Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
2.
Leukemia ; 30(3): 555-61, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26522083

ABSTRACT

DNA methylation changes are a constant feature of acute myeloid leukemia. Hypomethylating drugs such as azacitidine are active in acute myeloid leukemia (AML) as monotherapy. Azacitidine monotherapy is not curative. The AML-AZA trial tested the hypothesis that DNA methyltransferase inhibitors such as azacitidine can improve chemotherapy outcome in AML. This randomized, controlled trial compared the efficacy of azacitidine applied before each cycle of intensive chemotherapy with chemotherapy alone in older patients with untreated AML. Event-free survival (EFS) was the primary end point. In total, 214 patients with a median age of 70 years were randomized to azacitidine/chemotherapy (arm-A) or chemotherapy (arm-B). More arm-A patients (39/105; 37%) than arm-B (25/109; 23%) showed adverse cytogenetics (P=0.057). Adverse events were more frequent in arm-A (15.44) versus 13.52 in arm-B, (P=0.26), but early death rates did not differ significantly (30-day mortality: 6% versus 5%, P=0.76). Median EFS was 6 months in both arms (P=0.96). Median overall survival was 15 months for patients in arm-A compared with 21 months in arm-B (P=0.35). Azacitidine added to standard chemotherapy increases toxicity in older patients with AML, but provides no additional benefit for unselected patients.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/therapeutic use , Induction Chemotherapy/methods , Leukemia, Myeloid, Acute/drug therapy , Aged , Cytarabine/therapeutic use , Cytogenetic Analysis , Daunorubicin/therapeutic use , Female , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Remission Induction , Survival Analysis
3.
Blood Cancer J ; 4: e219, 2014 Jun 13.
Article in English | MEDLINE | ID: mdl-24927408

ABSTRACT

Despite new treatment modalities, the clinical outcome in a substantial number of patients with multiple myeloma (MM) has yet to be improved. Antibody-based targeted therapies for myeloma patients could make use of the HM1.24 antigen (CD317), a surface molecule overexpressed on malignant plasma cells and efficiently internalized. Here, a novel immunotoxin, HM1.24-ETA', is described. HM1.24-ETA' was generated by genetic fusion of a CD317-specific single-chain Fv (scFv) antibody and a truncated variant of Pseudomonas aeruginosa exotoxin A (ETA'). HM1.24-ETA' inhibited growth of interleukin 6 (IL-6)-dependent and -independent myeloma cell lines. Half-maximal growth inhibition was observed at concentrations as low as 0.3 nM. Target cell killing occurred via induction of apoptosis and was unaffected in co-culture experiments with bone marrow stromal cells. HM1.24-ETA' efficiently triggered apoptosis of freshly isolated/cryopreserved cells of patients with plasma cell leukemia and MM and was active in a preclinical severe combined immunodeficiency (SCID) mouse xenograft model. Importantly, HM1.24-ETA' was not cytotoxic against CD317-positive cells from healthy tissue (monocytes, human umbilical vein endothelial cells). These results indicate that CD317 may represent a promising target structure for specific and efficient immunotoxin therapy for patients with plasma cell tumors.


Subject(s)
ADP Ribose Transferases/pharmacology , Bacterial Toxins/pharmacology , Exotoxins/pharmacology , Immunotoxins/pharmacology , Multiple Myeloma/drug therapy , Virulence Factors/pharmacology , ADP Ribose Transferases/chemistry , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/pharmacology , Antigens, CD/immunology , Apoptosis/drug effects , Apoptosis/immunology , Bacterial Toxins/chemistry , Epitopes , Exotoxins/chemistry , Female , GPI-Linked Proteins/immunology , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Immunotoxins/chemistry , Immunotoxins/immunology , Jurkat Cells , Mice , Mice, SCID , Multiple Myeloma/immunology , Multiple Myeloma/pathology , Virulence Factors/chemistry , Xenograft Model Antitumor Assays , Pseudomonas aeruginosa Exotoxin A
5.
Leukemia ; 27(1): 190-201, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22660187

