Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 29
Filtrer
1.
Ann Hematol ; 101(5): 1031-1038, 2022 May.
Article de Anglais | MEDLINE | ID: mdl-35262868

RÉSUMÉ

Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) can be late complications following mutagenic treatment. Limited data is available on the outcome of patients developing therapy-related MDS and AML after treatment for multiple myeloma (MM). We identified 250 patients with therapy-associated MDS or AML in the Duesseldorf MDS registry. Of those, 50 patients were previously diagnosed with multiple myeloma (mm-MDS/AML). We compared them to patients with de novo MDS (n = 4862) and to patients with MDS following other underlying diseases (tMDS) (n = 200). mm-MDS patients and tMDS patients showed similar karyotypes and degrees of cytopenia. However, mm-MDS patients had significantly higher blast counts and more often belonged to the high-risk group according to the International Prognostic Scoring System (IPSS) (both p < 0.05). Although the rate of progression to AML was similar in mm-MDS and tMDS, both transformed significantly more often than de novo MDS (p < 0.05). Median overall survival of patients with mm-MDS (13 months; range: 1-99) and tMDS (13 months; range 0-160) was also similar yet significantly shorter than patients with de novo MDS (32 months; range 0-345 months; p < 0.05). Furthermore, survival of mm-MDS patients was not affected by myeloma activity. Despite significantly more high-risk disease and higher blast cell counts, myeloma-associated MDS-patients show features akin to other tMDS. Survival is similar to other tMDS and irrespective of myeloma remission status or transformation to AML. Thus, patient outcome is not determined by competing clones but rather by MDS governing the stem cell niche.


Sujet(s)
Leucémie aigüe myéloïde , Myélome multiple , Syndromes myélodysplasiques , Seconde tumeur primitive , Humains , Caryotypage , Leucémie aigüe myéloïde/induit chimiquement , Leucémie aigüe myéloïde/diagnostic , Leucémie aigüe myéloïde/thérapie , Myélome multiple/complications , Myélome multiple/thérapie , Syndromes myélodysplasiques/induit chimiquement , Syndromes myélodysplasiques/complications , Syndromes myélodysplasiques/thérapie , Seconde tumeur primitive/étiologie
2.
Leukemia ; 30(3): 683-91, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26601782

RÉSUMÉ

Hematopoietic insufficiency is the hallmark of acute myeloid leukemia (AML) and predisposes patients to life-threatening complications such as bleeding and infections. Addressing the contribution of mesenchymal stromal cells (MSC) to AML-induced hematopoietic failure we show that MSC from AML patients (n=64) exhibit significant growth deficiency and impaired osteogenic differentiation capacity. This was molecularly reflected by a specific methylation signature affecting pathways involved in cell differentiation, proliferation and skeletal development. In addition, we found distinct alterations of hematopoiesis-regulating factors such as Kit-ligand and Jagged1 accompanied by a significantly diminished ability to support CD34+ hematopoietic stem and progenitor cells in long-term culture-initiating cells (LTC-ICs) assays. This deficient osteogenic differentiation and insufficient stromal support was reversible and correlated with disease status as indicated by Osteocalcin serum levels and LTC-IC frequencies returning to normal values at remission. In line with this, cultivation of healthy MSC in conditioned medium from four AML cell lines resulted in decreased proliferation and osteogenic differentiation. Taken together, AML-derived MSC are molecularly and functionally altered and contribute to hematopoietic insufficiency. Inverse correlation with disease status and adoption of an AML-like phenotype after exposure to leukemic conditions suggests an instructive role of leukemic cells on bone marrow microenvironment.


