Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
Add more filters

Publication year range
1.
Palliat Med ; 35(6): 1108-1117, 2021 06.
Article in English | MEDLINE | ID: mdl-33908288

ABSTRACT

BACKGROUND: International oncology societies recommend early palliative care. Specific models to integrate early palliative care efficiently into clinical practice are debated. The authors designed a study to look at the quantitative and qualitative outcomes of an early palliative care intervention in oncological care to decrease stress and improve quality of life. AIMS: To compare a single structured early palliative care intervention added to a usual oncology care in terms of distress and health-related quality of life at baseline compared to 6 months after enrollment. DESIGN: This multicenter randomized controlled trial (NCT01983956) enrolled adult patients with advanced cancer. Participants were either randomly assigned to usual oncology care alone or usual care plus a structured early palliative care intervention. SETTING/PARTICIPANTS: One hundred fifty adult patients with a variety of advanced cancer diagnoses were randomized. Seventy-four participants were in the intervention and 76 participants in the control group. The primary outcome was the change in patient distress assessed by the National Comprehensive Cancer Network distress thermometer at 6 months. Health-related quality of life, the secondary outcome, was assessed by the Functional Assessment of Cancer Therapy-General Questionnaire. RESULTS: The results showed no significant effect of the early palliative care intervention neither on patient distress nor on health-related quality of life. CONCLUSION: The addition of an early intervention to usual care for patients with advanced cancer did not improve distress or quality of life. Thus, patients may need more intensive early palliative care with continuous professional support to identify and address their palliative needs early.


Subject(s)
Hospice and Palliative Care Nursing , Neoplasms , Adult , Humans , Neoplasms/therapy , Palliative Care , Quality of Life
4.
Blood ; 119(21): 4963-70, 2012 May 24.
Article in English | MEDLINE | ID: mdl-22498738

ABSTRACT

The transcription factor PU.1 is a master regulator of myeloid differentiation and function. On the other hand, only scarce information is available on PU.1-regulated genes involved in cell survival. We now identified the glycolytic enzyme hexokinase 3 (HK3), a gene with cytoprotective functions, as transcriptional target of PU.1. Interestingly, HK3 expression is highly associated with the myeloid lineage and was significantly decreased in acute myeloid leukemia patients compared with normal granulocytes. Moreover, HK3 expression was significantly lower in acute promyelocytic leukemia (APL) compared with non-APL patient samples. In line with the observations in primary APL patient samples, we observed significantly higher HK3 expression during neutrophil differentiation of APL cell lines. Moreover, knocking down PU.1 impaired HK3 induction during neutrophil differentiation. In vivo binding of PU.1 and PML-RARA to the HK3 promoter was found, and PML-RARA attenuated PU.1 activation of the HK3 promoter. Next, inhibiting HK3 in APL cell lines resulted in significantly reduced neutrophil differentiation and viability compared with control cells. Our findings strongly suggest that HK3 is: (1) directly activated by PU.1, (2) repressed by PML-RARA, and (3) functionally involved in neutrophil differentiation and cell viability of APL cells.


Subject(s)
Cell Differentiation/genetics , Hexokinase/physiology , Leukemia, Promyelocytic, Acute/pathology , Neutrophils/physiology , Proto-Oncogene Proteins/physiology , Trans-Activators/physiology , Anthracyclines/pharmacology , Anthracyclines/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Differentiation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Cells, Cultured , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/physiology , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/physiology , Glycolysis/genetics , Hexokinase/genetics , Hexokinase/metabolism , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/genetics , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Oncogene Proteins, Fusion/physiology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Tretinoin/pharmacology , Tretinoin/therapeutic use
5.
Future Oncol ; 10(2): 211-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24490607

