Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 245
Filter
2.
Leukemia ; 31(3): 602-613, 2017 03.
Article in English | MEDLINE | ID: mdl-27538486

ABSTRACT

Apart from its unique histopathological appearance with rare tumor cells embedded in an inflammatory background of bystander cells, classical Hodgkin lymphoma (cHL) is characterized by an unusual activation of a broad range of signaling pathways involved in cellular activation. This includes constitutive high-level activity of nuclear factor-κB (NF-κB), Janus kinase/signal transducer and activator of transcription (JAK/STAT), activator protein-1 (AP-1) and interferon regulatory factor (IRF) transcription factors (TFs) that are physiologically only transiently activated. Here, we demonstrate that inactivation of the putative ubiquitin E3-ligase PDLIM2 contributes to this TF activation. PDLIM2 expression is lost at the mRNA and protein levels in the majority of cHL cell lines and Hodgkin and Reed-Sternberg (HRS) cells of nearly all cHL primary samples. This loss is associated with PDLIM2 genomic alterations, promoter methylation and altered splicing. Reconstitution of PDLIM2 in HRS cell lines inhibits proliferation, blocks NF-κB transcriptional activity and contributes to cHL-specific gene expression. In non-Hodgkin B-cell lines, small interfering RNA-mediated PDLIM2 knockdown results in superactivation of TFs NF-κB and AP-1 following phorbol 12-myristate 13-acetate (PMA) stimulation. Furthermore, expression of PDLIM2 is lost in anaplastic large cell lymphoma (ALCL) that shares key biological aspects with cHL. We conclude that inactivation of PDLIM2 is a recurrent finding in cHL and ALCL, promotes activation of inflammatory signaling pathways and thereby contributes to their pathogenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Hodgkin Disease/genetics , LIM Domain Proteins/genetics , Lymphoma, Large-Cell, Anaplastic/genetics , Microfilament Proteins/genetics , Base Sequence , Cell Line, Tumor , Cluster Analysis , DNA Methylation , Enzyme Activation , Female , Gene Silencing , Genetic Loci , Hodgkin Disease/metabolism , Humans , LIM Domain Proteins/metabolism , Lymphoma, Large-Cell, Anaplastic/metabolism , Male , Microfilament Proteins/metabolism , Mutation , NF-kappa B/metabolism , Promoter Regions, Genetic , Proteolysis , RNA Splice Sites , Transcription Factors , Ubiquitin-Protein Ligases
3.
Leukemia ; 31(7): 1547-1554, 2017 07.
Article in English | MEDLINE | ID: mdl-27890934

ABSTRACT

Recurrent mutations within EGR2 were recently reported in advanced-stage chronic lymphocytic leukemia (CLL) patients and associated with a worse outcome. To study their prognostic impact, 2403 CLL patients were examined for mutations in the EGR2 hotspot region including a screening (n=1283) and two validation cohorts (UK CLL4 trial patients, n=366; CLL Research Consortium (CRC) patients, n=490). Targeted deep-sequencing of 27 known/postulated CLL driver genes was also performed in 38 EGR2-mutated patients to assess concurrent mutations. EGR2 mutations were detected in 91/2403 (3.8%) investigated cases, and associated with younger age at diagnosis, advanced clinical stage, high CD38 expression and unmutated IGHV genes. EGR2-mutated patients frequently carried ATM lesions (42%), TP53 aberrations (18%) and NOTCH1/FBXW7 mutations (16%). EGR2 mutations independently predicted shorter time-to-first-treatment (TTFT) and overall survival (OS) in the screening cohort; they were confirmed associated with reduced TTFT and OS in the CRC cohort and independently predicted short OS from randomization in the UK CLL4 cohort. A particularly dismal outcome was observed among EGR2-mutated patients who also carried TP53 aberrations. In summary, EGR2 mutations were independently associated with an unfavorable prognosis, comparable to CLL patients carrying TP53 aberrations, suggesting that EGR2-mutated patients represent a new patient subgroup with very poor outcome.