Sujet(s)
Régulation de l'expression des gènes dans la leucémie , Leucémie aigüe myéloïde/génétique , Cellules souches mésenchymateuses/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigènes CD34/génétique , Antigènes CD34/métabolisme , Protéines de liaison au calcium/génétique , Protéines de liaison au calcium/métabolisme , Études cas-témoins , Différenciation cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Milieux de culture conditionnés/pharmacologie , Femelle , Hématopoïèse/génétique , Humains , Protéines et peptides de signalisation intercellulaire/génétique , Protéines et peptides de signalisation intercellulaire/métabolisme , Protéine jagged-1 , Leucémie aigüe myéloïde/métabolisme , Leucémie aigüe myéloïde/anatomopathologie , Mâle , Protéines membranaires/génétique , Protéines membranaires/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/anatomopathologie , Adulte d'âge moyen , Ostéocalcine/génétique , Ostéocalcine/métabolisme , Ostéogenèse/effets des médicaments et des substances chimiques , Phénotype , Protéines serrate-jagged , Transduction du signal , Facteur de croissance des cellules souches/génétique , Facteur de croissance des cellules souches/métabolisme
4.
Leukemia ; 27(9): 1841-51, 2013 Sep.
Article de Anglais | MEDLINE | ID: mdl-23797473

RÉSUMÉ

Ineffective hematopoiesis is a major characteristic of myelodysplastic syndromes (MDS) causing relevant morbidity and mortality. Mesenchymal stromal cells (MSC) have been shown to physiologically support hematopoiesis, but their contribution to the pathogenesis of MDS remains elusive. We show that MSC from patients across all MDS subtypes (n=106) exhibit significantly reduced growth and proliferative capacities accompanied by premature replicative senescence. Osteogenic differentiation was significantly reduced in MDS-derived MSC, indicated by cytochemical stainings and reduced expressions of Osterix and Osteocalcin. This was associated with specific methylation patterns that clearly separated MDS-MSC from healthy controls and showed a strong enrichment for biological processes associated with cellular phenotypes and transcriptional regulation. Furthermore, in MDS-MSC, we detected altered expression of key molecules involved in the interaction with hematopoietic stem and progenitor cells (HSPC), in particular Osteopontin, Jagged1, Kit-ligand and Angiopoietin as well as several chemokines. Functionally, this translated into a significantly diminished ability of MDS-derived MSC to support CD34+ HSPC in long-term culture-initiating cell assays associated with a reduced cell cycle activity. Taken together, our comprehensive analysis shows that MSC from all MDS subtypes are structurally, epigenetically and functionally altered, which leads to impaired stromal support and seems to contribute to deficient hematopoiesis in MDS.


Sujet(s)
Cellules souches mésenchymateuses/métabolisme , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Études cas-témoins , Différenciation cellulaire , Prolifération cellulaire , Vieillissement de la cellule , Analyse de regroupements , Test clonogénique , Méthylation de l'ADN , Épigenèse génétique , Femelle , Analyse de profil d'expression de gènes , Humains , Immunophénotypage , Mâle , Cellules souches mésenchymateuses/cytologie , Adulte d'âge moyen , Ostéogenèse/génétique , Phénotype
6.
Leukemia ; 27(6): 1229-35, 2013 Jun.
Article de Anglais | MEDLINE | ID: mdl-23314834

RÉSUMÉ

The combination of azacitidine and donor lymphocyte infusions (DLI) as first salvage therapy for relapse after allogeneic transplantation (allo-HSCT) was studied in 30 patients with acute myeloid leukemia (AML; n=28) or myelodysplastic syndromes (MDS; n=2) within a prospective single-arm multicenter phase-II trial. Treatment schedule contained up to eight cycles azacitidine (100 mg/m(2)/day, days 1-5, every 28 days) followed by DLI (from 1-5 × 10(6) to 1-5 × 10(8) CD3(+)cells/kg) after every second azacitidine cycle. A median of three courses azacitidine (range 1-8) were administered, and 22 patients (73%) received DLI. Overall response rate was 30%, including seven complete remissions (CRs, 23%) and two partial remissions (7%). Five patients remain in CR for a median of 777 days (range 461-888). Patients with MDS or AML with myelodysplasia-related changes were more likely to respond (P=0.011), and a lower blast count (P=0.039) as well as high-risk cytogenetics (P=0.035) correlated with the likelihood to achieve CR. Incidence of acute and chronic graft-versus-host disease was 37% and 17%, respectively. Neutropenia and thrombocytopenia grade III/IV occurred during 65% and 63% of treatment cycles, while infections were the most common grade III/IV non-hematological toxicity. Azacitidine and DLI as salvage therapy is safe, induces long-term remissions and may become an alternative for patients with AML or MDS relapsing after allo-HSCT.


