylate synthase (TS) and dihydropyrimidine dehydrogenase (DPD) expression in colorectal cancer patients treated with adjuvant 5-FU. EXPERIMENTAL DESIGN: A real-time reverse transcription-PCR technique for quantitation of relative gene expression from paraffin-embedded specimen was established first. In a second step, archival paraffin-embedded primary tumor tissue of 309 patients who participated in adjuvant colorectal cancer trials was analyzed for TS and DPD mRNA expression. RESULTS: TS mRNA expression determined by real-time reverse transcription-PCR correlated with TS protein levels determined by TS immunoblot and immunohistochemistry in cultured colon cancer cell lines and paraffin-embedded primary tumor tissue. TS mRNA levels in fresh-frozen tissues also correlated with TS mRNA levels in corresponding paraffin sections. Among the patients receiving adjuvant 5-FU therapy, those with high TS survived longer than those with low TS, and in each TS subgroup, the ones with low DPD survived longer than the ones with high DPD levels. Multiple Cox regression analysis showed that besides tumor stage (P = 0.010), only the combination of TS and DPD expression turned out to be an independent prognostic factor for survival (P = 0.030). CONCLUSIONS: This suggests that TS and DPD quantitation may be helpful to evaluate prognosis of patients receiving adjuvant 5-FU and that patients with high TS and low DPD may benefit from adjuvant 5-FU chemotherapy.


Subject(s)
Colonic Neoplasms/drug therapy , Colonic Neoplasms/mortality , Dihydrouracil Dehydrogenase (NADP)/genetics , Fluorouracil/therapeutic use , RNA, Messenger/genetics , Rectal Neoplasms/drug therapy , Thymidylate Synthase/metabolism , Aged , Antimetabolites, Antineoplastic/therapeutic use , Chemotherapy, Adjuvant , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Rectal Neoplasms/mortality , Survival Analysis , Transcription, Genetic/genetics
16.
Lab Invest ; 83(6): 825-36, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12808117

ABSTRACT

TRAIL (TNF-related apoptosis-inducing ligand) induces apoptosis by cross-linking of the two TRAIL receptors that contain a death domain, TRAIL-R1 and TRAIL-R2. TRAIL-R3 and TRAIL-R4 are receptors that do not transmit an apoptotic signal. Our aim was to determine the expression of TRAIL and its receptors in normal pancreas and chronic pancreatitis. We applied real-time PCR, immunohisto(cyto)chemistry, and nick-end labeling of apoptosis. In normal pancreas, a minor subset of acinar cells coexpressed TRAIL-R2 and TRAIL-R4, whereas ductular epithelium and interstitial fibroblast-like cells (FLC) expressed TRAIL-R4. TRAIL-R1 and TRAIL-R3 were not detected in normal pancreas. In chronic pancreatitis, the exocrine epithelium strongly expressed TRAIL-R1, -R2, -R4, and, to a lesser extent, TRAIL-R3. Islets focally neoexpressed TRAIL-R1 and -R2 and intensely expressed TRAIL-R4. Changes in TRAIL receptor expression were most pronounced in areas of inflammatory infiltration and active fibrosis. In normal pancreas, expression of TRAIL was low on the mRNA level and undetectable on the protein level. In chronic pancreatitis, FLC in areas of active fibrosis expressed TRAIL. In addition, apoptosis were most numerous in these areas. We show that these FLC are pancreatic stellate cells. Pancreatic stellate cells express TRAIL in vivo and in vitro, and TRAIL expression is enhanced by IFN-gamma. Our findings indicate that the TRAIL/TRAIL receptor system is likely to be involved in chronic pancreatitis and suggest that pancreatic stellate cells may directly contribute to acinar regression by inducing apoptosis of parenchymal cells in a TRAIL-dependent manner.


Subject(s)
Membrane Glycoproteins/genetics , Pancreatitis/pathology , Pancreatitis/physiopathology , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/genetics , Apoptosis , Apoptosis Regulatory Proteins , Base Sequence , Cells, Cultured , Chronic Disease , DNA Primers , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/pathology , Epithelial Cells/physiology , Fibroblasts/pathology , Fibrosis , GPI-Linked Proteins , Humans , In Situ Nick-End Labeling , Islets of Langerhans/pathology , Islets of Langerhans/physiopathology , Pancreatectomy , Pancreatitis/genetics , RNA, Messenger/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor, Member 10c , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand , Tumor Necrosis Factor Decoy Receptors
17.
Clin Cancer Res ; 8(12): 3734-40, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12473583