Subject(s)
Early Growth Response Protein 2/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Mutation , Adult , Aged , Female , Genes, p53 , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/classification , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Male , Middle Aged , Proportional Hazards Models
5.
Rheumatol Int ; 36(11): 1563-1568, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27522225

ABSTRACT

Issues of fertility and pregnancy require special attention in the long-term care of patients with autoimmune diseases (AD), who are candidates for haematopoietic stem cell transplantation (HSCT). In this single-centre observational study, we report fertility status and pregnancy outcomes in 15 patients (11 female and 4 male) after immunoablation with cyclophosphamide, antithymocyte globulin and autologous CD34+-selected HSCT for severe, refractory AD. The median follow-up after HSCT was 12 years (range 2-16 years). Impaired fertility was observed in six patients (five females and one male) before HSCT based on sexual hormone measurements. Higher age and cumulative cyclophosphamide dosage before HSCT correlated with fertility impairment. Median serum level of follicle-stimulating hormone (FSH) was significantly higher in female patients at 1 year after HSCT compared to baseline values, but premature ovarian failure developed in only one patient. Four women had five pregnancies and six healthy offsprings during follow-up, and no miscarriages were observed. The mothers were in treatment-free remissions during conception. No peripartal flare of their AD occurred. Although AD patients undergoing HSCT are at risk of developing infertility, pre-HSCT treatment and patients' age seem to have higher impact on long-term fertility status than HSCT itself. HSCT offers the opportunity to conceive during treatment-free remissions with favourable pregnancy outcomes.


Subject(s)
Autoimmune Diseases/therapy , Cyclophosphamide/adverse effects , Fertility/physiology , Hematopoietic Stem Cell Transplantation/adverse effects , Immunosuppressive Agents/adverse effects , Adult , Autoimmune Diseases/complications , Cyclophosphamide/therapeutic use , Female , Follicle Stimulating Hormone/blood , Follow-Up Studies , Humans , Immunosuppressive Agents/therapeutic use , Infertility/blood , Infertility/etiology , Male , Middle Aged , Pregnancy , Pregnancy Outcome , Pregnancy Rate , Primary Ovarian Insufficiency/blood , Primary Ovarian Insufficiency/etiology , Young Adult
6.
Cancer Gene Ther ; 23(6): 162-7, 2016 06.
Article in English | MEDLINE | ID: mdl-27056671

ABSTRACT

Chemokines are key regulators of both innate and adaptive immune responses. CCL4 (macrophage inflammatory protein-1ß, MIP-1ß) is a CC chemokine that has a broad spectrum of target cells including immature dendritic cells, which express the cognate receptor CCR5. We asked whether a plasmid encoding CCL4 is able to improve tumor protection and immune responses in a Her2/neu+ mouse tumor model. Balb/c mice were immunized twice intramuscularly with plasmid DNA on days 1 and 15. On day 25, a tumor challenge was performed with 2 × 10(5) syngeneic Her2/neu+ D2F2/E2 tumor cells. Different groups of mice were vaccinated with pDNA(Her2/neu) plus pDNA(CCL4), pDNA(Her2/neu), pDNA(CCL4) or mock vector alone. Our results show that CCL4 is able to (i) improve tumor protection and (ii) augment a TH1-polarized immune response against Her2/neu. Although Her2/neu-specific humoral and T-cell immune responses were comparable with that induced in previous studies using CCL19 or CCL21 as adjuvants, tumor protection conferred by CCL4 was inferior. Whether this is due to a different spectrum of (innate) immune cells, remains to be clarified. However, combination of CCL19/21 with CCL4 might be a reasonable approach in the future, particularly for DNA vaccination in Her2/neu+ breast cancer in the situation of minimal residual disease.


Subject(s)
Adjuvants, Immunologic , Cancer Vaccines/immunology , Chemokine CCL4 , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Receptor, ErbB-2/genetics , Vaccines, DNA/immunology , Animals , Cancer Vaccines/genetics , Cell Line , Chemokine CCL4/genetics , Disease Models, Animal , Female , Gene Expression , Gene Order , Humans , Immunization , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/therapy , Mice , Plasmids/genetics , Tumor Burden , Vaccines, DNA/genetics
7.
Radiat Oncol ; 10: 266, 2015 Dec 30.
Article in English | MEDLINE | ID: mdl-26715553

ABSTRACT

BACKGROUND: Total body irradiation (TBI) has been part of standard conditioning regimens before allogeneic stem cell transplantation for many years. Its effect on normal tissue in these patients has not been studied extensively. METHOD: We studied the in vivo cytogenetic effects of TBI and high-dose chemotherapy on skin fibroblasts from 35 allogeneic stem cell transplantation (SCT) patients. Biopsies were obtained prospectively (n = 18 patients) before, 3 and 12 months after allogeneic SCT and retrospectively (n = 17 patients) 23-65 months after SCT for G-banded chromosome analysis. RESULTS: Chromosomal aberrations were detected in 2/18 patients (11 %) before allogeneic SCT, in 12/13 patients (92 %) after 3 months, in all patients after 12 months and in all patients in the retrospective group after allogeneic SCT. The percentage of aberrant cells was significantly higher at all times after allogeneic SCT compared to baseline analysis. Reciprocal translocations were the most common aberrations, but all other types of stable, structural chromosomal aberrations were also observed. Clonal aberrations were observed, but only in three cases they were detected in independently cultured flasks. A tendency to non-random clustering throughout the genome was observed. The percentage of aberrant cells was not different between patients with and without secondary malignancies in this study group. CONCLUSION: High-dose chemotherapy and TBI leads to severe chromosomal damage in skin fibroblasts of patients after SCT. Our long-term data suggest that this damage increases with time, possibly due to in vivo radiation-induced chromosomal instability.


Subject(s)
Chromosome Aberrations/radiation effects , Fibroblasts/radiation effects , Hematopoietic Stem Cell Transplantation/methods , Transplantation Conditioning/adverse effects , Whole-Body Irradiation/adverse effects , Adolescent , Adult , Allografts , Female , Humans , Male , Middle Aged , Retrospective Studies , Skin/radiation effects , Transplantation Conditioning/methods , Young Adult
8.
Leukemia ; 29(7): 1578-86, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25708834

ABSTRACT

Antibody drug conjugates (ADCs), in which cytotoxic drugs are linked to antibodies targeting antigens on tumor cells, represent promising novel agents for the treatment of malignant lymphomas. Pinatuzumab vedotin is an anti-CD22 ADC and polatuzumab vedotin an anti-CD79B ADC that are both linked to the microtubule-disrupting agent monomethyl auristatin E (MMAE). In the present study, we analyzed the activity of these agents in different molecular subtypes of diffuse large B-cell lymphoma (DLBCL) both in vitro and in early clinical trials. Both anti-CD22-MMAE and anti-CD79B-MMAE were highly active and induced cell death in the vast majority of activated B-cell-like (ABC) and germinal center B-cell-like (GCB) DLBCL cell lines. Similarly, both agents induced cytotoxicity in models with and without mutations in the signaling molecule CD79B. In line with these observations, relapsed and refractory DLBCL patients of both subtypes responded to these agents. Importantly, a strong correlation between CD22 and CD79B expression in vitro and in vivo was not detectable, indicating that patients should not be excluded from anti-CD22-MMAE or anti-CD79B-MMAE treatment because of low target expression. In summary, these studies suggest that pinatuzumab vedotin and polatuzumab vedotin are active agents for the treatment of patients with different subtypes of DLBCL.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD79 Antigens/immunology , Immunoconjugates/pharmacology , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/immunology , Sialic Acid Binding Ig-like Lectin 2/immunology , Apoptosis/drug effects , Blotting, Western , CD79 Antigens/genetics , Cell Cycle/drug effects , Cell Proliferation/drug effects , Clinical Trials, Phase I as Topic , Cohort Studies , Flow Cytometry , Follow-Up Studies , Humans , Immunoenzyme Techniques , Lymphoma, Large B-Cell, Diffuse/classification , Lymphoma, Large B-Cell, Diffuse/pathology , Mutation/genetics , Neoplasm Staging , Prognosis , Sialic Acid Binding Ig-like Lectin 2/genetics , Tumor Cells, Cultured
10.
Br J Cancer ; 111(10): 1917-23, 2014 Nov 11.
Article in English | MEDLINE | ID: mdl-25314063