Sujet(s)
Antimétabolites antinéoplasiques/usage thérapeutique , Azacitidine/usage thérapeutique , Transplantation cellulaire , Leucémie aigüe myéloïde/thérapie , Lymphocytes/cytologie , Syndromes myélodysplasiques/thérapie , Thérapie de rattrapage , Transplantation de cellules souches , Adulte , Sujet âgé , Association thérapeutique , Femelle , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/chirurgie , Mâle , Adulte d'âge moyen , Syndromes myélodysplasiques/traitement médicamenteux , Syndromes myélodysplasiques/chirurgie , Récidive , Transplantation homologue
8.
Bone Marrow Transplant ; 46(5): 747-55, 2011 May.
Article de Anglais | MEDLINE | ID: mdl-20661230

RÉSUMÉ

In the current study, we evaluated a combination of tacrolimus and mycophenolate mofetil (MMF) as GvHD prophylaxis in 50 patients undergoing truly nonmyeloablative (NM; 90 mg/m(2) fludarabine, 2 Gy TBI) hematopoietic SCT (HSCT) from unrelated donors. Median patient age was 51 years (range, 25-67 years). After a median follow-up of 1123 days (range, 47-2729 days), 20 patients (40%) are alive and free from disease. The probabilities of 1-, 2- and 3-year survival were 57, 47 and 39%, respectively. Patients who achieved a remission before HSCT had a significantly better OS compared with those who had active disease (P=0.01). The incidences of grade II-IV and III-IV acute GvHD (aGvHD) were 54% (n=27) and 16% (n=8). Remarkably, using tacrolimus and MMF, the median onset of aGvHD occurred distinctly late on day +66 (range, 12-119 days). A total of 46 patients were evaluable for chronic GvHD (cGvHD). Out of these, 26 (56%) patients developed cGvHD, with 16 (34%) of them showing limited and 10 (21%) showing extensive disease. We conclude that the combination of tacrolimus and MMF as post transplant immunosuppression for patients receiving NM unrelated donor HSCT permits stable engraftment and effective prophylaxis for acute and cGvHD. In particular, the occurrence of severe early-onset aGvHD was attenuated.


Sujet(s)
Maladie du greffon contre l'hôte/prévention et contrôle , Transplantation de cellules souches hématopoïétiques , Immunosuppresseurs/usage thérapeutique , Acide mycophénolique/analogues et dérivés , Tacrolimus/administration et posologie , Adulte , Sujet âgé , Femelle , Hémopathies/traitement médicamenteux , Transplantation de cellules souches hématopoïétiques/effets indésirables , Humains , Mâle , Adulte d'âge moyen , Acide mycophénolique/administration et posologie , Acide mycophénolique/usage thérapeutique , Tacrolimus/usage thérapeutique , Chimère obtenue par transplantation , Conditionnement pour greffe/effets indésirables , Conditionnement pour greffe/méthodes , Transplantation homologue , Résultat thérapeutique , Vidarabine/analogues et dérivés , Vidarabine/usage thérapeutique , Irradiation corporelle totale
9.
Ann Oncol ; 21(9): 1898-1904, 2010 Sep.
Article de Anglais | MEDLINE | ID: mdl-20233744