ABSTRACT

AIMS: ID1 is an important component of the MET-SRC signaling pathway, which is a regulator of cell migration and invasion. We hypothesized that the ALK/MET inhibitor crizotinib inhibits migration via MET-SRC-ID1, rather than ALK. MATERIALS & METHODS: We used ALK fusion-positive and -negative lung cancer cell lines; crizotinib, PHA-665752, and saracatinib, and stable transfection with shMET. We performed western blotting for p-ALK, ALK, p-MET, MET, p-SRC, SRC and ID1, and quantitative real-time PCR for ID1. RESULTS: Crizotinib decreased p-MET, p-SRC and ID1 levels in ALK- and or MET-positive cell lines and inhibited cell migration. Knockdown of MET was comparable with the effect of crizotinib. CONCLUSION: The effects of crizotinib on ID1 expression and cancer cell migration were associated with the presence of activated MET, rather than ALK fusion.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Movement/drug effects , Inhibitor of Differentiation Protein 1/metabolism , Lung Neoplasms/metabolism , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/metabolism , Pyrazoles/pharmacology , Pyridines/pharmacology , Cell Line, Tumor , Crizotinib , Humans , Inhibitor of Differentiation Protein 1/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Molecular Targeted Therapy , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-met/genetics
6.
Ther Umsch ; 71(12): 765-70, 2014 Dec.
Article in German | MEDLINE | ID: mdl-25447092

ABSTRACT

Modern oncology has much to offer, and the prognosis of cancer patients has undoubtedly improved over the last few decades. Nevertheless, medical and economic toxicities of modern cancer medicine demand that our many tools are viewed with a critical eye. Screening mammography and PSA screening for the detection of early breast, or prostate cancer, resp., are widely used. However, reduction of mortality from these disorders through early detection and treatment is quantitatively modest, whereas overdiagnosis of low-risk cancers and non-malignant conditions with these two methods is frequent. Cancer drug therapy increasingly uses predictive bio-markers to select patients (and their tumors) particularly likely to benefit from new drugs. Nevertheless palliative systemic treatment in oncology suffers from over-use of drugs with at times considerable toxicity and cost but little patient gain. It is still common practice to validate new cancer drugs in clinical trials using progression-free survival as a primary endpoint. PFS is easily measurable but often inadequate to discover true clinical benefit (i. e. prolonged overall survival and/or improved lasting quality of life). Licensing agencies such as the FDA or Swissmedic as well as clinical trialists are therefore challenged to review their criteria for the validation of new cancer drugs. Follow-up of cancer patients after treatment is widely practiced, often with "subscription series" for CT- or PET-scans and repetitive lab tests including tumor markers. Early detection of asymptomatic cancer relapse with these methods, however, is only warranted, if early re-treatment is proven to prolong survival, as cancer treatment in asymptomatic patients produces toxicity and costs that can only be justified if chances for long-term benefits are bettered.


Subject(s)
Antineoplastic Agents/therapeutic use , Early Detection of Cancer/methods , Neoplasms/diagnosis , Neoplasms/drug therapy , Patient Selection , Unnecessary Procedures , Antineoplastic Agents/adverse effects , Evidence-Based Medicine , Humans , Neoplasms/prevention & control , Primary Prevention/methods , Prognosis , Risk Assessment/methods , Treatment Outcome
7.
Biochem Biophys Res Commun ; 438(2): 283-8, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23891751

ABSTRACT

Autophagy is an intracellular bulk degradation process involved in cell survival upon stress induction, but also with a newly identified function in myeloid differentiation. The autophagy-related (ATG)8 protein family, including the GABARAP and LC3 subfamilies, is crucial for autophagosome biogenesis. In order to evaluate the significance of the GABARAPs in the pathogenesis of acute myeloid leukemia (AML), we compared their expression in primary AML patient samples, CD34(+) progenitor cells and in granulocytes from healthy donors. GABARAPL1 and GABARAPL2/GATE-16, but not GABARAP, were significantly downregulated in particular AML subtypes compared to normal granulocytes. Moreover, the expression of GABARAPL1 and GATE-16 was significantly induced during ATRA-induced neutrophil differentiation of acute promyelocytic leukemia cells (APL). Lastly, knocking down GABARAPL2/GATE-16 in APL cells attenuated neutrophil differentiation and decreased autophagic flux. In conclusion, low GABARAPL2/GATE-16 expression is associated with an immature myeloid leukemic phenotype and these proteins are necessary for neutrophil differentiation of APL cells.


Subject(s)
Adaptor Proteins, Signal Transducing/antagonists & inhibitors , Autophagy , Gene Expression Regulation, Leukemic , Leukemia, Promyelocytic, Acute/metabolism , Microfilament Proteins/antagonists & inhibitors , Neutrophils/cytology , Tretinoin/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Adolescent , Adult , Aged , Antigens, CD34/metabolism , Apoptosis Regulatory Proteins , Autophagy-Related Protein 8 Family , Cell Differentiation , Cell Line , Female , Humans , Leukemia, Promyelocytic, Acute/genetics , Male , Microfilament Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Middle Aged , Young Adult
8.
Biochem Biophys Res Commun ; 431(2): 264-9, 2013 Feb 08.
Article in English | MEDLINE | ID: mdl-23313490

ABSTRACT

MET, also known as hepatocyte growth factor receptor (HGFR), is a receptor tyrosine kinase with an important role, both in normal cellular function as well as in oncogenesis. In many cancer types, abnormal activation of MET is related to poor prognosis and various strategies to inhibit its function, including small molecule inhibitors, are currently in preclinical and clinical evaluation. Autophagy, a self-digesting recycling mechanism with cytoprotective functions, is induced by cellular stress. This process is also induced upon cytotoxic drug treatment of cancer cells and partially allows these cells to escape cell death. Thus, since autophagy protects different tumor cells from chemotherapy-induced cell death, current clinical trials aim at combining autophagy inhibitors with different cancer treatments. We found that in a gastric adenocarcinoma cell line GTL-16, where MET activity is deregulated due to receptor overexpression, two different MET inhibitors PHA665752 and EMD1214063 lead to cell death paralleled by the induction of autophagy. A combined treatment of MET inhibitors together with the autophagy inhibitor 3-MA or genetically impairing autophagy by knocking down the key autophagy gene ATG7 further decreased cell viability of gastric cancer cells. In general, we observed the induction of cytoprotective autophagy in MET expressing cells upon MET inhibition and a combination of MET and autophagy inhibition resulted in significantly decreased cell viability in gastric cancer cells.


Subject(s)
Adenocarcinoma/enzymology , Autophagy/drug effects , Drug Resistance, Neoplasm , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Stomach Neoplasms/enzymology , Adenine/analogs & derivatives , Adenine/pharmacology , Autophagy/genetics , Autophagy-Related Protein 7 , Cell Line, Tumor , Cell Survival , Gene Knockdown Techniques , Humans , Indoles/pharmacology , Pyridazines/pharmacology , Pyrimidines/pharmacology , Sulfones/pharmacology , Ubiquitin-Activating Enzymes/genetics
9.
Blood ; 118(23): 6037-42, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21951683

ABSTRACT

We report the results of a prospective, randomized phase 3 trial evaluating autologous peripheral blood stem cell transplantation (ASCT) versus intensive consolidation chemotherapy in newly diagnosed AML patients in complete remission (CR1). Patients with AML (16-60 years) in CR1 after 2 cycles of intensive chemotherapy and not eligible for allogeneic SCT were randomized between intensive chemotherapy with etoposide and mitoxantrone or ASCT ater high-dose cyclophosphamide and busulfan. Of patients randomized (chemotherapy, n = 259; ASCT, n = 258), more than 90% received their assigned treatment. The 2 groups were comparable with regard to prognostic factors. The ASCT group showed a markedly reduced relapse rate (58% vs 70%, P = .02) and better relapse-free survival at 5 years (38% vs 29%, P = .065, hazard ratio = 0.82; 95% confidence interval, 0.66-1.1) with nonrelapse mortality of 4% versus 1% in the chemotherapy arm (P = .02). Overall survival was similar (44% vs 41% at 5 years, P = .86) because of more opportunities for salvage with second-line chemotherapy and stem cell transplantation in patients relapsing on the chemotherapy arm. This large study shows a relapse advantage for ASCT as postremission therapy but similar survival because more relapsing patients on the chemotherapy arm were salvaged with a late transplantation for relapse. This trial is registered at www.trialregister.nl as #NTR230 and #NTR291.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation/methods , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/therapy , Adolescent , Adult , Antineoplastic Agents/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Agents, Phytogenic/therapeutic use , Busulfan/therapeutic use , Combined Modality Therapy/methods , Cyclophosphamide/therapeutic use , Etoposide/therapeutic use , Humans , Leukemia, Myeloid, Acute/mortality , Middle Aged , Mitoxantrone/therapeutic use , Prognosis , Prospective Studies , Remission Induction , Transplantation, Autologous , Treatment Outcome , Young Adult
10.
Environ Sci Technol ; 47(21): 12089-96, 2013.
Article in English | MEDLINE | ID: mdl-24099463

ABSTRACT

The spontaneous colonization of a bauxite residue (alumina refining tailings) deposit by local vegetation in Linden, Guyana, over 30 years, indicates that natural weathering processes can ameliorate tailings to the extent that it can support vegetation. Samples were collected from vegetated and unvegetated areas to investigate the relationships between bauxite residue properties and vegetation cover. Compared to unvegetated areas, bauxite residue in vegetated areas had lower pH (mean pH 7.9 vs 10.9), lower alkalinity (mean titratable alkalinity 0.4 vs 1.4 mol H(+) kg(-1)), lower electrical conductivity (mean EC 0.3 vs 2.1 mS cm(-1)), lower total Al (mean Al2O3 19.8 vs 25.8% wt) and Na (mean Na2O 0.9 vs 3.7% wt), and less sodalite and calcite. Accumulation of N, NH4(+), and organic C occurred under vegetation, demonstrating the capacity for plants to modify residue to suit their requirements as a soil-like growth medium. Aeolian redistribution of coarse grained tailings appeared to support vegetation establishment by providing a thin zone of enhanced drainage at the surface. Natural pedogenic processes may be supplemented by irrigation, enhanced drainage, and incorporation of sand and organic matter at other tailings deposits to accelerate the remediation process and achieve similar results in a shorter time frame.


Subject(s)
Aluminum Oxide , Environmental Restoration and Remediation , Plants , Soil , Aluminum/analysis , Aluminum Oxide/chemistry , Carbon/analysis , Electric Conductivity , Guyana , Hydrogen-Ion Concentration , Minerals/analysis , Nitrates/analysis , Nitrogen/analysis , Sodium/analysis
12.
Support Care Cancer ; 20(1): 141-7, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21234609

ABSTRACT

BACKGROUND: We assessed adherence to the European Society of Medical Oncology (ESMO)/Multinational Association of Supportive Care in Cancer recommendations for prophylaxis of chemotherapy-induced nausea and vomiting (CINV) at our institution. PATIENTS AND METHODS: The charts of 299 patients starting a new chemotherapy between November 2008 and April 2009 were reviewed. Baseline characteristics and prophylaxis of CINV during the first cycle were recorded, and adherence to ESMO recommendations was determined. Chi-square tests and logistic regression were used to test for predictors of adherence. RESULTS: Prophylaxis of acute CINV was not adherent in 39% of the patients: 39 of 54 patients with low emetogenic chemotherapy had a serotonin antagonist, and 24 of 100 with moderately emetogenic therapy had a neurokinin antagonist. Nevertheless, 71% of the patients treated with highly emetogenic therapy received the guideline-specified prescription. Prophylaxis of delayed CINV was not adherent in 89% of the patients: 101 of 125 patients with highly or moderately emetogenic single-day chemotherapy received a serotonin antagonist. Male gender (odds ratio (OR) 0.484, 95% confidence interval (CI) 0.291-0.806; P = 0.005) and hematologic neoplasia (OR 2.151, 95% CI 1.19-3.887; P = 0.011) were independent predictors of non-adherence. Age (OR 0.981, 95% CI 0.964-0.998; P = 0.029) and inpatient treatment (OR 0.457, 95% CI 0.25-0.836; P = 0.011) indicated a lower risk of non-adherence. CONCLUSION: Contrary to older studies reporting frequent omissions of corticosteroids, the current study demonstrated significant overuse of serotonin antagonists for prophylaxis of delayed CINV.


Subject(s)
Antiemetics/therapeutic use , Antineoplastic Agents/adverse effects , Nausea/prevention & control , Vomiting/prevention & control , Adult , Aged , Aged, 80 and over , Antiemetics/pharmacology , Antineoplastic Agents/therapeutic use , Chi-Square Distribution , Female , Guideline Adherence , Humans , Logistic Models , Male , Middle Aged , Nausea/chemically induced , Neoplasms/drug therapy , Neurokinin-1 Receptor Antagonists , Practice Guidelines as Topic , Retrospective Studies , Serotonin Antagonists/therapeutic use , Vomiting/chemically induced , Young Adult
13.
Ther Umsch ; 68(11): 618-30, 2011 Nov.
Article in German | MEDLINE | ID: mdl-22045524

ABSTRACT

The in vitro production of recombinant protein molecules has fostered a tremendous interest in their clinical application for treatment and support of cancer patients. Therapeutic proteins include monoclonal antibodies, interferons, and haematopoietic growth factors. Clinically established monoclonal antibodies include rituximab (targeting CD20-positive B-cell lymphomas), trastuzumab (active in HER-2 breast and gastric cancer), and bevacizumab (blocking tumor-induced angiogenesis through blockade of vascular-endothelial growth factor and its receptor). Interferons have lost much of their initial appeal, since equally or more effective treatments with more pleasant side effects have become available, for example in chronic myelogenous leukaemia or hairy cell leukaemia. The value of recombinant growth factors, notably granulocyte colony stimulating factor (G-CSF) and erythropoietin is rather in the field of supportive care than in targeted anti-cancer therapy. Adequately powered clinical phase III trials are essential to estimate the true therapeutic impact of these expensive compounds, with appropriate selection of clinically relevant endpoints and sufficient follow-up. Monoclonal antibodies, interferons, and growth factors must also, and increasingly so, be subjected to close scrutiny by appropriate cost-effectiveness analyses to ensure that their use results in good value for money. With these caveats and under the condition of their judicious clinical use, recombinant proteins have greatly enriched the therapeutic armamentarium in clinical oncology, and their importance is likely to grow even further.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Recombinant Proteins/therapeutic use , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/adverse effects , Erythropoietin/adverse effects , Erythropoietin/therapeutic use , Granulocyte Colony-Stimulating Factor/adverse effects , Granulocyte Colony-Stimulating Factor/therapeutic use , Humans , Intercellular Signaling Peptides and Proteins/adverse effects , Intercellular Signaling Peptides and Proteins/therapeutic use , Interferons/adverse effects , Interferons/therapeutic use , Leukemia/drug therapy , Lymphoma/drug therapy , Recombinant Proteins/adverse effects
14.
J Exp Med ; 199(10): 1343-54, 2004 May 17.
Article in English | MEDLINE | ID: mdl-15148334

ABSTRACT

Survivin has received great attention due to its expression in many human tumors and its potential as a therapeutic target in cancer. Survivin expression has been described to be cell cycle-dependent and restricted to the G2-M checkpoint, where it inhibits apoptosis in proliferating cells. In agreement with this current view, we found that survivin expression was high in immature neutrophils, which proliferate during differentiation. In contrast with immature cells, mature neutrophils contained only little or no survivin protein. Strikingly, these cells reexpressed survivin upon granulocyte/macrophage colony-stimulating factor (CSF) or granulocyte CSF stimulation in vitro and under inflammatory conditions in vivo. Moreover, survivin-deficient mature neutrophils were unable to increase their lifespan after survival factor exposure. Together, our findings demonstrate the following: (a) overexpression of survivin occurs in primary, even terminally differentiated cells and is not restricted to proliferating cells; and (b) survivin acts as an inhibitor of apoptosis protein in a cell cycle-independent manner. Therefore, survivin plays distinct and independent roles in the maintenance of the G2-M checkpoint and in apoptosis control, and its overexpression is not restricted to proliferating cells. These data provide new insights into the regulation and function of survivin and have important implications for the pathogenesis, diagnosis, and treatment of inflammatory diseases and cancer.


Subject(s)
Apoptosis/physiology , Microtubule-Associated Proteins/pharmacology , Neutrophils/cytology , Animals , Apoptosis/drug effects , Bone Marrow Cells/drug effects , Bone Marrow Cells/pathology , Cell Cycle/drug effects , Cell Differentiation , Cells, Cultured , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Inflammation/pathology , Inflammation/physiopathology , Inhibitor of Apoptosis Proteins , Mice , Mice, Knockout , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Neoplasm Proteins , Neoplasm Staging , Neoplasms/pathology , Neutrophils/drug effects , Neutrophils/immunology , Neutrophils/physiology , Recombinant Proteins , Survivin
15.
Br J Haematol ; 149(1): 55-64, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20096012

ABSTRACT

Acute promyelocytic leukaemia (APL) patients are successfully treated with all-trans retinoic acid (ATRA). However, concurrent chemotherapy is still necessary and less toxic therapeutic approaches are needed. Earlier studies suggested that in haematopoietic neoplasms, the green tea polyphenol epigallocatechin-3-gallate (EGCG) induces cell death without adversely affecting healthy cells. We aimed at deciphering the molecular mechanism of EGCG-induced cell death in acute myeloid leukaemia (AML). A significant increase of death-associated protein kinase 2 (DAPK2) levels was found in AML cells upon EGCG treatment paralleled by increased cell death that was significantly reduced upon silencing of DAPK2. Moreover, combined ATRA and EGCG treatment resulted in cooperative DAPK2 induction and potentiated differentiation. EGCG toxicity of primary AML blasts correlated with 67 kDa laminin receptor (67LR) expression. Pretreatment of AML cells with ATRA, causing downregulation of 67LR, rendered these cells resistant to EGCG-mediated cell death. In summary, it was found that (i) DAPK2 is essential for EGCG-induced cell death in AML cells, (ii) ATRA and EGCG cotreatment significantly boosted neutrophil differentiation, and 67LR expression correlates with susceptibility of AML cells to EGCG. We thus suggest that EGCG, by selectively targeting leukaemic cells, may improve differentiation therapies for APL and chemotherapy for other AML subtypes.


Subject(s)
Antineoplastic Agents/pharmacology , Catechin/analogs & derivatives , Leukemia, Myeloid, Acute/pathology , Neutrophils/drug effects , Tretinoin/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis Regulatory Proteins/physiology , Calcium-Calmodulin-Dependent Protein Kinases/physiology , Catechin/pharmacology , Cell Death/drug effects , Cell Differentiation/drug effects , Death-Associated Protein Kinases , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Gene Knockdown Techniques , Humans , Reverse Transcriptase Polymerase Chain Reaction/methods , Tumor Cells, Cultured , Up-Regulation/drug effects
16.
Lancet ; 373(9674): 1532-42, 2009 May 02.
Article in English | MEDLINE | ID: mdl-19410717

ABSTRACT

BACKGROUND: Erythropoiesis-stimulating agents reduce anaemia in patients with cancer and could improve their quality of life, but these drugs might increase mortality. We therefore did a meta-analysis of randomised controlled trials in which these drugs plus red blood cell transfusions were compared with transfusion alone for prophylaxis or treatment of anaemia in patients with cancer. METHODS: Data for patients treated with epoetin alfa, epoetin beta, or darbepoetin alfa were obtained and analysed by independent statisticians using fixed-effects and random-effects meta-analysis. Analyses were by intention to treat. Primary endpoints were mortality during the active study period and overall survival during the longest available follow-up, irrespective of anticancer treatment, and in patients given chemotherapy. Tests for interactions were used to identify differences in effects of erythropoiesis-stimulating agents on mortality across prespecified subgroups. FINDINGS: Data from a total of 13 933 patients with cancer in 53 trials were analysed. 1530 patients died during the active study period and 4993 overall. Erythropoiesis-stimulating agents increased mortality during the active study period (combined hazard ratio [cHR] 1.17, 95% CI 1.06-1.30) and worsened overall survival (1.06, 1.00-1.12), with little heterogeneity between trials (I(2) 0%, p=0.87 for mortality during the active study period, and I(2) 7.1%, p=0.33 for overall survival). 10 441 patients on chemotherapy were enrolled in 38 trials. The cHR for mortality during the active study period was 1.10 (0.98-1.24), and 1.04 (0.97-1.11) for overall survival. There was little evidence for a difference between trials of patients given different anticancer treatments (p for interaction=0.42). INTERPRETATION: Treatment with erythropoiesis-stimulating agents in patients with cancer increased mortality during active study periods and worsened overall survival. The increased risk of death associated with treatment with these drugs should be balanced against their benefits. FUNDING: German Federal Ministry of Education and Research, Medical Faculty of University of Cologne, and Oncosuisse (Switzerland).


Subject(s)
Anemia/drug therapy , Erythrocyte Transfusion , Hematinics/adverse effects , Neoplasms/mortality , Randomized Controlled Trials as Topic , Adolescent , Adult , Aged , Anemia/etiology , Antineoplastic Agents/therapeutic use , Effect Modifier, Epidemiologic , Erythropoietin/adverse effects , Female , Hematinics/therapeutic use , Humans , Linear Models , Male , Middle Aged , Neoplasms/complications , Neoplasms/drug therapy , Proportional Hazards Models , Recombinant Proteins , Research Design , Survival Rate , Treatment Outcome , Young Adult
17.
Ther Umsch ; 67(10): 491-5, 2010 Oct.
Article in German | MEDLINE | ID: mdl-20886453

ABSTRACT

The literature on malignant lymphomas has grown to an extent which is hardly palatable for general practitioners and non-lymphoma aficionados. Nevertheless some lymphomas exhibit typical clinical and radiological features which permit to suspect the correct histopathological diagnosis even before a pathology report becomes available. The present article points out such "case vignettes" of particular types of malignant lymphoma.


Subject(s)
Lymphoma/pathology , Adult , Aged , Biopsy , Burkitt Lymphoma/classification , Burkitt Lymphoma/diagnosis , Burkitt Lymphoma/pathology , Diagnosis, Differential , Enteropathy-Associated T-Cell Lymphoma/classification , Enteropathy-Associated T-Cell Lymphoma/diagnosis , Enteropathy-Associated T-Cell Lymphoma/pathology , Female , Hodgkin Disease/classification , Hodgkin Disease/diagnosis , Hodgkin Disease/pathology , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/classification , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma/classification , Lymphoma/diagnosis , Lymphoma, B-Cell/classification , Lymphoma, B-Cell/diagnosis , Lymphoma, B-Cell/pathology , Lymphoma, Follicular/classification , Lymphoma, Follicular/diagnosis , Lymphoma, Follicular/pathology , Lymphoma, Mantle-Cell/classification , Lymphoma, Mantle-Cell/diagnosis , Lymphoma, Mantle-Cell/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Tomography, X-Ray Computed , Waldenstrom Macroglobulinemia/classification , Waldenstrom Macroglobulinemia/diagnosis , Waldenstrom Macroglobulinemia/pathology , Young Adult
18.
Breast Cancer Res Treat ; 117(2): 319-24, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19137426

ABSTRACT

The current therapeutic strategy in breast cancer is to identify a target, such as estrogen receptor (ER) status, for tailoring treatments. We investigated the patterns of recurrence with respect to ER status for patients treated in two randomized trials with 25 years' median follow-up. In the ER-negative subpopulations most breast cancer events occurred within the first 5-7 years after randomization, while in the ER-positive subpopulations breast cancer events were spread through 10 years. In the ER-positive subpopulation, 1 year endocrine treatment alone significantly prolonged disease-free survival (DFS) with no additional benefit observed by adding 1 year of chemotherapy. In the small ER-negative subpopulation chemo-endocrine therapy had a significantly better DFS than endocrine alone or no treatment. Despite small numbers of patients, "old-fashioned" treatments, and competing causes of treatment failure, the value of ER status as a target for response to adjuvant treatment is evident through prolonged follow-up.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/metabolism , Receptors, Estrogen/biosynthesis , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Neoplasm Recurrence, Local/pathology , Randomized Controlled Trials as Topic , Treatment Outcome
19.
Breast Cancer Res Treat ; 116(3): 491-500, 2009 Aug.
Article in English | MEDLINE | ID: mdl-18953651

ABSTRACT

To compare the efficacy of chemoendocrine treatment with that of endocrine treatment (ET) alone for postmenopausal women with highly endocrine responsive breast cancer. In the International Breast Cancer Study Group (IBCSG) Trials VII and 12-93, postmenopausal women with node-positive, estrogen receptor (ER)-positive or ER-negative, operable breast cancer were randomized to receive either chemotherapy or endocrine therapy or combined chemoendocrine treatment. Results were analyzed overall in the cohort of 893 patients with endocrine-responsive disease, and according to prospectively defined categories of ER, age and nodal status. STEPP analyses assessed chemotherapy effect. The median follow-up was 13 years. Adding chemotherapy reduced the relative risk of a disease-free survival event by 19% (P = 0.02) compared with ET alone. STEPP analyses showed little effect of chemotherapy for tumors with high levels of ER expression (P = 0.07), or for the cohort with one positive node (P = 0.03). Chemotherapy significantly improves disease-free survival for postmenopausal women with endocrine-responsive breast cancer, but the magnitude of the effect is substantially attenuated if ER levels are high.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Endocrine System/drug effects , Lymph Nodes/pathology , Neoplasms, Hormone-Dependent/drug therapy , Receptors, Estrogen/metabolism , Breast Neoplasms/pathology , Chemotherapy, Adjuvant , Cyclophosphamide/administration & dosage , Female , Fluorouracil/administration & dosage , Humans , International Agencies , Lymph Nodes/drug effects , Lymphatic Metastasis , Methotrexate/administration & dosage , Middle Aged , Neoplasms, Hormone-Dependent/pathology , Postmenopause , Prognosis , Survival Rate , Tamoxifen/administration & dosage , Treatment Outcome
20.
Future Oncol ; 5(2): 245-57, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19284382

ABSTRACT

Tumor suppressor genes, such as p53, RB, the INK4-ARF family and PML, suppress malignant transformation by regulating cell cycle progression, ensuring the fidelity of DNA replication and chromosomal segregation, or by inducing apoptosis in response to potentially deleterious events. In myeloid leukemia, hematopoietic differentiation resulting from highly coordinated, stage-wise expression of myeloid transcription and soluble signaling factors is disrupted leading to a block in terminal differentiation and uncontrolled proliferation. This virtually always involves functional inactivation or genetic disruption of one or several tumor suppressor genes in order to circumvent their checkpoint control and apoptosis-inducing functions. Hence, reactivation of tumor suppressor gene function has therapeutic potential and can possibly enhance conventional cytotoxic chemotherapy. In this review, we focus on the role of different tumor suppressor genes in myeloid differentiation and leukemogenesis, and discuss implications for therapy.


Subject(s)
Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Genes, Tumor Suppressor/physiology , Hematopoietic Stem Cells/cytology , Leukemia, Myeloid/genetics , Hematopoietic Stem Cells/physiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL