Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
Add more filters

Publication year range
1.
Eur J Clin Invest ; 38(8): 578-84, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18717827

ABSTRACT

BACKGROUND: A mutation of Janus kinase 2 V617F is present in most patients with polycythaemia vera (PV). However, it is generally believed that JAK2(V617F) is not the sole molecular abnormality in PV. Since dasatinib is currently evaluated in patients with PV, it is of interest to study the effects of dasatinib on the growth of clonal progenitor cells in vitro. DESIGN AND METHODS: Peripheral blood mononuclear cells from patients with PV, chronic myeloid leukaemia (CML) and controls were exposed to dasatinib (0.1 to 500 nm mL(-1)). Colony growth was stimulated by interleukin-3, granulocyte-macrophage colony-stimulating factor and erythropoietin. Endogenous erythroid colony (EEC) growth was investigated without exogenous cytokines. Real-time PCR was performed to assess the percentage of JAK2(V617F) cells. RESULTS: 10 nm of dasatinib suppressed EEC growth from PV by 89% (P = 0.002). This inhibition was dose dependent and occurred at pharmacological concentrations. Erythroid and myeloid colony growth was also significantly suppressed in the presence of exogenous cytokines. When compared to PV the inhibition of stimulated colony growth was significantly less pronounced in controls but tended to be more vigorous in CML. Interestingly, despite the potent inhibition of PV cells real-time PCR revealed that the numbers of JAK2(V617F) transcripts did not decrease upon exposure to dasatinib. CONCLUSION: This study shows a marked inhibition of the proliferative capacity of progenitor cells from PV. Although JAK2(V617F) transcript levels did not decrease upon exposure to dasatinib, the drug might suppress PV progenitors through inhibition of a yet undefined molecular target.


Subject(s)
Cell Proliferation/drug effects , Janus Kinase 2/antagonists & inhibitors , Polycythemia Vera/pathology , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology , Adult , Aged , Aged, 80 and over , Cells, Cultured , Dasatinib , Dose-Response Relationship, Drug , Erythroid Precursor Cells/pathology , Female , Humans , Janus Kinase 2/genetics , Male , Middle Aged , Myeloid Progenitor Cells/pathology , Polycythemia Vera/genetics , Reverse Transcriptase Polymerase Chain Reaction
2.
Eur J Clin Invest ; 38(1): 53-60, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18173551

ABSTRACT

BACKGROUND: Presenting the same histological diagnosis, multiple myeloma (MM) shows a large genomic variety, resulting in variable times of overall survival. MATERIALS AND METHODS: To investigate major cytogenetic categories (any 14q-translocation, t(11;14), t(4;14), 13q-deletions, 17p-deletions) and their clinical consequences in MM after a pre-existing monoclonal gammopathy (MM post-MGUS), we performed a comparative analysis of 41 patients with MM post-MGUS and 287 patients with unknown prior history MM (U-MM). RESULTS: In MM post-MGUS, a t(11;14) was found to be more frequent than in U-MM (24% vs. 14%) and it was associated with significantly shortened survival (24 months vs. 70 months in U-MM; P = 0.01). MM post-MGUS was further characterized by a higher frequency of 13q-deletions only (absence of all other specific abnormalities; 28% vs. 12% in U-MM; P = 0.02). A 13q-deletion only was an indicator of long survival in MM post-MGUS (median not yet reached) as opposed to U-MM (median survival, 29 months; P = 0.001). 17p-deletions were infrequent in MM post-MGUS (3% vs. 16% in U-MM; P = 0.04). Survival times for patients with t(4;14) and/or 17p-deletions and other abnormalities were similar in both MM patient cohorts. CONCLUSIONS: Our data suggest that t(11;14) and 13q-deletions have distinct prognostic implications in the context of MM post-MGUS.