RÉSUMÉ

BACKGROUND: We retrospectively determined whether a 3-day short course of palifermin could reduce the toxicity of high-dose therapy (HDT) and autologous blood stem-cell transplantation (ASCT) in patients with multiple myeloma (MM). PATIENTS AND METHODS: Sixty-seven consecutive patients received 60 mug/kg palifermin for 3 days before HDT with melphalan 200 or 140 mg/m(2) for patients with renal failure (group A). Granulocyte colony-stimulating factor (G-CSF) was applied after ASCT. Data on haematopoietic reconstitution and toxicity were compared with two previously published patient groups from our institution who had received pegfilgrastim but not palifermin (group B, n = 21) and patients who had received neither palifermin nor G-CSF (group C, n = 21). RESULTS: In group A, patients with renal failure had a significantly higher risk for severe mucositis (64% versus 16%, P < 0.002). Patients with normal renal function who received palifermin experienced significantly less days of hospitalisation (P < 0.05) and less need for narcotic analgesia (P < 0.05), parenteral nutrition (P < 0.05) and erythrocyte transfusions (P < 0.05) in comparison with groups B and C. Time to haematopoietic reconstitution was not compromised by the use of palifermin. CONCLUSIONS: In conclusion, a short 3-day course of palifermin may be able to reduce the toxicity of HDT and ASCT in patients with MM. Patients with impaired renal function at the time of HDT need additional strategies to further reduce the incidence of severe mucositis.


Sujet(s)
Facteur de croissance fibroblastique de type 7/usage thérapeutique , Inflammation muqueuse/prévention et contrôle , Myélome multiple/thérapie , Récidive tumorale locale/prévention et contrôle , Transplantation de cellules souches de sang périphérique , Adulte , Sujet âgé , Antinéoplasiques alcoylants/administration et posologie , Femelle , Filgrastim , Études de suivi , Facteur de stimulation des colonies de granulocytes/administration et posologie , Humains , Mâle , Melphalan/administration et posologie , Adulte d'âge moyen , Inflammation muqueuse/étiologie , Myélome multiple/anatomopathologie , Récidive tumorale locale/anatomopathologie , Stadification tumorale , Soins palliatifs , Polyéthylène glycols , Protéines recombinantes , Études rétrospectives , Taux de survie , Transplantation autologue , Résultat thérapeutique
10.
Bone Marrow Transplant ; 45(5): 872-6, 2010 May.
Article de Anglais | MEDLINE | ID: mdl-19820729

RÉSUMÉ

Patients with AML or myelodysplastic syndrome who relapse after allo-SCT have a poor prognosis. In the search for novel treatment strategies for these patients, we conducted a multicenter retrospective analysis and identified 22 patients treated with the DNA-methylation inhibitor 5-azacytidine (5-Aza). Patients received a median number of two cycles 5-Aza (range 1-8) at a dose of 100 mg/m(2) over 5 days following relapse. Eighteen patients (82%) also received a median number of two donor lymphocyte infusions (DLI, range 1-5). Sixteen patients (72%) responded to 5-Aza treatment and five patients (23%) achieved a CR. 5-Aza-induced CR lasted for 433 days (median, range 114-769). Median survival and the estimated 2-year survival rate were 144 days and 23%, respectively. Acute GVHD after DLI was seen in six patients (33%) and four of these patients developed chronic GVHD of the skin. There were no treatment-related deaths. Patients who achieved halving of leukocyte counts after the first 5-Aza cycle had a superior median survival of 802 days compared with 135 days (P=0.0025) in all other patients. On univariate analysis, the achievement of this halving of leukocyte counts was identified as a significant predictor of survival.


Sujet(s)
Azacitidine/usage thérapeutique , Leucémie aigüe myéloïde/thérapie , Syndromes myélodysplasiques/thérapie , Transplantation de cellules souches/effets indésirables , Adulte , Sujet âgé , Femelle , Maladie du greffon contre l'hôte/complications , Maladie du greffon contre l'hôte/traitement médicamenteux , Humains , Mâle , Adulte d'âge moyen , Récidive , Études rétrospectives , Facteurs de risque , Thérapie de rattrapage , Analyse de survie , Transplantation homologue
11.
Bone Marrow Transplant ; 44(12): 785-92, 2009 Dec.
Article de Anglais | MEDLINE | ID: mdl-19430496