ABSTRACT

Bispecific antibodies (bsab) offer a promising approach for optimizing antibody-based therapies. In the present study, [(CD20)(2)xCD16], a recombinant CD20- and CD16-directed bsab in the tribody format, was designed to optimize recruitment of FcγRIII (CD16)-positive effector cells. [(CD20)(2)xCD16] retained the antigen specificities of the parental monoclonal antibodies and binding to FcγRIIIa was not compromised by the F/V polymorphism at amino-acid position 158. [(CD20)(2)xCD16] mediated potent lysis of lymphoma cell lines and freshly isolated tumor cells from patients, even at low picomolar concentrations (∼10 pM). Irrespective of the CD16a allotype, potency as well as efficacy of lysis obtained with the tribody was significantly higher than lysis triggered by rituximab. Tumor cell killing also occurred when autologous NK cells were used as effector cells. Compared with rituximab, the tribody demonstrated depletion of autologous B cells in ex vivo whole blood assays at 100-fold lower antibody concentration. In mice with a reconstituted humanized hematopoietic system, established by transplantation of human CD34-positive cord blood cells, this novel tribody significantly depleted autologous human B cells. Thus, tribodies such as [(CD20)(2)xCD16], recruiting CD16-positive effector cells, may represent promising candidates for clinical development.


Subject(s)
Antibodies, Bispecific/therapeutic use , Antibody-Dependent Cell Cytotoxicity , Antigens, CD20/immunology , Leukemia, B-Cell/therapy , Lymphoma, B-Cell/therapy , Receptors, IgG/immunology , Adult , Aged , Aged, 80 and over , Animals , Animals, Newborn , Antibody Specificity , Female , Fetal Blood/cytology , Fetal Blood/metabolism , Humans , Killer Cells, Natural/immunology , Leukemia, B-Cell/immunology , Lymphocyte Depletion , Lymphoma, B-Cell/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Receptors, IgG/metabolism
6.
Ann Hematol ; 91(11): 1765-72, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22926531

ABSTRACT

In most patients, mantle cell lymphoma (MCL) shows an aggressive clinical course with a continuous relapse pattern and a median survival of only 3-5 years. In the current study generation of the European MCL Network, the addition of high-dose Ara-C to R-CHOP (rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone)-like regimen followed by myeloablative consolidation achieved a significant improvement of progression-free survival in younger patients. In elderly patients, rituximab maintenance led to a marked prolongation of remission duration. Emerging strategies include mammalian target of rapamycin (mTOR) inhibitors, proteasome inhibitors, immune modulatory drugs, Bruton's tyrosine kinase inhibitors and others, all based on the dysregulated control of cell cycle machinery and impairment of several apoptotic pathways. Combination strategies are currently being investigated in numerous trials, but their introduction into clinical practice and current treatment algorithms remains a challenge. In the current survey, the application of the molecular targeted compounds were collected and evaluated by a representative national network of 14 haematological institutions. Optimised strategies are recommended for clinical routine. Future studies will apply individualised approaches according to the molecular risk profile of the patient.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Evidence-Based Medicine , Lymphoma, Mantle-Cell/drug therapy , Adult , Age Factors , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Child , Consensus Development Conferences as Topic , Consolidation Chemotherapy/adverse effects , Consolidation Chemotherapy/methods , European Union , Health Care Surveys , Humans , Induction Chemotherapy/adverse effects , Induction Chemotherapy/methods , Lymphoma, Mantle-Cell/prevention & control , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/prevention & control , Survival Analysis
8.
Bone Marrow Transplant ; 46(1): 143-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20348971

ABSTRACT

The efficacy and safety of CD52 antibody alemtuzumab to treat severe acute GVHD in 18 consecutive patients refractory to standard high-dose corticosteroid therapy is reported. Patients (age range 13-68 years) had developed acute GVHD grade III and IV with gut and/or liver involvement after stem cell transplantation from family donors (n= 7) or HLA-matched unrelated donors (n=11), including five donors with one or two HLA mismatches. Initially, in three patients, start doses of alemtuzumab in the range of 70-80 mg were applied and repeated after 3 to 4 weeks. Impressive responses were seen, but virus reactivation and bacterial infections were frequent. In an attempt to reduce this complication, the next nine patients received a reduced starting dose of 20-33 mg, and the last six patients received 3-13 mg repeated every 2-3 weeks. Seventeen of 18 patients responded to alemtuzumab, six patients are alive with a median follow-up of 108 weeks. Chronic GVHD was observed frequently. Although pronounced lymphocyte depletion requiring close monitoring for signs of infections seems inevitable for efficacy, alemtuzumab given in moderate doses has a substantial activity not only in intestinal but also in severe acute GVHD of the liver.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Host Disease/immunology , Immunosuppressive Agents/therapeutic use , Adolescent , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Alemtuzumab , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/administration & dosage , Antibodies, Neoplasm/adverse effects , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Bacterial Infections/complications , CD52 Antigen , Gastrointestinal Diseases/complications , Gastrointestinal Diseases/drug therapy , Gastrointestinal Diseases/immunology , Glycoproteins/metabolism , Graft vs Host Disease/complications , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Leukemia, Myeloid, Acute/complications , Leukemia, Myeloid, Acute/therapy , Liver Diseases/complications , Liver Diseases/drug therapy , Liver Diseases/immunology , Lymphocyte Depletion/adverse effects , Middle Aged , Survival Analysis , Treatment Outcome , Virus Diseases/complications , Young Adult
10.
Eur J Med Res ; 12(8): 337-40, 2007 Aug 16.
Article in English | MEDLINE | ID: mdl-17933709