Subject(s)
Chromosome Deletion , Monoclonal Gammopathy of Undetermined Significance/complications , Multiple Myeloma/genetics , Translocation, Genetic , Adult , Aged , Aged, 80 and over , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 14 , Female , Humans , In Situ Hybridization, Fluorescence , Male , Middle Aged , Multiple Myeloma/complications , Prognosis , Survival Rate
3.
Leukemia ; 21(2): 277-80, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17251900

ABSTRACT

A widely accepted definition of resistance or intolerance to hydroxyurea (HU) in patients with essential thrombocythemia (ET) is lacking. An international working group (WG) was convened to develop a consensus formulation of clinically significant criteria for defining resistance/intolerance to HU in ET. To this aim, an analytic hierarchy process (AHP), a multiple-attribute decision-making technique, was used. The steps consisted of selecting the candidate criteria for defining resistance/intolerance; identifying the motivations that could influence the preference of the WG for any individual criterion; comparing the candidate criteria in a pair-wise manner; and grading them according their ability to fulfill the motivations. Every step in the model was derived by questionnaires or group discussion. The WG proposed that the definition of resistance/intolerance should require the fulfillment of at least one of the following criteria: platelet count greater than 600,000/micro l after 3 months of at least 2 g/day of HU (2.5 g/day in patients with a body weight over 80 kg); platelet count greater than 400,000/micro l and WBC less than 2500/micro l or Hb less than 10 g/dl at any dose of HU; presence of leg ulcers or other unacceptable muco-cutaneous manifestations at any dose of HU; HU-related fever.


Subject(s)
Hydroxyurea/therapeutic use , Thrombocythemia, Essential/drug therapy , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Consensus Development Conferences as Topic , Drug Resistance , Humans , Hydroxyurea/adverse effects , Patient Selection , Reproducibility of Results
4.
Leukemia ; 32(2): 450-461, 2018 02.
Article in English | MEDLINE | ID: mdl-28744014

ABSTRACT

The multistep process of TP53 mutation expansion during myeloproliferative neoplasm (MPN) transformation into acute myeloid leukemia (AML) has been documented retrospectively. It is currently unknown how common TP53 mutations with low variant allele frequency (VAF) are, whether they are linked to hydroxyurea (HU) cytoreduction, and what disease progression risk they carry. Using ultra-deep next-generation sequencing, we examined 254 MPN patients treated with HU, interferon alpha-2a or anagrelide and 85 untreated patients. We found TP53 mutations in 50 cases (0.2-16.3% VAF), regardless of disease subtype, driver gene status and cytoreduction. Both therapy and TP53 mutations were strongly associated with older age. Over-time analysis showed that the mutations may be undetectable at diagnosis and slowly increase during disease course. Although three patients with TP53 mutations progressed to TP53-mutated or TP53-wild-type AML, we did not observe a significant age-independent impact on overall survival during the follow-up. Further, we showed that complete p53 inactivation alone led to neither blast transformation nor HU resistance. Altogether, we revealed patient's age as the strongest factor affecting low-burden TP53 mutation incidence in MPN and found no significant age-independent association between TP53 mutations and hydroxyurea. Mutations may persist at low levels for years without an immediate risk of progression.


Subject(s)
Hydroxyurea/administration & dosage , Janus Kinase 2/genetics , Mutation/genetics , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Tumor Suppressor Protein p53/genetics , Adult , Aged , Aged, 80 and over , Alleles , Disease Progression , Female , Gene Frequency/drug effects , Gene Frequency/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Mutation/drug effects , Retrospective Studies , Young Adult
5.
Leukemia ; 31(4): 896-902, 2017 04.
Article in English | MEDLINE | ID: mdl-27773929

ABSTRACT

RBC-transfusion dependence is common in persons with myeloproliferative neoplasm (MPN)-associated myelofibrosis. The objective of this study was to determine the rates of RBC-transfusion independence after therapy with pomalidomide vs placebo in persons with MPN-associated myelofibrosis and RBC-transfusion dependence. Two hundred and fifty-two subjects (intent-to-treat (ITT) population) including 229 subjects confirmed by central review (modified ITT population) were randomly assigned (2:1) to pomalidomide or placebo. Trialists and subjects were blinded to treatment allocation. Primary end point was proportion of subjects achieving RBC-transfusion independence within 6 months. One hundred and fifty-two subjects received pomalidomide and 77 placebo. Response rates were 16% (95% confidence interval (CI), 11, 23%) vs 16% (8, 26%; P=0.87). Response in the pomalidomide cohort was associated with ⩽4 U RBC/28 days (odds ratio (OR)=3.1; 0.9, 11.1), age ⩽65 (OR=2.3; 0.9, 5.5) and type of MPN-associated myelofibrosis (OR=2.6; 0.7, 9.5). Responses in the placebo cohort were associated with ⩽4 U RBC/28 days (OR=8.6; 0.9, 82.3), white blood cell at randomization >25 × 109/l (OR=4.9; 0.8, 28.9) and interval from diagnosis to randomization >2 years (OR=4.9; 1.1, 21.9). Pomalidomide was associated with increased rates of oedema and neutropenia but these adverse effects were manageable. Pomalidomide and placebo had similar RBC-transfusion-independence response rates in persons with MPN-associated RBC-transfusion dependence.