RÉSUMÉ

In this prospective study, we examined the toxicity and efficacy of an intensified conditioning regimen for treatment of patients with relapsed or high-risk acute lymphoblastic leukemia who undergo allogeneic hematopoietic stem cell transplantation. Fifteen patients received fludarabine 30 mg/m(2), cytarabine 2000 mg/m(2), amsacrine 100 mg/m(2) on days -10, -9, -8 and -7, anti-thymocyte globulin (ATG-Fresenius) 20 mg/kg body weight on days -6, -5 and -4 and fractionated total body irradiation 2 x 2 Gy on days -3, -2 and -1 (FLAMSA-ATG-TBI) before allogeneic hematopoietic stem cell transplantation. At the time of hematopoietic stem cell transplantation, 10 patients were in complete remission (8 CR1; 2 CR2), 3 with primary refractory and 2 suffered from refractory relapse. All patients achieved a complete remission after hematopoietic stem cell transplantation; and after a median follow-up time of 1091 days (range, 334-1554 days), nine patients (60%) are alive and free from disease, including three patients with prior refractory disease. Three patients died due to treatment-related mortality. The most frequent and severe conditioning-related toxicities observed in 9 out of 15 patients were grade III/IV infections according to common toxicity criteria. Thus, conditioning with the FLAMSA-ATG-TBI regimen is a feasible and effective alternative for patients with relapsed or high-risk acute lymphoblastic leukemia.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Conditionnement pour greffe/méthodes , Adolescent , Adulte , Amsacrine/administration et posologie , Cytarabine/administration et posologie , Survie sans rechute , Études de suivi , Transplantation de cellules souches hématopoïétiques , Humains , Mâle , Adulte d'âge moyen , Leucémie-lymphome lymphoblastique à précurseurs B et T/mortalité , Récidive , Facteurs de risque , Taux de survie , Transplantation homologue , Vidarabine/administration et posologie , Vidarabine/analogues et dérivés , Irradiation corporelle totale/méthodes
12.
Bone Marrow Transplant ; 43(9): 669-77, 2009 May.
Article de Anglais | MEDLINE | ID: mdl-19308043

RÉSUMÉ

To date, G-CSF is the most favoured cytokine administered for PBSC mobilization because of its great efficacy and lack of serious toxicity. Recently, a pegylated filgrastim (pegfilgrastim) has been introduced. Attachment of the polyethylene glycol (PEG) moiety reduces renal excretion and masks proteolytic cleavage sites resulting in elevated G-CSF serum levels for up to 14 days after a single injection. As single-dose pegfilgrastim had similar effects in the prophylaxis of chemotherapy-induced neutropenia as the daily administration of the unconjugated drug, its capability for the mobilization of haematopoietic stem and progenitor cells has been assessed and presented to be at least equal to that of conventional G-CSF. Administration of pegfilgrastim following high-dose therapy and autologous blood SCT (BSCT) shortened the time to myeloid recovery as seen in conventional G-CSF. Plasma G-CSF levels were about 1 log higher with pegfilgrastim, but in the setting of autologous BSCT this did not translate into a faster haematopoietic recovery. Only few data exist on the biological effects of pegfilgrastim. Still, these data suggest that pegfilgrastim-stimulation results in different functional properties of haematopoietic stem and progenitor cells compared with G-CSF.


Sujet(s)
Facteur de stimulation des colonies de granulocytes/usage thérapeutique , Mobilisation de cellules souches hématopoïétiques/méthodes , Transplantation de cellules souches hématopoïétiques/méthodes , Filgrastim , Hématopoïèse/effets des médicaments et des substances chimiques , Humains , Cinétique , Polyéthylène glycols , Protéines recombinantes , Transplantation autologue
13.
Leuk Res ; 33(2): 348-50, 2009 Feb.
Article de Anglais | MEDLINE | ID: mdl-18573526