ABSTRACT

Systemic candidiasis is a rare but life threatening complication in immunosuppressed patients undergoing allogeneic SCT. Combination of new antifungal agents may improve outcome in this patient population. Here, triple anti-mycotic therapy is described in an relapsed ALL patient in urgent need of allogeneic bone marrow transplantation. The patient with T-cell acute lymphoblastic leukemia of thymic differentiation achieved remission after treatment according to the German ALL protocol 07/03. Two months after the consolidation therapy relapse occurred requiring high dose chemotherapy with allogeneic stem cell transplantation. One day after start of the conditioning regimen the patient showed skin manifestations typical for septic mycosis and blood cultures became positive for Candida krusei while on fluconazole prophylaxis. Because of the limited sensibility of fluconazole resistant candida species to liposomal amphotericin B and the high mortality rate in patients with systemic candidiasis, voriconazole was added immediately to liposomal amphotericin B. Since fever did not resolve and the conditioning therapy for allogeneic transplantation was not yet completed caspofungin was added. Skin manifestation responded to this triple anti-mycotic combination and peripheral blood stem cells from an unrelated donor were transplanted. With the triple antifungal therapy the patient finally became afebrile, skin manifestations showed complete resolution and blood cultures became negative. Three months after the onset of systemic candidiasis the patient was fully active with no signs of fungal infection and in haematological and molecular remission. Mycotic sepsis at the start of myeloablative conditioning therapy in heavily pretreated acute leukemia patients is usually considered as not allowing successful allogeneic transplantation. Thus this case demonstrates, that allogeneic stem cell transplantation is feasible in patients presenting with systemic candidiasis if combined antifungal therapy with liposomal amphotericin B, caspofungin and voriconazole is given.


Subject(s)
Antifungal Agents/therapeutic use , Bone Marrow Transplantation , Candidiasis/therapy , Hematopoietic Stem Cell Transplantation , Transplantation Conditioning , Adult , Amphotericin B/administration & dosage , Amphotericin B/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Candidiasis/immunology , Caspofungin , Drug Therapy, Combination , Echinocandins , Humans , Immunocompromised Host , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/therapy , Lipopeptides , Liposomes , Male , Peptides, Cyclic/therapeutic use , Pyrimidines/therapeutic use , Transplantation, Homologous , Triazoles/therapeutic use , Voriconazole
11.
Eur J Med Res ; 11(2): 73-6, 2006 Feb 21.
Article in English | MEDLINE | ID: mdl-16504964

ABSTRACT

There is now evidence that the tolerability and response to systemic chemotherapy in HIV-infected patients with AIDS-related lymphoma (ARL) is significantly improved by highly active antiretroviral therapy. Here we report an severely immunocompromised AIDS patient with recurrent ARL who was successfully treated with autologous stem cell transplantation (ASCT). We also review the current literature of ASCT in HIV-infected patients.