Subject(s)
Immunologic Factors/therapeutic use , Myeloproliferative Disorders/complications , Primary Myelofibrosis/etiology , Primary Myelofibrosis/therapy , Thalidomide/analogs & derivatives , Adult , Aged , Aged, 80 and over , Biomarkers , Erythrocyte Transfusion/methods , Female , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Male , Middle Aged , Myeloproliferative Disorders/diagnosis , Phenotype , Primary Myelofibrosis/diagnosis , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/therapeutic use , Treatment Outcome , Workflow
6.
Blood Rev ; 30(6): 453-459, 2016 11.
Article in English | MEDLINE | ID: mdl-27341755

ABSTRACT

Clinical evidence supports the need of changing the diagnostic criteria of the 2008 updated WHO classification for polycythemia vera (PV), essential thrombocythemia (ET) and primary myelofibrosis (PMF). In JAK2-mutated patients who show characteristic bone marrow (BM) morphology, clinical studies demonstrated that a hemoglobin level of 16.5g/dL in men and 16.0g/dl for women or a hematocrit value of 49% in men and 48% in women are the optimal cut off levels for distinguishing JAK2-mutated ET from "masked/prodromal" PV. Therefore BM morphology was upgraded to a major diagnostic criterion. Regarding ET the key issue was to improve standardization of prominent BM features enhancing differentiation between "true" ET and prefibrotic/early primary myelofibrosis (prePMF). These two entities have shown a different epidemiology and clinical outcomes. Concerning prePMF a more explicit clinical characterization of minor criteria is mandated for an improved distinction from ET and overt PMF and accurate diagnosis and outcome prediction.


Subject(s)
Myeloproliferative Disorders/diagnosis , Practice Guidelines as Topic , Disease Susceptibility , Humans , Myeloproliferative Disorders/etiology , World Health Organization
7.
Leukemia ; 30(8): 1701-7, 2016 08.
Article in English | MEDLINE | ID: mdl-27211272

ABSTRACT

Ruxolitinib is a Janus kinase (JAK) (JAK1/JAK2) inhibitor that has demonstrated superiority over placebo and best available therapy (BAT) in the Controlled Myelofibrosis Study with Oral JAK Inhibitor Treatment (COMFORT) studies. COMFORT-II was a randomized (2:1), open-label phase 3 study in patients with myelofibrosis; patients randomized to BAT could crossover to ruxolitinib upon protocol-defined disease progression or after the primary end point, confounding long-term comparisons. At week 48, 28% (41/146) of patients randomized to ruxolitinib achieved ⩾35% decrease in spleen volume (primary end point) compared with no patients on BAT (P<0.001). Among the 78 patients (53.4%) in the ruxolitinib arm who achieved ⩾35% reductions in spleen volume at any time, the probability of maintaining response was 0.48 (95% confidence interval (CI), 0.35-0.60) at 5 years (median, 3.2 years). Median overall survival was not reached in the ruxolitinib arm and was 4.1 years in the BAT arm. There was a 33% reduction in risk of death with ruxolitinib compared with BAT by intent-to-treat analysis (hazard ratio (HR)=0.67; 95% CI, 0.44-1.02; P=0.06); the crossover-corrected HR was 0.44 (95% CI, 0.18-1.04; P=0.06). There was no unexpected increased incidence of adverse events with longer exposure. This final analysis showed that spleen volume reductions with ruxolitinib were maintained with continued therapy and may be associated with survival benefits.


Subject(s)
Primary Myelofibrosis/drug therapy , Pyrazoles/administration & dosage , Adult , Aged , Aged, 80 and over , Cross-Over Studies , Disease Progression , Female , Follow-Up Studies , Humans , Male , Middle Aged , Nitriles , Organ Size/drug effects , Primary Myelofibrosis/mortality , Pyrimidines , Spleen , Survival Rate
8.
Leukemia ; 30(5): 1126-32, 2016 05.
Article in English | MEDLINE | ID: mdl-26710883

ABSTRACT

Essential thrombocythemia (ET) is currently diagnosed either by the British Committee of Standards in Haematology (BCSH) criteria that are predominantly based on exclusion and not necessarily on bone marrow (BM) morphology, or the World Health Organization (WHO) criteria that require BM examination as essential criterion. We studied the morphological and clinical features in patients diagnosed according either to the BCSH (n=238) or the WHO guidelines (n=232). The BCSH-defined ET cohort was re-evaluated by applying the WHO classification. At presentation, patients of the BCSH group showed significantly higher values of serum lactate dehydrogenase and had palpable splenomegaly more frequently. Following the WHO criteria, the re-evaluation of the BCSH-diagnosed ET cohort displayed a heterogeneous population with 141 (59.2%) ET, 77 (32.4%) prefibrotic primary myelofibrosis (prePMF), 16 (6.7%) polycythemia vera and 4 (1.7%) primary myelofibrosis. Contrasting WHO-confirmed ET, the BCSH cohort revealed a significant worsening of fibrosis-free survival and prognosis. As demonstrated by the clinical data and different outcomes between WHO-diagnosed ET and prePMF, these adverse features were generated by the inadvertent inclusion of prePMF to the BCSH group. Taken together, the diagnosis of ET without a scrutinized examination of BM biopsy specimens will generate a heterogeneous cohort of patients impairing an appropriate clinical management.


Subject(s)
Bone Marrow/pathology , Practice Guidelines as Topic/standards , Thrombocythemia, Essential/diagnosis , Academies and Institutes , Adolescent , Adult , Aged , Aged, 80 and over , Biopsy , Bone Marrow Examination , Humans , L-Lactate Dehydrogenase/blood , Middle Aged , Prognosis , Splenomegaly , World Health Organization , Young Adult
9.
Leukemia ; 30(5): 1018-24, 2016 05.
Article in English | MEDLINE | ID: mdl-26854026

ABSTRACT

The Philadelphia-negative myeloproliferative neoplasms (MPNs) are clonal disorders involving hematopoietic stem and progenitor cells and are associated with myeloproliferation, splenomegaly and constitutional symptoms. Similar signs and symptoms can also be found in patients with chronic inflammatory diseases, and inflammatory processes have been found to play an important role in the pathogenesis and progression of MPNs. Signal transduction pathways involving JAK1, JAK2, STAT3 and STAT5 are causally involved in driving both the malignant cells and the inflammatory process. Moreover, anti-inflammatory and immune-modulating drugs have been used successfully in the treatment of MPNs. However, to date, many unresoved issues remain. These include the role of somatic mutations that are present in addition to JAK2V617F, CALR and MPL W515 mutations, the interdependency of malignant and nonmalignant cells and the means to eradicate MPN-initiating and -maintaining cells. It is imperative for successful therapeutic approaches to define whether the malignant clone or the inflammatory cells or both should be targeted. The present review will cover three aspects of the role of inflammation in MPNs: inflammatory states as important differential diagnoses in cases of suspected MPN (that is, in the absence of a clonal marker), the role of inflammation in MPN pathogenesis and progression and the use of anti-inflammatory drugs for MPNs. The findings emphasize the need to separate the inflammatory processes from the malignancy in order to improve our understanding of the pathogenesis, diagnosis and treatment of patients with Philadelphia-negative MPNs.


Subject(s)
Inflammation/drug therapy , Myeloproliferative Disorders/drug therapy , Neoplasms/pathology , Anti-Inflammatory Agents/therapeutic use , Clone Cells/pathology , Humans , Myeloproliferative Disorders/pathology
10.
J Clin Oncol ; 18(4): 804-12, 2000 Feb.
Article in English | MEDLINE | ID: mdl-10673522

ABSTRACT

PURPOSE: Recent metaphase cytogenetic studies suggested that specific chromosomal abnormalities are of prognostic significance in patients with multiple myeloma (MM). Because the true incidence of chromosomal abnormalities in MM is much higher than that detected by metaphase analysis, we used interphase fluorescence in situ hybridization (FISH) to determine the prognostic value of specific chromosomal aberrations. PATIENTS AND METHODS: Bone marrow plasma cells from 89 previously untreated patients with MM were studied consecutively by FISH to detect the deletions of 13q14, 17p13, and 11q and the presence of t(11;14)(q13;q32). FISH results were analyzed in the context of clinical parameters (response to treatment and survival after conventional-dose chemotherapy), and a multivariate analysis of prognostic factors was performed. RESULTS: By FISH, the deletion of 13q14 occurred in 40 patients (44.9%), deletion of 17p13 in 22 (24.7%), and 11q abnormalities in 14 (15.7%; seven with t(11;14)). Deletions of 13q14 and 17p13 were associated with poor response to induction treatment (46.9% v 77.3% in those without deletions, P =.006 and 40.0% v 73.2%, P =.008, respectively) and short median overall survival (OS) time (24.2 v 88.1 months, P =. 008 and 16.2 v 51.3 months, P =.008, respectively). Short median OS time was also observed for patients with 11q abnormalities (13.1 v 41.6 months, P =.02). According to the number of unfavorable cytogenetic features (deletion of 13q14, deletion of 17p13, and aberrations of 11q) that were present in each patient (0 v 1 v 2 or 3), patients with significantly different OS times could be discriminated from one another (102.4 v 29.6 v 13.9 months, P <.001, respectively). CONCLUSION: For patients with MM who were treated with conventional-dose chemotherapy, interphase FISH for 13q14, 17p13, and 11q provides prognostically relevant information in addition to that provided by standard prognostic factors. This observation may be considered for risk-adapted stratifications of MM patients in future clinical trials.


Subject(s)
Chromosome Aberrations/genetics , Interphase/genetics , Multiple Myeloma/genetics , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bone Marrow Cells/pathology , Chromosome Deletion , Chromosome Disorders , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 17/genetics , Cytogenetics , Female , Forecasting , Humans , In Situ Hybridization, Fluorescence , Incidence , Male , Metaphase/genetics , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/pathology , Multivariate Analysis , Plasma Cells/pathology , Prognosis , Regression Analysis , Remission Induction , Survival Rate , Trisomy/genetics
11.
Leukemia ; 11 Suppl 5: S52-6, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9436940

ABSTRACT

Response rates of 20% were achieved when interferon alpha (IFN) was applied as single agent treatment for multiple myeloma. A synergistic activity was observed when IFN was combined with chemotherapeutic agents in vitro and in vivo. These observations led to a series of randomized trials comparing chemotherapy alone with the combination of IFN and chemotherapy which yielded controversial results. Ludwig et al have analyzed presently available data of randomized trials in a systematic overview. In 16 trials, 2286 patients were randomized either for induction therapy with chemotherapy or IFN combined with chemotherapy. The IFN dose ranged between 4.8 and 18.7 MU/week. The overall response rate was 45.9% for patients treated with chemotherapy alone compared to 54.4% for patients treated with IFN alpha and chemotherapy. The gain of relapse-free survival was 6 months and the gain of overall survival was 5 months for IFN-treated patients. Advances in treatment were due to the expense of more severe side-effects in IFN-treated patients expressed by hematological side-effects, fever, nausea and neurological as well as psychological alterations. IFN maintenance therapy was evaluated from eight trials comprising 929 patients randomized for IFN-treatment or 'wait and see'. IFN maintenance treatment prolonged the average relapse-free survival by 7 months and prolonged the average overall survival by 5 months. Younger patients, patients in good performance status and patients with lower tumor burden seem to benefit most from the addition of IFN to chemotherapy or from IFN maintenance therapy. In conclusion, IFN shows a minor beneficial effect on response rates, progression-free and overall survival in the above mentioned patient group and might be applied when high-dose chemotherapy with transplantation is not feasible. IFN has been shown to be effective for maintenance therapy in patients with lower tumor burden who have responded to induction therapy.


Subject(s)
Interferon-alpha/therapeutic use , Multiple Myeloma/therapy , Clinical Trials as Topic , Disease-Free Survival , Humans , Interferon alpha-2 , Interferon-alpha/adverse effects , Multiple Myeloma/mortality , Recombinant Proteins , Survival Rate
12.
Leukemia ; 4(2): 91-4, 1990 Feb.
Article in English | MEDLINE | ID: mdl-2106057

ABSTRACT

The myelodysplastic syndrome (MDS) is characterized by a high probability of leukemic transformation and frequently lethal infections or bleeding episodes. Up to now, no generally accepted form of therapy has been established for MDS. Previous trials with alpha-interferon (IFN-alpha) have shown some beneficial effects. We studied the effects of long-term application of IFN-alpha at higher dosages in patients with "low-risk" MDS. Ten patients were included in the study; eight were treated for a period of 6-36 months. IFN-alpha was administered at a median dosage of 9, 6, 4 mU/week during the first, second, and third year, respectively. Response was determined by the status of peripheral blood and bone marrow. Prolonged exposure to IFN resulted in a response rate of 3/10 (30%). In an additional case, disease progression was retarded during the third year of therapy. The incidences of infections and bleeding events subsided notably. After the withdrawal of IFN, hematological and clinical parameters rapidly deteriorated in some patients. The observed improvement of the patients' susceptibility to infections possibly prolongs their survival and seems to justify further trials on IFN treatment in patients with MDS.


Subject(s)
Interferon Type I/therapeutic use , Myelodysplastic Syndromes/therapy , Aged , Female , Hematopoiesis , Hemoglobins/analysis , Humans , Infection Control , Interferon Type I/adverse effects , Male , Middle Aged , Myelodysplastic Syndromes/blood
13.
Leukemia ; 18(11): 1879-82, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15385925

ABSTRACT

Molecular and genetic events associated with the transition from monoclonal gammopathy of undetermined significance (MGUS) to multiple myeloma (MM) are still poorly characterized. We investigated serial bone marrow specimens from 11 patients with MGUS who eventually progressed to MM (MM post-MGUS) by interphase fluorescence in situ hybridization for immunoglobulin heavy-chain gene (IgH) translocations and chromosome 13q deletions (del(13q)). In nine patients, IgH translocations were present both in MGUS and MM post-MGUS plasma cells, including three t(11;14)(q13;q32) and one t(4;14)(p16;q32), which was observed already 92 months prior to MM. Similarly, all five MM patients with del(13q) had this aberration already at the MGUS stage. Two patients without IgH translocation and del(13q) had chromosomal gains suggesting hyperdiploidy, but IgH translocations and/or del(13q) did not emerge at MM post-MGUS. IgH translocations and del(13q) are early genetic events in monoclonal gammopathies, suggesting that additional events are required for the transition from stable MGUS to progressive MM.


Subject(s)
Chromosomes, Human, Pair 13 , Gene Deletion , Immunoglobulin Heavy Chains/genetics , Multiple Myeloma/genetics , Paraproteinemias/genetics , Translocation, Genetic , Aged , Bone Marrow , Disease Progression , Humans , In Situ Hybridization, Fluorescence , Interphase , Middle Aged
14.
Leukemia ; 14(11): 1975-9, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11069034

ABSTRACT

Since deletion of chromosome 13q is a clinically relevant feature in multiple myeloma (MM), we analyzed bone marrow plasma cells from 29 patients with monoclonal gammopathy of undetermined significance (MGUS) to investigate the chromosome 13 status in MGUS. Studies were performed by interphase fluorescence in situ hybridization (FISH) with a panel of 13q14-specific probes (RB1, D13S319, D13S25, D13S31). Plasma cells with a deletion of at least one of the 13q14 loci were detected in 13 patients (44.8%) with MGUS. In five patients (17.2%), deletions of all four 13q14-specific probes were observed, and the additional deletion of a 13q telomeric region (D13S327) suggested loss of the entire 13q arm or monosomy 13. Loss of 13q14 was observed to be monoallelic and to occur in 11.0 to 35.0% of plasma cells (cut-off levels for a deletion <10% with all probes). Nine of 17 patients (52.9%) with MM progressing from a pre-existing MGUS had evidence for a deletion of 13q14 as determined by FISH with the RB1 probe. These results suggest that deletion of 13q14 is an early event in the development of monoclonal gammopathies, but its role for the eventual progression to MM remains to be determined prospectively.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 13/ultrastructure , Paraproteinemias/genetics , Adult , Aged , Aged, 80 and over , Chromosomes, Human, Pair 13/genetics , Disease Progression , Follow-Up Studies , Humans , In Situ Hybridization, Fluorescence , Interphase , Middle Aged , Monosomy , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Paraproteinemias/pathology , Paraproteins/analysis , Plasma Cells/ultrastructure
15.
Leukemia ; 12(8): 1182-6, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9697871

ABSTRACT

Among 379 patients with AML with FAB type M1, 2 and M4-7 diagnosed between 1978 and 1997 in our institution, 19 (5%) had hypofibrinogenemia (HF), ie a fibrinogen level <180 mg/dl. Compared to patients with normal fibrinogen (n = 360) patients with HF had significantly elevated markers of activation of coagulation (TAT, F1.2, FPA) and fibrinolysis (D-dimer, FDP) indicating that disseminated intravascular coagulation/hyperfibrinolysis was the cause of hypofibrinogenemia. Patients with HF had significantly longer prothrombin times, thrombin clotting and reptilase times. Factor X and VIII were significantly lower than in patients without HF. With the exception of M7, HF occurred in all FAB subtypes, but was most common in M5 (12.1%). Patients with HF did not differ from those with normal fibrinogen with regard to age, sex, leukocyte count and other hematological parameters. During induction chemotherapy fibrinogen normalized rapidly (median 5 days) and there was no increased incidence of early hemorrhagic death. The overall and disease-free survival was similar to that of patients without HF.


Subject(s)
Blood Coagulation Disorders/complications , Fibrinogen/metabolism , Leukemia, Myeloid, Acute/complications , Adult , Antineoplastic Agents/therapeutic use , Blood Coagulation Disorders/epidemiology , Disease-Free Survival , Female , Humans , Incidence , Karyotyping , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Neoplasm Proteins/genetics , Oncogene Proteins, Fusion/genetics , Prognosis , Tretinoin/therapeutic use
16.
Clin Cancer Res ; 5(9): 2426-30, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10499614

ABSTRACT

Expression of the lung resistance protein (LRP) is associated with resistance to various anticancer drugs including melphalan and, therefore, may affect the clinical outcome in multiple myeloma (MM). To determine the clinical significance of LRP, we have compared LRP expression in bone marrow plasma cells with clinical parameters including response to chemotherapy and survival of previously untreated patients with MM (n = 72). LRP expression immunocytochemically assessed by means of the LRP-56 monoclonal antibody was positive (> or =10% staining plasma cells) in 44 (61%) samples. There was no correlation between LRP expression and age, sex, type of the paraprotein, serum creatinine, stage, beta2-microglobulin, serum lactate dehydrogenase, or C-reactive protein. However, LRP expression was more frequently observed in patients with a p53 deletion than in those without such a deletion (P = 0.01). The overall response rate for all of the patients evaluable for response to induction chemotherapy (n = 58) was 67%. The response rate was 87% for patients without LRP expression but only 54% for patients with LRP expression (P = 0.01). Kaplan-Meier analysis revealed that patients with LRP expression had a shorter overall survival (median, 33 months) than those without LRP expression (median not reached; P = 0.04). These data show that LRP expression is an important marker for clinical drug resistance and predicts a poor outcome in MM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Neoplasm Proteins/biosynthesis , Vault Ribonucleoprotein Particles/biosynthesis , Adult , Aged , Aged, 80 and over , Drug Resistance, Neoplasm , Female , Humans , Immunohistochemistry , Male , Middle Aged , Multiple Myeloma/blood , Multiple Myeloma/pathology , Prognosis , Survival Analysis , Treatment Outcome
17.
Leukemia ; 29(1): 20-6, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25151955

ABSTRACT

The discovery of somatic mutations, primarily JAK2V617F and CALR, in classic BCR-ABL1-negative myeloproliferative neoplasms (MPNs) has generated interest in the development of molecularly targeted therapies, whose accurate assessment requires a standardized framework. A working group, comprised of members from European LeukemiaNet (ELN) and International Working Group for MPN Research and Treatment (IWG-MRT), prepared consensus-based recommendations regarding trial design, patient selection and definition of relevant end points. Accordingly, a response able to capture the long-term effect of the drug should be selected as the end point of phase II trials aimed at developing new drugs for MPNs. A time-to-event, such as overall survival, or progression-free survival or both, as co-primary end points, should measure efficacy in phase III studies. New drugs should be tested for preventing disease progression in myelofibrosis patients with early disease in randomized studies, and a time to event, such as progression-free or event-free survival should be the primary end point. Phase III trials aimed at preventing vascular events in polycythemia vera and essential thrombocythemia should be based on a selection of the target population based on new prognostic factors, including JAK2 mutation. In conclusion, we recommended a format for clinical trials in MPNs that facilitates communication between academic investigators, regulatory agencies and drug companies.


Subject(s)
Consensus , Endpoint Determination , Fusion Proteins, bcr-abl/genetics , Myeloproliferative Disorders/drug therapy , Humans , Myeloproliferative Disorders/genetics , Prognosis
18.
Am J Clin Pathol ; 112(1): 93-100, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10396290

ABSTRACT

Chronic myeloproliferative diseases (MPDs) are not associated with consistent cytogenetic or molecular abnormalities. Demonstration of clonal cell growth by analysis of X-chromosome inactivation (XCI) patterns in females provides a promising tool for diagnosis. However, this technique can be complicated by excessive lyonization of normal cells mimicking clonal cell growth: We analyzed XCI patterns at the human androgen receptor (HUMARA) locus in 146 healthy females, 65 women with secondary neutrophilia, 31 women with reactive thrombocytosis, and 86 women with chronic MPDs. A skewed XCI pattern with greater than 75% amplification of 1 allele (allele ratio > 3:1) was found in 22 (9.1%) of 242 control subjects. The incidence of skewing was statistically significantly lower in women younger than 30 years (2/73) compared with women older than 60 years (10/53). Of 86 patients with a chronic MPD, 71 (82%) exhibited an allele ratio greater than 3:1, whereas only 10 (12%) of 86 age-matched control subjects showed a skewed XCI pattern. Although statistical evaluation of the data showed a significant difference between patients with a chronic MPD and control subjects, proof of clonality in individual, especially elderly, patients is difficult.


Subject(s)
Dosage Compensation, Genetic , Leukocytosis/genetics , Myeloproliferative Disorders/genetics , Neutrophils/pathology , Receptors, Androgen/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Chronic Disease , Clone Cells , Cohort Studies , DNA Primers/chemistry , Female , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukocytosis/pathology , Middle Aged , Myeloproliferative Disorders/blood , Myeloproliferative Disorders/pathology , Polycythemia Vera/blood , Polycythemia Vera/genetics , Polymerase Chain Reaction , Primary Myelofibrosis/blood , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Thrombocytosis/blood , Thrombocytosis/genetics , Thrombocytosis/pathology
19.
Cancer Genet Cytogenet ; 107(1): 43-7, 1998 Nov.
Article in English | MEDLINE | ID: mdl-9809033

ABSTRACT

Pancytopenia and fulminant disseminated intravascular coagulation in a 68-year-old woman suggested an acute hematologic malignancy. However, cytogenetic analysis on a bone marrow sample revealed a near-tetraploid karyotype with an isochromosome 1q and a translocation (2;13) (q35;q14), which was suggestive of an alveolar rhabdomyosarcoma (ARMS). This diagnosis was subsequently confirmed by indirect immunohistochemistry. ARMS has not yet been observed in a patient of this age. Thus, our case underlines the importance of cytogenetics, to establish an a priori unexpected tumor diagnosis.


Subject(s)
Cervical Vertebrae , Chromosomes, Human, Pair 13/genetics , Chromosomes, Human, Pair 2/genetics , Rhabdomyosarcoma, Alveolar/genetics , Spinal Neoplasms/genetics , Translocation, Genetic , Aged , Bone Marrow Examination , Chromosomes, Human, Pair 1/genetics , Fatal Outcome , Female , Fluorescent Antibody Technique, Indirect , Genetic Markers , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Rhabdomyosarcoma, Alveolar/pathology , Spinal Neoplasms/pathology
20.
Leuk Lymphoma ; 15(3-4): 201-8, 1994 Oct.
Article in English | MEDLINE | ID: mdl-7866269

ABSTRACT

Activation of monocytes and granulocytes in vitro by cytokines, in vivo administration of cytokines, as well as in vivo cytokine production due to infectious and inflammatory diseases causes changes of the surface expression density of certain membrane molecules. In recent studies we attempted to determine the feasibility of using flow cytometric immunophenotyping as a tool to develop a sensitive parameter for detecting infections at an early stage of disease when clinical parameters are still negative. Since infections are an important factor determining the clinical course of myelodysplastic syndromes (MDS), early detection of infection might be beneficial for these immunocompromised patients. We indeed found activation-associated immunophenotypic changes of cell surface antigens on monocytes and granulocytes of clinically infection free MDS patients suggesting enhanced immune activity in these patients, most likely due to latent or beginning infections. In particular, analyses of the expression density of receptors for IgG (Fc gamma Rs), complement receptors, and certain activation-associated surface molecules such as the CD67 and the M5 molecule seem to be of clinical relevance. We will also discuss findings concerning changes of cytokine levels and functional alterations of immunologic parameters in MDS patients.


Subject(s)
Myelodysplastic Syndromes/immunology , Humans , Immunophenotyping
SELECTION OF CITATIONS
SEARCH DETAIL