RÉSUMÉ

The fms-related tyrosine kinase 3 internal tandem duplication (FLT3-ITD) can be found in about one quarter of patients with acute myeloid leukemia (AML) [Small D. FLT3 mutations: biology and treatment. Hematology Am Soc Hematology. Educ. Program 2006;178-84 [Review]]. Patients who carry this mutation have a high risk of relapse even after allogeneic stem cell transplantation [Sheikhha MH, Awan A, Tobal K, Liu Yin JA. Prognostic significance of FLT3 ITD and D835 mutations in AML patients. Hematol J 2003;4:41-6; Meshinchi S, Arceci RJ, Sanders JE, Smith FO, Woods WB, Radich JP, et al. Role of allogeneic stem cell transplantation in FLT3/ITD-positive AML. Blood 2006;108(1):400-1]. Recent reports show that Sorafenib, a multikinase inhibitor has significant activity against FLT3-ITD(+) blasts in vitro [Auclair D, Miller D, Yatsula V, Pickett W, Carter C, Chang Y, et al. Antitumor activity of sorafenib in FLT3-driven leukemic cells. Leukemia 2007;21(3):439-45]. We here report the first clinical case of molecular remission induced by Sorafenib in a patient with FLT3-ITD(+) AML and extramedullary disease after allogenic stem cell transplantation.


Sujet(s)
Benzènesulfonates/usage thérapeutique , Leucémie aigüe myéloïde/traitement médicamenteux , Pyridines/usage thérapeutique , Tyrosine kinase-3 de type fms/génétique , Humains , Leucémie aigüe myéloïde/génétique , Nicotinamide/analogues et dérivés , Phénylurées , Induction de rémission/méthodes , Sorafénib , Séquences répétées en tandem
14.
Horm Metab Res ; 40(2): 108-16, 2008 Feb.
Article de Anglais | MEDLINE | ID: mdl-18283628

RÉSUMÉ

The existence of inherited aggressive forms of medullary thyroid carcinoma (MTC) and their resistance to classical therapies make it a prime candidate for adoptive immunotherapy. Highly potent antigen-presenting cells, namely dendritic cells (DCs), may serve as an interesting tool for anticancer vaccination. Here we report on the IN VITRO findings of a vaccination trial in five MTC patients, who were treated with a new DC generation protocol consisting of granulocyte-macrophage colony-stimulating factor and interferon-alpha (IFN-DCs). These cells were pulsed with tumor-specific calcitonin and administered twice. In two patients who responded to therapy we found a large increase (in mean 2.9+/-1.9%) of antigen-specific IFN-gamma-secreting CD4+ cells as well as an increase of granzyme B positive CD8+ cells (mean 2.2+/-0.2%) in the peripheral blood. In parallel, a decrease of CD4+/CD25+/FoxP3+ regulatory T cells was seen. Importantly, IN VITRO stimulation of PBMC with 10 different 15mer calcitonin peptides resulted in the identification of two HLA class II epitope regions within the central part of full-length calcitonin. These data were in accordance with the results drawn from the computer-based algorithm epitope prediction software SYFPEITHI. Measurement of different pro- and anti-angiogenic factors did not allow for a distinct outcome of prediction of the treated patients. In summary, we have demonstrated that immunization with IFN-DCs leads to a tumor epitope-specific immune response in MTC patients and may, therefore, represent a promising tool for future vaccination trials.


Sujet(s)
Antigènes néoplasiques/immunologie , Vaccins anticancéreux/usage thérapeutique , Carcinome médullaire/immunologie , Cellules dendritiques/immunologie , Lymphocytes auxiliaires Th1/immunologie , Tumeurs de la thyroïde/immunologie , Séquence d'acides aminés , Agents angiogéniques/sang , Agents angiogéniques/métabolisme , Calcitonine/synthèse chimique , Calcitonine/immunologie , Calcitonine/usage thérapeutique , Vaccins anticancéreux/synthèse chimique , Vaccins anticancéreux/immunologie , Carcinome médullaire/thérapie , Séparation cellulaire , Cellules dendritiques/transplantation , Cartographie épitopique , Cytométrie en flux , Facteur de stimulation des colonies de granulocytes et de macrophages/immunologie , Humains , Immunothérapie adoptive , Interféron alpha/immunologie , Données de séquences moléculaires , Lymphocytes T/cytologie , Lymphocytes T/immunologie , Tumeurs de la thyroïde/thérapie , Vaccins sous-unitaires/synthèse chimique , Vaccins sous-unitaires/immunologie , Vaccins sous-unitaires/usage thérapeutique
15.
Horm Metab Res ; 40(2): 117-21, 2008 Feb.
Article de Anglais | MEDLINE | ID: mdl-18283629