ABSTRACT

PURPOSE: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selectively induces apoptosis in cancer cells and may be involved in protection from metastases. TRAIL receptor (TRAIL-R) 1 and TRAIL-R2, but not TRAIL-R3 and TRAIL-R4, mediate apoptosis. We examined the expression of TRAIL and its receptors in normal and neoplastic colon epithelium, and studied its correlation with prognosis in colon cancer. EXPERIMENTAL DESIGN: Immunohistochemistry was performed on normal colon mucosa (n = 10), colon adenomas (n = 20), and R0-resected Unio Internationale Contra Cancrum stage II/III colon carcinomas (n = 129). Disease-free survival was examined by Kaplan-Meier estimates and the log-rank test. Prognostic factors were determined by multivariate Cox-analysis. RESULTS: In normal colon mucosa, TRAIL and TRAIL-R2 were expressed mostly in the surface epithelium, whereas TRAIL-R1 and TRAIL-R4 were detected all along the crypt axis. In adenomas, this expression pattern was mostly retained, although some adenomas also neoexpressed TRAIL-R3. In carcinomas, the expression of TRAIL and TRAIL receptors was much more variable. TRAIL, TRAIL-R2, TRAIL-R3, and TRAIL-R4 expression did not correlate statistically with disease-free survival (multivariate analysis: P = 0.54, P = 0.67, P = 0.45, and P = 0.69, respectively), but TRAIL-R1 expression was significantly associated with disease-free survival in colon cancer (multivariate analysis: P = 0.003). CONCLUSIONS: TRAIL-R1 is an independent prognostic factor in R0-resected Unio Internationale Contra Cancrum stage II/III colon cancer.


Subject(s)
Colon/metabolism , Colonic Neoplasms/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma, Mucinous/metabolism , Adenocarcinoma, Mucinous/pathology , Adult , Aged , Apoptosis , Case-Control Studies , Cell Differentiation , Colonic Neoplasms/pathology , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , GPI-Linked Proteins , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Staging , Prognosis , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor, Member 10c , Survival Rate , Tumor Necrosis Factor Decoy Receptors
18.
Cell Commun Adhes ; 9(1): 1-14, 2002.
Article in English | MEDLINE | ID: mdl-12200961

ABSTRACT

Pleural cavities are lined by a polarized monolayer of mesothelial cells (MC). During pleuritis, MC are shed into effusions, and pleural obstruction may occur. Integrins are cell surface receptors mediating interactions with extracellular matrix (ECM) proteins. The distribution of beta 1-, beta 3-, beta 4-integrins and fibronectin and laminin in normal and chronically inflamed pleura and in/on MC from pleural effusions was examined by immunomorphology and flow cytometry. Adhesion assays of MC to fibronectin and laminin were performed. In situ, resting MC expressed beta 1-, beta 3-, and beta 4-, and alpha v-subunits. Activated MC were beta 1- and alpha v-positive and also expressed alpha 3 and alpha 6; beta 4 was confined to the basal surface of MC; beta 3 was absent. Floating MC from effusions neoexpressed alpha 5 and reexpressed beta 3. In vitro, MC surface expressed beta 1, beta 3, alpha 3, alpha 5, alpha 6, alpha v, and also alpha 1 and alpha 2. In normal pleura, fibronectin and laminin were components of the basement membrane. In pleuritis, the basement membrane was desintegrated. Instead, newly formed fibronectin/laminin containing fibrils extended into the submesothelial connective tissue. Floating MC freshly isolated from effusions carried fibronectin and laminin on their surface and showed specific binding to these ECM proteins. Binding was blocked by anti-beta 1 or anti-alpha 5 and anti-alpha 6 antibodies, respectively. MC incubated with fibronectin showed a clear shift to the S phase, while laminin had no effect. In conclusion, activated and detached MC progressively enrich their integrin repertoire. By capturing soluble fibronectin and laminin and by matrix-mediated bridging, readhering MC may contribute to pleural obstruction. Further, soluble fibronectin bound to alpha 5 beta 1 might be life-sustaining for floating MC by driving cells into cell cycle.


Subject(s)
Epithelium/metabolism , Fibronectins/metabolism , Integrins/biosynthesis , Laminin/metabolism , Antigens, CD/biosynthesis , Cell Adhesion , Cell Cycle , Cells, Cultured , Flow Cytometry , Humans , Immunohistochemistry , Integrin alphaV , Integrin beta1/biosynthesis , Integrin beta3 , Integrin beta4 , Lung/pathology , Platelet Membrane Glycoproteins/biosynthesis , Pleural Diseases/metabolism
19.
J Gastrointest Surg ; 6(3): 331-7, 2002.
Article in English | MEDLINE | ID: mdl-12022983

ABSTRACT

Patients with International Union Against Cancer (UICC) stage IIb and III colon cancer and stage II and III rectal cancer may receive adjuvant chemotherapy with 5-fluorouracil (5-FU). High levels of thymidylate synthase (TS) and dihydropyrimidine dehydrogenase (DPD) have been associated with resistance to 5-FU in advanced colorectal cancer. The aim of this study was to investigate the association of TS and DPD mRNA levels with recurrence-free survival in patients with colorectal cancer who are receiving adjuvant 5-FU-based chemotherapy. TS and DPD mRNA quantitation was retrospectively performed in primary colorectal cancer specimens from patients receiving adjuvant 5-FU using a reverse transcription- polymerase chain reaction technique. The median TS mRNA level in patients with a recurrence (n = 142) was 0.68, and in patients without a recurrence (n = 206) the median level was 0.80 (P < 0.01). Patients with a recurrence who had a low TS level (TS < or = 0.9; n = 102) had a median recurrence-free survival of 18 months (range 3.0 to 54 months), and those with a high TS level (TS > 0.9; n = 40) had a median recurrence-free survival of 11 months (range 1.7 to 53 months; P = 0.0024). There was no difference in the median recurrence-free survival of patients with low and high DPD mRNA levels. The TS mRNA level may be a useful marker to predict the time to recurrence in patients with colorectal cancer who are receiving adjuvant 5-FU treatment.


Subject(s)
Colonic Neoplasms/enzymology , Colonic Neoplasms/mortality , Neoplasm Recurrence, Local/enzymology , Oxidoreductases/metabolism , Rectal Neoplasms/enzymology , Rectal Neoplasms/mortality , Thymidylate Synthase/metabolism , Aged , Antimetabolites, Antineoplastic/therapeutic use , Chemotherapy, Adjuvant , Colonic Neoplasms/drug therapy , Colonic Neoplasms/pathology , Dihydrouracil Dehydrogenase (NADP) , Disease-Free Survival , Female , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , RNA, Messenger/metabolism , Rectal Neoplasms/drug therapy , Rectal Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction
20.
Gastroenterology ; 122(3): 659-66, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11874999

ABSTRACT

BACKGROUND & AIMS: Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a member of the tumor necrosis factor family and induces apoptosis by cross-linking either of the 2 TRAIL receptors containing a death domain (TRAIL-R1 or TRAIL-R2). TRAIL-R3 and TRAIL-R4 are receptors that do not transmit an apoptotic signal. The aim of this study was to investigate the expression and function of TRAIL and its receptors in normal colonic epithelium. METHODS: TRAIL and TRAIL receptor expression was studied by reverse-transcriptase polymerase chain reaction and immunohistochemistry. TRAIL sensitivity of epithelial cells was determined in vitro. RESULTS: Normal colonic epithelial cells express TRAIL, TRAIL-R1, TRAIL-R2, and TRAIL-R4. Interestingly, TRAIL and TRAIL-R2 are coexpressed mostly in the luminal surface epithelium. Despite the expression of apoptosis-mediating TRAIL receptors, the normal colonic crypt epithelium is completely resistant to TRAIL-induced apoptosis in vitro. Using an infection model with restricted human cytomegalovirus gene expression or productive adenovirus infection in the colon carcinoma cell line Caco-2, we show that TRAIL sensitivity of colonic epithelial cells is induced on virus infection along with an up-regulation of TRAIL-R1 and TRAIL-R2 on the cell surface. CONCLUSIONS: We conclude that the TRAIL system may play a role in the early elimination of virus-infected epithelial cells in the normal gut.


Subject(s)
Adenoviridae Infections/pathology , Colon/pathology , Cytomegalovirus Infections/pathology , Membrane Glycoproteins/genetics , Receptors, Tumor Necrosis Factor/genetics , Tumor Necrosis Factor-alpha/genetics , Apoptosis/physiology , Apoptosis Regulatory Proteins , Caco-2 Cells , Colon/chemistry , Colon/virology , Gene Expression/physiology , Humans , Immunity, Innate , Immunohistochemistry , Intestinal Mucosa/chemistry , Intestinal Mucosa/pathology , Intestinal Mucosa/virology , Membrane Glycoproteins/analysis , Receptors, TNF-Related Apoptosis-Inducing Ligand , Receptors, Tumor Necrosis Factor/analysis , TNF-Related Apoptosis-Inducing Ligand , Transfection , Tumor Necrosis Factor-alpha/analysis , Up-Regulation/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...