ABSTRACT

BACKGROUND: Previous investigations in pancreatic cancer suggest a prognostic role for α-smooth muscle actin (α-SMA) expression and stromal density in the peritumoural stroma. The aim of this study was to further validate the impact of α-SMA expression and stromal density in resectable pancreatic cancer patients treated with adjuvant gemcitabine compared with untreated patients. METHODS: CONKO-001 was a prospective randomised phase III study investigating the role of adjuvant gemcitabine as compared with observation. Tissue samples of 162 patients were available for immunohistochemistry on tissue microarrays to evaluate the impact of α-SMA expression and stromal density impact on patient outcome. RESULTS: High α-SMA expression in tumour stroma was associated with worse patient outcome (DFS: P=0.05, OS: P=0.047). A dense stroma reaction was associated with improved disease-free survival (DFS) and overall survival (OS) in the overall study population (DFS: P=0.001, OS: P=0.001). This positive prognostic impact was restricted to patients with no adjuvant treatment (DFS: P<0.001, OS: P<0.001). In multivariable analysis, α-SMA and stromal density expression were independently predictive factors for survival. CONCLUSIONS: Our data confirm the negative prognostic impact of high α-SMA expression in pancreatic cancer patients after curatively intended resection. In contrast to former investigations, we found a positive prognostic impact for a dense stroma. This significant influence was restricted to patients who received no adjuvant therapy.


Subject(s)
Actins/metabolism , Adenocarcinoma/metabolism , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/metabolism , Stromal Cells/metabolism , Tumor Microenvironment , Adenocarcinoma/drug therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/therapeutic use , Biomarkers, Tumor/metabolism , Chemotherapy, Adjuvant , Deoxycytidine/therapeutic use , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , Male , Middle Aged , Neoplasm Grading , Neoplasm Invasiveness , Neoplasm Staging , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Prospective Studies , Stromal Cells/pathology , Survival Rate , Tissue Array Analysis , Gemcitabine
11.
Blood Cancer J ; 4: e217, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24927406

ABSTRACT

Despite evidence that deregulated Notch signalling is a master regulator of multiple myeloma (MM) pathogenesis, its contribution to myeloma bone disease remains to be resolved. Notch promotes survival of human MM cells and triggers human osteoclast activity in vitro. Here, we show that inhibition of Notch through the γ-secretase inhibitor XII (GSI XII) induces apoptosis of murine MOPC315.BM myeloma cells with high Notch activity. GSI XII impairs murine osteoclast differentiation of receptor activator of NF-κB ligand (RANKL)-stimulated RAW264.7 cells in vitro. In the murine MOPC315.BM myeloma model GSI XII has potent anti-MM activity and reduces osteolytic lesions as evidenced by diminished myeloma-specific monoclonal immunoglobulin (Ig)-A serum levels and quantitative assessment of bone structure changes via high-resolution microcomputed tomography scans. Thus, we suggest that Notch inhibition through GSI XII controls myeloma bone disease mainly by targeting Notch in MM cells and possibly in osteoclasts in their microenvironment. We conclude that Notch inhibition is a valid therapeutic strategy in MM.


Subject(s)
Bone Diseases/drug therapy , Dipeptides/pharmacology , Multiple Myeloma/drug therapy , Receptors, Notch/antagonists & inhibitors , Animals , Apoptosis/drug effects , Bone Diseases/metabolism , Bone Diseases/pathology , Cell Differentiation/drug effects , Cell Line, Tumor , Disease Models, Animal , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Female , Humans , Mice , Mice, Inbred BALB C , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Random Allocation , Receptors, Notch/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
Ann Oncol ; 25(5): 1025-32, 2014 May.
Article in English | MEDLINE | ID: mdl-24562449

ABSTRACT

BACKGROUND: Previous investigations in pancreatic cancer suggested a prognostic role for secreted protein acidic and rich in cysteine (SPARC) expression in the peritumoral stroma but not for cytoplasmic SPARC expression. The aim of this study was to evaluate the impact of SPARC expression in pancreatic cancer patients treated with gemcitabine compared with untreated patients. PATIENTS AND METHODS: CONKO-001 was a prospective randomized phase III study investigating the role of adjuvant gemcitabine when compared with observation. Tissue samples of 160 patients were available for SPARC immunohistochemistry on tissue microarrays to evaluate its impact on patient outcome. RESULTS: Strong stromal SPARC expression was associated with worse disease-free survival (DFS) and overall survival (OS) in the overall study population (DFS: P = 0.005, OS: P = 0.033). Its negative prognostic impact was restricted to patients treated with gemcitabine (DFS: P = 0.007, OS: P = 0.006). High cytoplasmic SPARC expression also was associated with worse patient outcome (DFS: P = 0.041, OS: P = 0.011). Again the effect was restricted to patients treated with gemcitabine (DFS: P = 0.002, OS: P = 0.003). In multivariable analysis, SPARC expression was independently predictive of patient outcome. CONCLUSIONS: Our data confirm the prognostic significance of SPARC expression after curatively intended resection. The negative prognostic impact was restricted to patients who received adjuvant treatment with gemcitabine, suggesting SPARC as a predictive marker for response to gemcitabine.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Deoxycytidine/analogs & derivatives , Osteonectin/metabolism , Pancreatic Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/therapy , Chemotherapy, Adjuvant , Deoxycytidine/therapeutic use , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/therapy , Proportional Hazards Models , Prospective Studies , Treatment Outcome , Gemcitabine
13.
Ann Oncol ; 25(1): 210-5, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24356632

ABSTRACT

BACKGROUND: Marginal zone lymphoma (MZL) is a non-Hodgkin lymphoma that occurs as extra nodal, nodal, or splenic. While MZL is generally considered an indolent disease, a substantial percentage of patients follow an unfavorable course. The objective of this retrospective analysis was to identify predictors for a reduced overall survival (OS), or conversely an increased OS. PATIENTS AND METHODS: One hundred and ninety-seven MZL patients were analyzed. Apart from assessing previously published risk factors, concomitant morbidity at diagnosis, transformation into aggressive lymphoma, and occurrence of additional malignancies were evaluated. RESULTS: Next to the known risk factors, i.e. above 60 years of age and elevated serum lactate dehydrogenase (LDH), we demonstrate that transformation into aggressive lymphoma, as well as additional malignancies, are important independent risk factors for a shortened OS in a multivariate analysis, irrespective of the MZL localization. Impressively, in the group of patients lacking LDH elevation, transformation, and/or additional malignancies, only 1 of 63 patients died during follow-up compared with 37 of 87 patients in the high-risk group (HR = 22.8; 95% confidence interval 3.1-167.0; P = 0.002). CONCLUSIONS: Our analysis proposes novel risk factors and warrants for a continuous follow-up to detect the occurrence of transformation and additional malignancies early on.


Subject(s)
Lymphoma, B-Cell, Marginal Zone/pathology , Adult , Aged , Aged, 80 and over , Cell Transformation, Neoplastic/metabolism , Disease Progression , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , L-Lactate Dehydrogenase/blood , Lymphoma, B-Cell, Marginal Zone/blood , Lymphoma, B-Cell, Marginal Zone/mortality , Male , Middle Aged , Multivariate Analysis , Proportional Hazards Models , Risk Factors , Treatment Outcome , Young Adult
14.
Ann Oncol ; 24(12): 3065-9, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24121121

ABSTRACT

BACKGROUND: Treatment options for patients with nonbulky stage IA-IIA Hodgkin lymphoma include combined modality therapy (CMT) using doxorubicin, bleomycin, vinblastine and dacarbazine (ABVD) plus involved-field radiation therapy (IFRT), and chemotherapy with ABVD alone. There are no mature randomized data comparing ABVD with CMT using modern radiation techniques. PATIENTS AND METHODS: Using German Hodgkin Study Group HD10/HD11 and NCIC Clinical Trials Group HD.6 databases, we identified 588 patients who met mutually inclusive eligibility criteria from the preferred arms of HD10 or 11 (n = 406) and HD.6 (n = 182). We evaluated time to progression (TTP), progression-free (PFS) and overall survival, including in three predefined exploratory subset analyses. RESULTS: With median follow-up of 91 (HD10/11) and 134 (HD.6) months, respective 8-year outcomes were for TTP, 93% versus 87% [hazard ratio (HR) 0.44, 95% confidence interval (CI) 0.24-0.78]; for PFS, 89% versus 86% (HR 0.71, 95% CI 0.42-1.18) and for overall survival, 95% versus 95% (HR 1.09, 95% CI 0.49-2.40). In the exploratory subset analysis including HD10 eligible patients who achieved complete response (CR) or unconfirmed complete response (CRu) after two cycles of ABVD, 8-year PFS was 87% (HD10) versus 95% (HD.6) (HR 2.8; 95% CI 0.64-12.5) and overall survival 96% versus 100%. In contrast, among those without CR/CRu after two cycles of ABVD, 8-year PFS was 88% versus 74% (HR 0.35; 95% CI 0.16-0.79) and overall survival 95% versus 91%, respectively (HR 0.42; 95% CI 0.12-1.44). CONCLUSIONS: In patients with nonbulky stage IA-IIA Hodgkin lymphoma, CMT provides better disease control than ABVD alone, especially among those not achieving complete response after two cycles of ABVD. Within the follow-up duration evaluated, overall survivals were similar. Longer follow-up is required to understand the implications of radiation and chemotherapy-related late effects. CLINICAL TRIALS: The trials included in this analysis were registered at ClinicalTrials.gov: HD10 - NCT00265018, HD11 - NCT00264953, HD.6 - NCT00002561.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hodgkin Disease/drug therapy , Adult , Bleomycin/therapeutic use , Chemoradiotherapy , Dacarbazine/therapeutic use , Disease-Free Survival , Doxorubicin/therapeutic use , Hodgkin Disease/mortality , Humans , Proportional Hazards Models , Randomized Controlled Trials as Topic , Retrospective Studies , Treatment Outcome , Vinblastine/therapeutic use
15.
Front Physiol ; 4: 118, 2013.
Article in English | MEDLINE | ID: mdl-23745115

ABSTRACT

BACKGROUND: Genetic risk factors for sporadic pancreatic cancer are largely unknown but actually under high exposure. Findings of correlations between the AB0 blood group system (Chromosome 9q34,1-q34,2) and the risk of pancreatic cancer (PC) in patients from Asia, America and south Europe have already been published. So far it is unclear, whether this correlation between blood group an PC incidence can be found in German patients as well. METHODS: One hundred and sixty-six patients who underwent a resection of PC were evaluated in a period between 2000 and 2010. Blood group reference distribution for the German population is given as: 0: 41%; A: 43%; B: 11%; AB: 5%; Rhesus positive: 85%; Rhesus negative: 15%. Analyses were done using the non-parametric Chi(2)-test (p-value two sided; SPSS 19.0). RESULTS: Median age was 62 (34-82) years. Gender: female 73/44%; male: 93/56%. Observed blood group proportions: 0: 43 (25.9%)/A: 94 (56.6%)/B: 16 (9.6%)/AB: 13 (7.8%)/Rhesus positive: 131 (78.9%)/negative: 35 (21.1%). We detected a significant difference to the German reference distribution of the AB0 system (Chi(2) 19.34, df 3, p < 0.001). Rhesus factor has no impact on AB0-distribution (Chi(2) 4.13, df 3, p = 0.25), but differs significantly from reference distribution-probably due to initial AB0-variation (Chi(2) 4.82, df 1, p = 0.028). The odds ratio for blood group A is 2.01 and for blood group 0 is 0.5. CONCLUSIONS: The incidence of PC in the German cohort is highly associated with the AB0-system as well. More patients with blood group A suffer from PC (p < 0.001) whereas blood group 0 was less frequent in patients with PC (p < 0.001). Thus, our findings support the results from other non-German surveys. The causal trigger points of this carcinogenesis correlation are still not known.

16.
Leukemia ; 27(6): 1316-21, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23459449

ABSTRACT

Several retrospective studies have described the clinical manifestation of peripheral artery occlusive disease (PAOD) in patients receiving nilotinib. We thus prospectively screened for PAOD in patients with chronic phase chronic myeloid leukemia (CP CML) being treated with tyrosine kinase inhibitors (TKI), including imatinib and nilotinib. One hundred and fifty-nine consecutive patients were evaluated for clinical and biochemical risk factors for cardiovascular disease. Non-invasive assessment for PAOD included determination of the ankle-brachial index (ABI) and duplex ultrasonography. A second cohort consisted of patients with clinically manifest PAOD recruited from additional collaborating centers. Pathological ABI were significantly more frequent in patients on first-line nilotinib (7 of 27; 26%) and in patients on second-line nilotinib (10 of 28; 35.7%) as compared with patients on first-line imatinib (3 of 48; 6.3%). Clinically manifest PAOD was identified in five patients, all with current or previous nilotinib exposure only. Relative risk for PAOD determined by a pathological ABI in first-line nilotinib-treated patients as compared with first-line imatinib-treated patients was 10.3. PAOD is more frequently observed in patients receiving nilotinib as compared with imatinib. Owing to the severe nature of clinically manifest PAOD, longitudinal non-invasive monitoring and careful assessment of risk factors is warranted.


Subject(s)
Antineoplastic Agents/adverse effects , Arterial Occlusive Diseases/complications , Benzamides/adverse effects , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Peripheral Arterial Disease/complications , Piperazines/adverse effects , Pyrimidines/adverse effects , Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Cohort Studies , Humans , Imatinib Mesylate , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/complications , Piperazines/therapeutic use , Pyrimidines/therapeutic use
17.
Dig Dis Sci ; 58(8): 2399-405, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23525734

ABSTRACT

BACKGROUND: Effective and tolerable chemotherapy with gemcitabine and cisplatin for advanced biliary tract cancer (BTC) has been established recently. However, overall prognosis is still poor, and additional therapeutic approaches are needed for patients with locally advanced, irresectable and/or pretreated tumors. Hepatic arterial infusion (HAI) of chemotherapy represents a safe and well-established treatment modality, but data on its use in patients with BTC are still sparse. METHODS: Patients with irresectable BTC predominant to the liver were included in a prospective, open phase II study investigating HAI provided through interventionally implanted port catheters. Intraarterial chemotherapy consisted of biweekly oxaliplatin (O) 85 mg/m(2) and folinic acid (F) 170 mg/m(2) with 5-FU (F) 600 mg/m(2). RESULTS: Between 2004 and 2010, 37 patients were enrolled. A total of 432 cycles of HAI were applied with a median of 9 (range 1-46) cycles. Objective response rate was 16 %, and tumor control was achieved in 24 of 37 (65 %) patients. Median progression-free survival was 6.5 months (range 0.5-26.0; 95 % CI 4.3-8.7), median overall survival was 13.5 (range 0.9-50.7; 95 % CI 11.1-15.9) months. The most frequent adverse event was sensory neuropathy grade 1/2 in 10/14 patients. CONCLUSIONS: Using a minimal invasive technique, repetitive HAI with OFF is feasible and results in clinically relevant tumor control with low toxicity in patients with liver predominant advanced BTC.


Subject(s)
Biliary Tract Neoplasms/drug therapy , Fluorouracil/therapeutic use , Leucovorin/therapeutic use , Liver/blood supply , Organoplatinum Compounds/therapeutic use , Adult , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Drug Administration Schedule , Drug Therapy, Combination , Female , Fluorouracil/administration & dosage , Humans , Leucovorin/administration & dosage , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Oxaliplatin
18.
Leukemia ; 27(6): 1381-90, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23257783

ABSTRACT

Myeloid cell leukemia-1 (MCL1) is an anti-apoptotic member of the BCL2 family that is deregulated in various solid and hematological malignancies. However, its role in the molecular pathogenesis of diffuse large B-cell lymphoma (DLBCL) is unclear. We analyzed gene expression profiling data from 350 DLBCL patient samples and detected that activated B-cell-like (ABC) DLBCLs express MCL1 at significantly higher levels compared with germinal center B-cell-like DLBCL patient samples (P=2.7 × 10(-10)). Immunohistochemistry confirmed high MCL1 protein expression predominantly in ABC DLBCL in an independent patient cohort (n=249; P=0.001). To elucidate molecular mechanisms leading to aberrant MCL1 expression, we analyzed array comparative genomic hybridization data of 203 DLBCL samples and identified recurrent chromosomal gains/amplifications of the MCL1 locus that occurred in 26% of ABC DLBCLs. In addition, aberrant STAT3 signaling contributed to high MCL1 expression in this subtype. Knockdown of MCL1 as well as treatment with the BH3-mimetic obatoclax induced apoptotic cell death in MCL1-positive DLBCL cell lines. In summary, MCL1 is deregulated in a significant fraction of ABC DLBCLs and contributes to therapy resistance. These data suggest that specific inhibition of MCL1 might be utilized therapeutically in a subset of DLBCLs.