RÉSUMÉ

The antitumor effects of IFNalpha is mainly mediated by the activation of cytotoxic T lymphocytes (CTLs), the activation of natural killer (NK) cells, and the generation of highly potent antigen-presenting dendritic cells (IFN-DCs). Recently, we demonstrated that these cells partially express the NK cell marker CD56 and reveal a direct cytotoxic immunity towards tumor cells. The aim of the present study was to explore these cells in more detail with respect to their phenotypical and functional characteristics. Flowcytometric analyses revealed that a 5-day incubation time of CD14+ monocytes with IFNalpha results in a steady increase of CD56 surface expression of these cells from 25% (+/-2%) on day 1 up to 68% (+/-11%) on day 5. Interestingly, additional culturing of negatively selected CD56- IFN-DCs also resulted in a partial CD56 surface expression. By comparing both cell types in more detail we found a significant decrease of CD14 expression on CD56+ IFN-DCs (66+/-6%) compared to CD56- IFN-DCs (76+/-6%). On the basis of functional tests, CD56+ IFN-DCs revealed a slightly increased phagocytosis capacity compared to CD56- IFN-DCs as only 82% of CD56- IFN-DCs showed a positive intracytoplasmatic signal after 60 minutes coculturing with FITC-labeled albumin, whereas 91% of CD56+ IFN-DCs were positive. Moreover, CD56+ IFN-DCs revealed a stronger T cell stimulation capacity compared to CD56- IFN-DCs. These results together with our previously described data suggest that CD56+ IFN-DCs and CD56- IFN-DCs may represent one identical cell population with different maturation status rather than two separate cell entities. Because of their high stimulating capacity and their direct cytolytic effects these cells represent a new promising tool for cellular anticancer therapy.


Sujet(s)
Cellules dendritiques/immunologie , Interféron alpha/immunologie , Monocytes/immunologie , Marqueurs biologiques/sang , Antigènes CD56/immunologie , Antigènes CD56/métabolisme , Séparation cellulaire , Techniques de coculture , Cellules dendritiques/composition chimique , Cellules dendritiques/cytologie , Cytométrie en flux/méthodes , Humains , Cellules tueuses naturelles/cytologie , Activation des lymphocytes , Monocytes/cytologie , Phagocytose
17.
Cancer Gene Ther ; 14(4): 431-9, 2007 Apr.
Article de Anglais | MEDLINE | ID: mdl-17235352

RÉSUMÉ

In this report, the effects of a combined treatment with the proteasome inhibitor bortezomib and either a recombinant adeno-associated virus type 2 (rAAV-2)-mediated p53 gene transfer or chemotherapeutic agents, docetaxel and pemetrexed, were tested on p53 positive and p53negative non-small cell lung cancer (NSCLC) cell lines. The combination of bortezomib and rAAV-p53 led to a significant synergistic inhibition of cell growth between 62-82% depending on the p53 status of the cell line and drug concentration. Surviving cells of the combined treatment showed a significant reduced ability to form colonies. Enhanced cell toxicity was associated with a 5.3-14.4-fold increase of the apoptotic rate and intracellular p53 level up to 50.4% following vector-mediated p53 restoration and bortezomib treatment. In contrast, an antagonistic effect on tumor cell growth and colony formation was observed for the combination of bortezomib and docetaxel or pemetrexed as a reduction of cell growth between 31 and 48% was found in comparison to 50% using the single agents. Lower cytotoxic effects were associated with significantly reduced apoptosis and an increase of clonogenic growth. The observed antagonistic effects between bortezomib and docetaxel or pemetrexed might influence clinical trials using these compounds. Conversely, p53 restoration and bortezomib treatment led to enhanced, synergistic tumor cell toxicity.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Acides boroniques/usage thérapeutique , Carcinome pulmonaire non à petites cellules/thérapie , Gènes p53/génétique , Thérapie génétique , Tumeurs du poumon/thérapie , Inhibiteurs de protéases/usage thérapeutique , Pyrazines/usage thérapeutique , Apoptose , Bortézomib , Carcinome pulmonaire non à petites cellules/traitement médicamenteux , Association thérapeutique , Docetaxel , Glutamates/usage thérapeutique , Guanine/analogues et dérivés , Guanine/usage thérapeutique , Humains , Tumeurs du poumon/traitement médicamenteux , Pémétrexed , Inhibiteurs du protéasome , Taxoïdes/usage thérapeutique , Transfection , Cellules cancéreuses en culture
19.
Hautarzt ; 57(2): 137-43, 2006 Feb.
Article de Allemand | MEDLINE | ID: mdl-15657729