Subject(s)
B-Lymphocytes/pathology , Hematopoietic Stem Cell Transplantation , Immunocompromised Host , Lymphoma, AIDS-Related/therapy , Adult , HIV Infections/complications , HIV Infections/pathology , Humans , Lymphoma, AIDS-Related/pathology , Male , Transplantation, Autologous , Treatment Outcome
12.
Leukemia ; 19(4): 611-7, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15729385

ABSTRACT

Severe congenital neutropenia (CN) is characterized by a maturation arrest of myelopoiesis at the promyelocyte stage. Treatment with pharmacological doses of recombinant human granulocyte colony-stimulating factor (rh-G-CSF) stimulates neutrophil production and decreases the risk of major infectious complications. However, approximately 15% of CN patients develop myeloid malignancies that have been associated with somatic mutations in the G-CSF receptor (G-CSFR) and RAS genes as well as with acquired monosomy 7. We report a CN patient with chronic myelomonocytic leukemia (CMML) who never received rh-G-CSF. Molecular analysis demonstrated a somatic G-CSFR mutation (C2390T), which led to expression of a truncated G-CSFR protein in the CMML. Normal G-CSFR expression was unexpectedly absent in primary and cultured CMML. In addition, CMML cells showed monosomy 7 and an oncogenic NRAS mutation. In vitro culture revealed a G-CSF-dependent proliferation of CMML cells, which subsequently differentiated along the monocytic/macrophage lineage. Our results provide direct evidence for the in vivo expression of a truncated G-CSFR in leukemic cells, which emerged in the absence of rh-G-CSF treatment and transduces proliferative signals.


Subject(s)
Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/pathology , Neutropenia/genetics , Neutropenia/pathology , Receptors, Granulocyte Colony-Stimulating Factor/genetics , Adolescent , Cell Division , Genes, ras/genetics , Granulocyte Colony-Stimulating Factor/pharmacology , Humans , In Vitro Techniques , Male , Neutropenia/congenital , RNA, Messenger/metabolism , Tumor Cells, Cultured
13.
Ann Oncol ; 15(11): 1691-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15520073

ABSTRACT

BACKGROUND: Patients with follicular (FL) or mantle cell lymphoma (MCL) are incurable with conventional therapy. We investigated the safety and efficacy of rituximab consolidation after autologous stem cell transplantation (ASCT) in order to prevent relapse by clearance of minimal residual disease (MRD). METHODS: Rituximab was given approximately 8 weeks after CD34+ cell enriched ASCT at 375 mg/m2, weekly for 4 weeks. Monitoring of MRD was performed by repetitive PCR analyses. RESULTS: Thirty-one patients were included; one died early after ASCT before rituximab administration. Thirty patients (20 FL, 10 MCL) were evaluable after rituximab consolidation, and 27 of these were assessable for MRD detection. Rituximab consolidation post-ASCT was safe, the most common toxicity being infection. At a median follow-up of 42 months (range 13-96) after ASCT, 25 patients were censored with an actuarial event-free survival (EFS) of 81% at 4 and 5 years. Four patients (two FL, two MCL) relapsed, and one additional MCL patient died unexpectedly in complete remission. PCR-negativity was observed in 22% of the patients before ASCT, 53% post-ASCT (P=0.0547), 72% after rituximab (P=0.0018) and 100% at 6 months post-transplant (P < 0.001). CONCLUSIONS: One single course of rituximab consolidation given after ASCT is safe, may help to eliminate MRD and may translate into improved EFS in both FL and MCL patients.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Lymphoma, Follicular/drug therapy , Lymphoma, Mantle-Cell/drug therapy , Adult , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Murine-Derived , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Infections/chemically induced , Leukopenia/chemically induced , Lymphoma, Follicular/therapy , Lymphoma, Mantle-Cell/therapy , Male , Middle Aged , Neoplasm Staging , Peripheral Blood Stem Cell Transplantation , Postoperative Care , Rituximab , Survival Analysis , Transplantation, Autologous , Treatment Outcome
14.
J Drug Target ; 12(1): 11-8, 2004 Jan.
Article in English | MEDLINE | ID: mdl-15203907

ABSTRACT

Success of human gene therapy depends upon the development of delivery vehicles or vectors, which can selectively deliver therapeutic genes to target cells with efficiency and safety. Previous studies have shown an efficient, systemic trans-gene expression in many cell lines (in vitro) by using an anionic liposomal vector, based on the composition of retroviral envelopes (artificial viral envelopes, AVEs). The AVE-liposomes and their complexes with plasmid (DNA) were characterized according to zeta potential measurements and transmission electron microscopy (TEM). We successfully demonstrated that AVE liposomes, dispersed in 10% serum-containing growth medium, efficiently delivered plasmid DNA to HuH-7 (human hepatoma cell line) cells. We assessed the utility of liver-targeted vesicles as a drug/gene delivery system for the treatment of liver diseases. We found that small unilamellar AVE vesicles containing 15 mol% digalactosyl diglyceride (DGDG) are efficiently targeted to the liver via the hepatic asialoglycoprotein receptor.


Subject(s)
DNA/administration & dosage , Genetic Therapy/methods , Serum/chemistry , Carbohydrates/chemistry , Cell Line, Tumor , DNA/chemistry , Green Fluorescent Proteins , Humans , Liposomes , Liver/metabolism , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Membrane Lipids/chemistry , Microscopy, Electron , Molecular Mimicry , Plasmids/administration & dosage , Polyethyleneimine/chemistry , Retroviridae/chemistry , Transfection
15.
Br J Haematol ; 124(1): 47-54, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14675407

ABSTRACT

Chromosomal translocations are a characteristic feature of leukaemia and other malignant diseases. As clonal markers, they can be applied to identify and quantify the number of malignant cells by polymerase chain reaction (PCR) methods. The translocation t(4;11) is present in >60% of infant leukaemia. In order to facilitate the sequencing of chromosomal breakpoints, we developed an optimized set of 30 PCR primers and a new approach, designated as asymmetric multiplex PCR (am-PCR). Due to the high number of primers, small breakpoint-spanning DNA fragments are obtained in one nested multiplex PCR reaction. All PCR products contain an identical binding site for the initiation of direct sequencing. By using am-PCR, the translocation t(4;11) was examined in bone marrow and blood samples from children with acute leukaemia. Compared with previously described methods for the determination of genomic breakpoints, am-PCR may be advantageous with regard to its simplicity and rapidity. Breakpoint-spanning sequences were also evaluated with regard to their applicability as unique clonal markers to design primers and probes for minimal residual disease quantification by real-time PCR. This approach can easily be adapted to other chromosomal translocations in malignant diseases for the detection and analysis of clone-specific DNA markers.


Subject(s)
Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 4/genetics , Polymerase Chain Reaction/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Translocation, Genetic/genetics , Chromosome Breakage , Fusion Proteins, bcr-abl/genetics , Humans , Polymerase Chain Reaction/standards , Sensitivity and Specificity , Sequence Analysis, DNA/methods , Taq Polymerase
16.
Br J Cancer ; 89(12): 2234-43, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14676800

ABSTRACT

A phase I study of the bispecific antibody MDX-H210 in combination with granulocyte colony-stimulating factor (G-CSF) was performed in stage IV breast carcinoma patients, overexpressing HER-2/neu. MDX-H210, constructed by crosslinking antigen binding fragments (F(ab') fragments) of monoclonal antibody (mAb) H22 to Fc gamma receptor I (FcgammaRI), and mAb 520C9 to HER-2/neu, respectively, mediates the lysis of tumour cells in vitro, and in human FcgammaRI transgenic mouse models. The proto-oncogene HER-2/neu is overexpressed in approximately 30% of breast cancer patients, and represents a promising target for antibody-based immunotherapy. Fc gamma receptor I (CD64) is an effective trigger molecule, which is expressed on monocytes/macrophages, immature dendritic cells, and G-CSF-primed polymorphonuclear cells (PMN). Patients received G-CSF (Filgrastim) for 8 consecutive days, and cohorts of three patients were treated on day 4 with escalating, single doses of MDX-H210. A total of 30 patients were included, and treatment was generally well tolerated, without reaching dose-limiting toxicity. Side effects consisted mainly of fever and short periods of chills, which were timely related to elevated plasma levels of interleukin 6 and tumour necrosis factor alpha. In the last two cohorts, MDX-H210 plasma levels exceeded 1 microg ml(-1), and on circulating myeloid cells >50% saturation of FcgammaRI was found until day 4. These effector cells were highly effective in antibody-dependent cell-mediated cytotoxicity. Immunohistochemical analyses of tumour biopsies in individual patients documented infiltration of monocytes and PMN after MDX-H210 infusion. Although the clinical course of the disease was not altered by the single dose of MDX-H210, a favourable toxicity profile--even at high doses--and remarkable biological effects were seen when combined with G-CSF. Therefore, the combination of G-CSF and MDX-H210 should be evaluated in further immunotherapeutical strategies.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antibodies, Bispecific/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Granulocyte Colony-Stimulating Factor/therapeutic use , Adult , Aged , Antibodies, Bispecific/immunology , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Biomarkers, Tumor/blood , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cohort Studies , Cytokines/immunology , Female , Filgrastim , Genes, erbB-2/genetics , Granulocyte Colony-Stimulating Factor/immunology , Humans , Immunotherapy/methods , Maximum Tolerated Dose , Middle Aged , Neoplasm Staging , Proto-Oncogene Mas , Recombinant Proteins
17.
Horm Res ; 60(3): 143-7, 2003.
Article in English | MEDLINE | ID: mdl-12931042

ABSTRACT

OBJECTIVE: Autosomal dominant familial neurohypophyseal diabetes insipidus is a rare disorder characterized by polydipsia and polyuria. We present the results of the molecular analysis of the AVP-NPII gene of a German kindred. METHODS: All three exons of the gene were amplified by polymerase chain reaction and sequenced. RESULTS: In 7 affected individuals a new missense mutation (1770G > T) in exon 2 was found predicting a cysteine to phenylalanine substitution at codon 58 in the neurophysin II domain (NPII). CONCLUSION: As a result of this mutation a cysteine residue is exchanged, which is involved in a disulfide bond with cysteine 44 of the NPII moiety, hypothesizing that the resulting misfolded protein may lead to chronic neurotoxicity by accumulation of these products in the endoplasmatic reticulum.


Subject(s)
Arginine Vasopressin/genetics , Diabetes Insipidus, Neurogenic/genetics , Neurophysins/genetics , Adolescent , Adult , Aged , Base Sequence , Child , Female , Genetic Carrier Screening , Heterozygote , Humans , Male , Middle Aged , Molecular Sequence Data , Mutation, Missense , Pedigree
18.
Cytometry B Clin Cytom ; 53(1): 4-10, 2003 May.
Article in English | MEDLINE | ID: mdl-12717685

ABSTRACT

BACKGROUND: The goal of this study concerned the pretherapeutic identification of high-risk acute myeloid leukemia (AML) patients by data pattern analysis from flow cytometric immunophenotype, cytogenetic, and clinical data. METHODS: Sixty-seven parameters of AML patients at diagnosis were classified for predictive information by algorithmic data sieving using iteratively self optimizing triple matrix data pattern analysis (http://www.biochem.mpg.de/valet/classif1.html). RESULTS: Pretherapeutic predictive values for nonsurvival within five years and two years were 100.0% and 83.2%, respectively, compared to 13.9% and 47.4% for the prediction of survival at five years and two years, respectively. At diagnosis, five-year nonsurvivors showed increased patient age and higher concentration of cells in the analyzed specimen, as well as increased levels of % CD2, CD4, CD13, CD36, and CD45 positive AML blasts. Two-year nonsurvivors were characterized by a data pattern of increased patient age and levels of % CD4, CD7, CD11b, CD24, CD45, TH126, and HLA-DR positive AML blasts and decreased levels of % CD1, CD65, CD95, and TC25 positive AML blasts. Cytogenetic abnormalities were not selected for the optimized discriminatory data patterns. CONCLUSIONS: The comparatively accurate pretherapeutic identification of high-risk AML patients may prove useful for the development of individualized therapy protocols in stratified clinical patients groups.


Subject(s)
Flow Cytometry , Leukemia, Myeloid/mortality , Leukemia, Myeloid/pathology , Acute Disease , Computational Biology , Humans , Immunophenotyping , Predictive Value of Tests , Prognosis , Risk Factors , Survival Analysis
19.
Cytometry B Clin Cytom ; 53(1): 11-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12717686

ABSTRACT

BACKGROUND: Chromosomal abnormalities are one of the most important prognostic factors in acute myeloid leukemia (AML). However, only a limited number of patients have such informative chromosomal abnormalities. The prognostic value of immunophenotyping in this disease is still unclear. METHODS: Seven hundred and eighty-three newly diagnosed AML patients treated in the German SHG-AML trials in 1991 and 1996 were analyzed with a panel of 33 antibodies. Expression was correlated to overall survival, complete remission-rate, and complete remission duration, and tested in a multivariate analysis including other clinical and biological markers. RESULTS: With a median follow-up of 4.3 years, patients with AML blasts negative for CD9, CD11b, CD13, CD34, and CD41, or positive for CD15, CD33, CD38, CD64, and MPO had superior overall survival. This effect was associated with a significantly higher complete remission rate (CD13, CD34, CD41, and CD64) or a longer complete remission duration (CD9, CD11b, and CD64). Cox-regression analysis, including cytogenetic, morphologic, and biologic parameters showed CD9, CD13, CD34, and CD64 as independent factors for overall survival. These markers were used for a prognostic score. Patients were pooled in three groups with highly significant differences of overall survival. The prognostic relevance of this score was confirmed in patients with normal karyotype and/or in younger patients

Subject(s)
Immunophenotyping/methods , Leukemia, Myeloid/mortality , Leukemia, Myeloid/pathology , Acute Disease , Adolescent , Adult , Aged , Antibiotics, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/administration & dosage , Cytarabine/administration & dosage , Daunorubicin/administration & dosage , Female , Humans , Leukemia, Myeloid/drug therapy , Male , Middle Aged , Multivariate Analysis , Predictive Value of Tests , Prognosis , Retrospective Studies , Risk Factors , Survival Analysis
20.
Ann Rheum Dis ; 61(12): 1065-70, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12429536

ABSTRACT

BACKGROUND: The survival rate in patients with systemic lupus erythematosus (SLE) has improved dramatically during the past four decades to 96.6% (five year) in the Erlangen cohort, but it is nearly three times as high as in an age and sex matched control population. Reasons for death are mainly cardiovascular diseases (37%) and infections (29%). OBJECTIVE: To find risk factors existing at disease onset for a severe outcome in the Erlangen cohort. PATIENTS AND METHODS: By using a database of 338 patients with SLE from a single centre, documented at least one to 15 years and including Systemic Lupus International Collaborating Clinics/American College of Rheumatology (SLICC/ACR) damage score data and index (SDI) and an activity score (European Consensus Lupus Activity Measurement (ECLAM)), a retrospective search was made for risk factors for a severe outcome like death, end stage renal disease (ESRD), and thromboembolic events (TE) in SLE. For this purpose, multivariable Cox regression models were analysed using the statistical package SPSS 10.0 for Windows. RESULTS: The following were defined as risk factors for death at disease onset: male sex (p<0.001, relative risk (RR)=3.5), age >40 at disease onset (p<0.0001, RR=19.9), nephritis (p<0.05, RR=1.6), a reduction of creatinine clearance (p<0.001, RR=1.8), heart disease (p=0.05, RR=1.5), and central nervous system (CNS) disease (p=0.06, RR=1.6). An increase in the SDI of two or more points from the first to the third year of disease was the worst prognostic factor (p<0.0001, RR=7.7). The existence of Ro or nRNP antibodies, or both, was protective (p<0.05, RR =0.1). A low C3 (p<0.01 RR=3.0) and splenomegaly (p<0.01 RR=2.7) at disease onset turned out to be risk factors for ESRD besides a nephritis. In patients with hypertension (p<0.05) and/or high titres of dsDNA antibodies (>70 U/l) (p<0.01) and/or a mean ECLAM score of 4 (p<0.01) in the course of disease, a prevalence of ESRD was recorded in 9% (p<0.05) and 10% (p<0.01), and 8% (p<0.01) v 4% in the whole group. Analysis of risk factors at disease onset for TE identified positive lupus anticoagulant (p=0.17, RR=1.6), cryoglobulins (p<0.05, RR=1.8), and nephritis (p=0.05, RR=1.4), in addition to an age >40 at disease onset. CONCLUSIONS: A subgroup of patients in the Erlangen cohort with a typical clinical and serological phenotype at disease onset that is at high risk for a worse outcome was identified. Identification of these white patients at risk at disease onset will enable treatment to be intensified and thereby possibly prevent or better control late stage manifestations.


Subject(s)
Kidney Failure, Chronic/mortality , Lupus Erythematosus, Systemic/mortality , Thromboembolism/mortality , Adult , Age Factors , Central Nervous System Diseases/complications , Cohort Studies , Creatinine/metabolism , Female , Heart Diseases/complications , Humans , Kidney Failure, Chronic/complications , Lupus Erythematosus, Systemic/complications , Male , Multivariate Analysis , Nephritis/complications , Nephritis/mortality , Risk Factors , Sex Factors , Thromboembolism/complications
SELECTION OF CITATIONS
SEARCH DETAIL
...