Subject(s)
Gene Expression Profiling , Lymphoma, Large B-Cell, Diffuse/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Antineoplastic Agents/therapeutic use , Comparative Genomic Hybridization , Humans , Immunohistochemistry , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/pathology , Myeloid Cell Leukemia Sequence 1 Protein , Real-Time Polymerase Chain Reaction
19.
Ann Oncol ; 24(5): 1363-70, 2013 May.
Article in English | MEDLINE | ID: mdl-23247660

ABSTRACT

BACKGROUND: The prognostic value of the NIH consensus criteria for graft-versus-host disease (GVHD) is not well defined yet. PATIENTS AND METHODS: We analyzed NIH-defined GVHD in 147 acute lymphoblastic leukemia (ALL) patients. RESULTS: The cumulative incidence of classic acute GVHD (aGVHD), late aGVHD and chronic GVHD (cGVHD) was 63%, 12% and 41%, respectively. cGVHD was subclassified as classic versus overlap syndrome in 40% versus 60% of cases. In multivariate Cox regression analysis with GVHD as time-dependent covariate, classic aGVHD grade III/IV had a negative impact on overall survival (OS) due to higher non-relapse mortality. cGVHD of any grade was associated with superior OS, which was due to lower relapse incidence. Classic cGVHD versus overlap syndrome had no differential impact. In 44 patients without GVHD after transplant who received donor lymphocyte infusions (DLI), the cumulative incidence of classic aGVHD, late aGVHD or cGVHD was 60%, 5% and 57%. Occurrence of cGVHD after DLI was associated with improved OS due to lower relapse incidence. CONCLUSIONS: The NIH consensus criteria for GVHD clearly define prognostic subgroups in patients transplanted for ALL. The improved OS in patients developing cGVHD after transplant or DLI gives clear evidence for a potent graft-versus-leukemia effect in this indication.


Subject(s)
Graft vs Host Disease , Graft vs Leukemia Effect , Hematopoietic Stem Cell Transplantation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Adolescent , Adult , Disease-Free Survival , Female , Humans , Male , Middle Aged , Neoplasm Recurrence, Local , Transplantation Conditioning , Transplantation, Homologous , Transplants , Young Adult
20.
Cancer Gene Ther ; 19(12): 880-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23099886

ABSTRACT

The aim of this study was to evaluate the efficacy of the chemokine CCL19 (ELC) as an adjuvant for intradermal gene gun delivery of Her2/neu DNA and to investigate the role of B cells in CCL19-mediated enhancement of immune responses. Balb/c mice were immunized intramuscularly (i.m.) on days 1 and 15 with plasmid DNA (pDNA) (100 µg DNA) or intradermally (i.d.) by gene gun delivery (1-2 µg DNA). Administration of pDNA encoding Her2/neu (pDNA(Her2/neu) was compared with pDNA(Her2/neu) plus pDNA(CCL19), pDNA(CCL19), mock vector or uncoated gold particles/phosphate-buffered saline (PBS). Tumor challenge was performed subcutaneously on day 25 with syngeneic Her2/neu(+) tumor cells (D2F2/E2). Intradermal immunization by gene gun led to an enhancement of tumor protection by the DNA vaccine as compared with i.m. immunization. The protective effect of the vaccine was further enhanced by coadministration of pDNA(CCL19) both after i.m. and i.d. immunization. Tumor protection was associated with Her2/neu-specific T cell and humoral immune responses. Experiments in B-cell-deficient µMT mice showed that B cells are crucial for CCL19-mediated enhancement of tumor rejection, most likely as antigen-presenting B cells. DNA vaccines against Her2/neu may play a future role in the treatment of Her2/neu-positive breast cancer patients in a clinical situation of minimal residual disease.


Subject(s)
B-Lymphocytes/immunology , Biolistics/methods , Cancer Vaccines/administration & dosage , Chemokine CCL19/genetics , Chemokine CCL19/immunology , Mammary Neoplasms, Experimental/therapy , Receptor, ErbB-2/immunology , Vaccines, DNA/administration & dosage , Animals , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Chemokine CCL19/metabolism , Disease Models, Animal , Female , Humans , Injections, Intradermal , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/immunology , Mice , Mice, Inbred BALB C , Receptor, ErbB-2/genetics , T-Lymphocytes/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...