RÉSUMÉ

We report on two female patients who presented with painful recurrent palpable purpura, ulcers and necroses on the extremities. The results of all examinations and laboratory tests considered together suggested a diagnosis of necrotizing leukocytoclastic vasculitis. Leukocytoclastic vasculitis is an inflammatory necrotizing condition of the superficial dermal vessels, presenting with variable clinical symptoms. In most cases it becomes manifest as palpable purpura, but hemorrhagic-necrotizing, bullous, nodular and urticarial presentations also occur. Common etiological factors include bacterial, viral or drug antigens, chronic infections (hepatitis B and C), non-Hodgkin lymphomas (monoclonal gammopathy, multiple myeloma), leukemia (hairy cell leukemia), and tumors (bronchial, breast, and gastric cancer) and also connective tissue disorders. In the course of the work-up, a plasmocytoma was discovered as the cause of the leukocytoclastic vasculitis, presenting in a similar way to livedo reticularis in one case and to pyoderma gangraenosum in the other.


Sujet(s)
Plasmocytome/complications , Plasmocytome/diagnostic , Tumeurs cutanées/complications , Tumeurs cutanées/diagnostic , Vascularite/diagnostic , Vascularite/étiologie , Sujet âgé , Diagnostic différentiel , Femelle , Humains , Mâle , Adulte d'âge moyen , Maladies rares/diagnostic , Maladies rares/étiologie
20.
Br J Haematol ; 130(4): 588-94, 2005 Aug.
Article de Anglais | MEDLINE | ID: mdl-16098074

RÉSUMÉ

We conducted a randomised trial comparing an intensified versus a standard conditioning regimen for high-dose chemotherapy followed by autologous stem-cell transplantation in patients with multiple myeloma. In this study, 56 patients were randomly assigned for high-dose therapy with melphalan 200 mg/m2 or with idarubicin 42 mg/m2, melphalan 200 mg/m2 and cyclophosphamide 120 mg/kg. The primary objective was response rate. Acute toxicity, mainly because of infections, was higher in the intensified treatment arm with a treatment-related mortality of 20% versus 0% in the standard arm. This lead to the early discontinuation of the study. Response rates did not differ significantly between both treatment arms {intensified versus standard: complete response+near complete remission 50% [95% confidence interval (CI) 26-74%] vs. 33% (95% CI 17-55%), partial remission 35% (95% CI 16-61%) vs. 50% (95% CI 30-70%)}. After a follow-up of 5 years, the median time-to-progression and overall survival were not significantly different between both patient groups. Analysis restricted to patients surviving the first 100 d after transplant showed a better outcome for patients with >or=2 bad prognostic risk factors in the intensified treatment arm, however all treatment-related deaths occurred within this group of patients. In conclusion, intensified conditioning for high-dose therapy had intolerably high toxicity without improving outcome in patients with multiple myeloma.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Myélome multiple/chirurgie , Transplantation de cellules souches/mortalité , Conditionnement pour greffe/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Cyclophosphamide/administration et posologie , Cyclophosphamide/usage thérapeutique , Survie sans rechute , Calendrier d'administration des médicaments , Femelle , Humains , Idarubicine/administration et posologie , Idarubicine/usage thérapeutique , Mâle , Melphalan/administration et posologie , Melphalan/usage thérapeutique , Adulte d'âge moyen , Myélome multiple/traitement médicamenteux , Myélome multiple/immunologie , Pronostic , Induction de rémission , Facteurs de risque , Taux de survie , Conditionnement pour greffe/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE