Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Vaccine ; 41(19): 3128-3136, 2023 05 05.
Article in English | MEDLINE | ID: mdl-37061372

ABSTRACT

BACKGROUND: Patients with chronic lymphocytic leukemia (CLL) show an immune dysfunction with increased risk of infections and poor response to vaccination. Streptococcus pneumoniae is a common cause of morbidity and mortality in CLL patients. In a previous randomized clinical trial, we found a superior immune response in CLL patients receiving conjugated pneumococcal vaccine compared to non-conjugated vaccine. The response to revaccination in CLL patients is scarcely studied. In this study, early humoral response to repeated revaccinations with pneumococcal vaccines was evaluated, by determination of B cell subsets and plasmablast dynamics in peripheral blood. METHOD: CLL patients (n = 14) and immunocompetent controls (n = 31) were revaccinated with a 13-valent pneumococcal conjugate vaccine (PCV13) after previous primary immunization (3-6 years ago) with PCV13 or a 23-valent pneumococcal polysaccharide vaccine (PPSV23). Eight weeks after the first revaccination, all CLL patients received a second revaccination with PCV13 or PPSV23. B cell subsets including plasmablasts were analyzed in peripheral blood by flow cytometry, before and after the first and the second revaccination. RESULTS: None of the CLL patients, but all controls, had detectable plasmablasts at baseline (p < 0.001). After the first revaccination with PCV13, the plasmablast proportions did not increase in CLL patients (p = 0.13), while increases were seen in controls (p < 0.001). However, after a second revaccination with PCV13 or PPSV23, plasmablasts increased compared to baseline also in CLL patients (p < 0.01). If no response was evident after first revaccination, only a second revaccination with PCV13 increased plasmablasts in contrast to PPSV23 revaccination. Patients with hypogammaglobulinemia and ongoing/previous CLL specific treatment responded poorly, also to a second revaccination. CONCLUSION: In CLL patients, pneumococcal revaccination induced minor early plasmablast response compared to controls, but the response improved using a strategy of repeated doses with of conjugated T cell dependent pneumococcal vaccine.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell , Pneumococcal Infections , Humans , Antibodies, Bacterial , Double-Blind Method , Immunization, Secondary , Leukemia, Lymphocytic, Chronic, B-Cell/complications , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Pneumococcal Vaccines , Streptococcus pneumoniae , Vaccination , Vaccines, Conjugate , Case-Control Studies
2.
Haematologica ; 107(7): 1528-1537, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35081688

ABSTRACT

With increasingly effective treatments, early death (ED) has become the predominant reason for therapeutic failure in patients with acute promyelocytic leukemia (APL). To better prevent ED, patients with high-risk of ED must be identified. Our aim was to develop a score that predicts the risk of ED in a real-life setting. We used APL patients in the populationbased Swedish AML Registry (n=301) and a Portuguese hospital-based registry (n=129) as training and validation cohorts, respectively. The cohorts were comparable with respect to age (median, 54 and 53 years) and ED rate (19.6% and 18.6%). The score was developed by logistic regression analyses, risk-per-quantile assessment and scoring based on ridge regression coefficients from multivariable penalized logistic regression analysis. White blood cell count, platelet count and age were selected by this approach as the most significant variables for predicting ED. The score identified low-, high- and very high-risk patients with ED risks of 4.8%, 20.2% and 50.9% respectively in the training cohort and with 6.7%, 25.0% and 36.0% as corresponding values for the validation cohort. The score identified an increased risk of ED already at sub-normal and normal white blood cell counts and, consequently, it was better at predicting ED risk than the Sanz score (AUROC 0.77 vs. 0.64). In summary, we here present an externally validated and population-based risk score to predict ED risk in a real-world setting, identifying patients with the most urgent need of aggressive ED prevention. The results also suggest that increased vigilance for ED is already necessary at sub-normal/normal white blood cell counts.


Subject(s)
Leukemia, Promyelocytic, Acute , Cohort Studies , Humans , Leukemia, Promyelocytic, Acute/diagnosis , Leukemia, Promyelocytic, Acute/epidemiology , Leukemia, Promyelocytic, Acute/therapy , Leukocyte Count , Risk Factors , Treatment Outcome
3.
Eur J Radiol Open ; 8: 100357, 2021.
Article in English | MEDLINE | ID: mdl-34095355

ABSTRACT

BACKGROUND: Patients with transfusion dependent anemia are at risk of complications from iron overload. Quantitative T2* magnetic resonance imaging (MRI) is the best non-invasive method to assess iron deposition in the liver and heart and to guide chelation therapy. PURPOSE: To investigate the image quality and inter-observer variations in T2* measurements of the myocardium and the liver, and to obtain the lower limit of cardiac and hepatic quantitative T2* values in patients without suspicion of iron overload. MATERIAL AND METHODS: Thirty-eight patients referred for cardiac MRI were prospectively included in the study. Three patients were referred with, and 35 without suspicion of iron overload. Quantitative T2* parametric maps were obtained on a 1.5 T MRI system in the cardiac short axis and liver axial view. Two readers independently assessed the image quality and the representative and the lowest T2* value in the myocardium and the liver. RESULTS: The normal range of representative T2* values in the myocardium and liver was 24-45 ms and 14-37 ms, respectively. None of the 35 participants (0 %, 95 % confidence interval 0-11 %) in the normal reference group demonstrated representative T2* values below previously reported lower limits in the myocardium (20 ms) or the liver (8 ms). Focal myocardial areas with T2* values near the lower normal range, 19-20 ms, were seen in two patients. The readers generally reported good image quality. CONCLUSION: T2* imaging for assessing iron overload can be performed in a non-dedicated center with sufficient image quality.

4.
Genes Chromosomes Cancer ; 60(6): 426-433, 2021 06.
Article in English | MEDLINE | ID: mdl-33433047

ABSTRACT

Acute myeloid leukemia (AML) with t(9;22)(q34;q11), also known as AML with BCR-ABL1, is a rare, provisional entity in the WHO 2016 classification and is considered a high-risk disease according to the European LeukemiaNet 2017 risk stratification. We here present a retrospective, population-based study of this disease entity from the Swedish Acute Leukemia Registry. By strict clinical inclusion criteria we aimed to identify genetic markers further distinguishing AML with t(9;22) as a separate entity. Twenty-five patients were identified and next-generation sequencing using a 54-gene panel was performed in 21 cases. Interestingly, no mutations were found in NPM1, FLT3, or DNMT3A, three frequently mutated genes in AML. Instead, RUNX1 was the most commonly mutated gene, with aberrations present in 38% of the cases compared to around 10% in de novo AML. Additional mutations were identified in genes involved in RNA splicing (SRSF2, SF3B1) and chromatin regulation (ASXL1, STAG2, BCOR, BCORL1). Less frequently, mutations were found in IDH2, NRAS, TET2, and TP53. The mutational landscape exhibited a similar pattern as recently described in patients with chronic myeloid leukemia (CML) in myeloid blast crisis (BC). Despite the concomitant presence of BCR-ABL1 and RUNX1 mutations in our cohort, both features of high-risk AML, the RUNX1-mutated cases showed a superior overall survival compared to RUNX1 wildtype cases. Our results suggest that the molecular characteristics of AML with t(9;22)/BCR-ABL1 and CML in myeloid BC are similar and do not support a distinction of the two disease entities based on their underlying molecular alterations.


Subject(s)
Fusion Proteins, bcr-abl/genetics , Gene Frequency , Genetic Loci , Leukemia, Myeloid, Acute/genetics , Adult , Aged , Aged, 80 and over , DNA Methyltransferase 3A/genetics , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Mutation , Nucleophosmin/genetics , Phenotype , Sweden , fms-Like Tyrosine Kinase 3/genetics
5.
Eur J Haematol ; 104(6): 588-593, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32115785

ABSTRACT

OBJECTIVE: Hemolysis is a sporadically reported but potentially serious side effect of the proteasome inhibitor carfilzomib. We aimed to investigate the frequency of hemolysis in an unselected cohort. METHODS: We performed a retrospective, single-center study of the incidence of hemolysis in patients treated with carfilzomib, based mainly on consecutive haptoglobin levels. The patients were diagnosed with myeloma (n = 20), AL amyloidosis (n = 3), and light-chain deposition disease (n = 1). Carfilzomib treatment was applied after a median of 3 (range: 1-7) therapy lines. RESULTS: Haptoglobin levels were normal/increased before, generally suppressed during, and normalized after treatment with carfilzomib. Very low haptoglobin (<0.1 g/L) implying the presence of hemolysis was observed in 16 of 24 (67%) patients during carfilzomib therapy. Hemolysis was mild in 11 of 16 (69%) affected patients, whereas 5 of 16 (31%) required transfusion. Severe hemolysis was explained by thrombotic microangiopathy (TMA) in one patient who died of the complication. Mechanisms were unclear in the remaining 15 patients. CONCLUSIONS: Hemolysis was surprisingly common but mostly mild during carfilzomib treatment. However, the possibility of TMA should be kept in mind in this setting. Hypothetically, non-TMA hemolysis could be attributed to the accumulation of globin chains due to the suppression of eukaryotic translation initiation inhibition by carfilzomib.


Subject(s)
Oligopeptides/adverse effects , Proteasome Inhibitors/adverse effects , Thrombotic Microangiopathies/diagnosis , Thrombotic Microangiopathies/etiology , Aged , Biomarkers , Female , Hemolysis , Humans , Male , Middle Aged , Multiple Myeloma/complications , Multiple Myeloma/drug therapy , Oligopeptides/therapeutic use , Proteasome Inhibitors/therapeutic use , Retrospective Studies , Thrombotic Microangiopathies/blood
8.
Biol Blood Marrow Transplant ; 25(10): 1970-1974, 2019 10.
Article in English | MEDLINE | ID: mdl-31173901

ABSTRACT

Allogeneic stem cell transplantation (SCT) as primary treatment for aplastic anemia (AA) is being increasingly used. Yet, age, stem cell source, and donor type are important outcome factors. We have recently performed a nationwide cohort study of all patients with AA in Sweden diagnosed from 2000 to 2011 and now present outcome data on SCT patients. In total, 68 patients underwent SCT, and 63% of them had failed immunosuppressive therapy. We found that, with a median follow-up of 109 months (range, 35 to 192 months), 5-year overall survival (OS) for all patients was 86.8%, whereas graft-versus-host disease-free, relapse/rejection-free survival (GRFS) at 5 years was 69.1%. There was no survival impact regarding the donor type or stem cell source. Patients aged ≥40 years had a higher transplant-related mortality (29.4% versus 7.8%; P = .023), which translated into a lower 5-year OS: 70.6% versus 92.2% (P = .022) and a trend of lower GRFS (52.9% versus 74.5%; P = .069). In conclusion, we found in this real-world setting that both OS and GRFS were high, but SCT for patients with AA aged ≥40 years is problematic, and clinical trials addressing this issue are warranted.


Subject(s)
Anemia, Aplastic/therapy , Adolescent , Adult , Aged , Anemia, Aplastic/pathology , Child , Child, Preschool , Cohort Studies , Female , Graft vs Host Disease/mortality , Humans , Male , Middle Aged , Recurrence , Sweden , Tissue Donors , Unrelated Donors , Young Adult
9.
Eur J Haematol ; 100(6): 613-620, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29532518

ABSTRACT

OBJECTIVES: Antithymocyte globulin (ATG)-based immunosuppression remains a cornerstone in aplastic anaemia (AA) treatment. However, most ATG studies are not population-based and knowledge about real-world results concerning response and outcome could offer important information for treating physicians. METHODS: We have recently performed a nationwide retrospective cohort study on all AA patients diagnosed in Sweden in 2000-2011 and now present treatment and outcome data on patients receiving first-line ATG. In total, 158 patients showed a 47.0% response rate which was similar in all age groups (range 41.5%-51.7%) with no difference regarding ATG formulation. The response was significantly associated with severity grade-especially at time of treatment initiation: very severe (VSAA) 22.7%; severe (SAA) 54.5% (P < .001); and non-severe 88.5% (P < .001). A logistic regression-based predictive model indicated that VSAA patients with an absolute reticulocyte count <25 × 109 /L had only a 19% probability of response. In a multivariable analysis, age and VSAA at the time of treatment were the independent factors for inferior survival. CONCLUSIONS: Real-world VSAA patients respond poorly to ATG which indicates the need for a different treatment approach. Our findings suggest that age alone should not be a discriminating factor for administering ATG treatment.


Subject(s)
Anemia, Aplastic/drug therapy , Antilymphocyte Serum/therapeutic use , Immunosuppressive Agents/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Anemia, Aplastic/diagnosis , Anemia, Aplastic/epidemiology , Anemia, Aplastic/immunology , Biomarkers , Child , Child, Preschool , Cohort Studies , Combined Modality Therapy , Female , Hematopoietic Stem Cell Transplantation , Humans , Incidence , Male , Middle Aged , Odds Ratio , Severity of Illness Index , Survival Analysis , Sweden/epidemiology , Young Adult
10.
Haematologica ; 102(10): 1683-1690, 2017 10.
Article in English | MEDLINE | ID: mdl-28751565

ABSTRACT

A plastic anemia is a rare life-threatening disease. However, since the introduction of immunosuppressive therapy and allogeneic stem cell transplantation, the outcome has improved considerably, and the 5-year survival is reported to be 70-80% in selected patient cohorts. Yet, contemporary population-based data on incidence and survival are lacking. We performed a national retrospective study to determine the incidence, treatment, and survival of patients with aplastic anemia diagnosed in Sweden from 2000-2011. Patients were included via the National Patient Registry, and diagnosed according to the Camitta criteria. In total, 257 confirmed cases were identified, with an overall incidence of 2.35 (95% CI: 2.06-2.64) cases per million inhabitants per year. Median age was 60 years (range: 2-92), and median follow up was 76 (0-193) months. Primary treatments included immunosuppressive therapy (63%), allogenic stem cell transplantation (10%), or single-agent cyclosporine/no specific therapy (27%). The 5-year survival was 90.7% in patients aged 0-18 years, 90.5% in patients aged 19-39 years, 70.7% in patients aged 40-59 years, and 38.1% in patients aged ≥60 years. Multivariate analysis showed that age (both 40-59 and ≥60 age groups), very severe aplastic anemia and single-agent cyclosporine/no specific therapy were independent risk factors for inferior survival. In conclusion, younger aplastic anemia patients experience a very good long-term survival, while that of patients ≥60 years in particular remains poor. Apparently, the challenge today is to improve the management of older aplastic anemia patients, and prospective studies to address this medical need are warranted.


Subject(s)
Anemia, Aplastic/epidemiology , Adolescent , Adult , Aged , Aged, 80 and over , Anemia, Aplastic/diagnosis , Anemia, Aplastic/history , Anemia, Aplastic/therapy , Child , Child, Preschool , Combined Modality Therapy , Female , History, 21st Century , Humans , Incidence , Male , Middle Aged , Patient Outcome Assessment , Population Surveillance , Proportional Hazards Models , Registries , Retrospective Studies , Severity of Illness Index , Survival Analysis , Sweden/epidemiology , Young Adult
11.
Eur J Haematol ; 98(5): 493-500, 2017 May.
Article in English | MEDLINE | ID: mdl-28152233

ABSTRACT

OBJECTIVES AND METHODS: To ascertain the incidence/clinical implications of isolated autosomal trisomies in adult acute myeloid leukemia (AML), all such cases were retrieved from the Swedish AML Registry. RESULTS: Of the 3179 cytogenetically informative AMLs diagnosed January 1997-May 2015, 246 (7.7%) had isolated trisomies. The frequency increased by age (2.4% at age 18-60 years vs. 23% at >60 years; P<.0001); the median age was 69 years. The five most common were +8 (4.0%), +13 (0.9%), +11 (0.8%), +21 (0.7%), and +4 (0.5%). Age and gender, types of AML and treatment, and complete remission and early death rates did not differ between the single trisomy and the intermediate risk (IR) groups or among cases with isolated gains of chromosomes 4, 8, 11, 13, or 21. The overall survival (OS) was similar in the single trisomy (median 1.6 years) and IR groups (1.7 years; P=.251). The OS differed among the most frequent isolated trisomies; the median OS was 2.5 years for +4, 1.9 years for +21, 1.5 years for +8, 1.1 years for +11, and 0.8 years for +13 (P=.013). CONCLUSION: AML with single trisomies, with the exception of +13, should be grouped as IR.


Subject(s)
Leukemia, Myeloid, Acute/epidemiology , Leukemia, Myeloid, Acute/genetics , Trisomy , Adolescent , Adult , Aged , Aged, 80 and over , Combined Modality Therapy , Female , Humans , In Situ Hybridization, Fluorescence , Incidence , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/therapy , Male , Middle Aged , Population Surveillance , Prognosis , Registries , Risk , Survival Analysis , Sweden/epidemiology , Young Adult
12.
Am J Hematol ; 90(9): 800-5, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26088289

ABSTRACT

To ascertain the clinical implications of high hyperdiploid (HH; 49-65 chromosomes) and triploid/tetraploid (TT; >65 chromosomes) adult acute myeloid leukemia (AML), all such cases were retrieved from the Swedish AML Registry. Of the 3,654 cytogenetically informative cases diagnosed between January 1997 and May 2014, 68 (1.9%) were HH (n = 50)/TT (n = 18). Patients with HH/TT were older than those with intermediate risk (IR) AML (median 71 years vs. 67 years; P = 0.042) and less often had de novo AML (63% vs. 79%; P = 0.004); no such differences were observed between HH/TT and complex karyotype (CK) AML. The overall survival (OS) was similar between patients with HH/TT and CK AML (median 0.9 years vs. 0.6 years; P = 0.082), whereas OS was significantly longer (median 1.6 years; P = 0.028) for IR AML. The OS was shorter for cases with HH than with TT (median 0.6 years vs. 1.4 years; P = 0.032) and for HH/TT AMLs with adverse abnormalities (median 0.8 years vs. 1.1 years; P = 0.044). In conclusion, HH/TT AML is associated with a poor outcome, but chromosome numbers >65 and absence of adverse aberrations seem to translate into a more favorable prognosis. Thus, HH/TT AMLs are clinically heterogeneous and should not automatically be grouped as high risk.


Subject(s)
Chromosome Aberrations , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/genetics , Ploidies , Registries , Adolescent , Adult , Aged , Antineoplastic Agents/therapeutic use , Female , Humans , Karyotyping , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Prognosis , Risk , Survival Analysis , Sweden
13.
Am J Hematol ; 90(3): 208-14, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25421221

ABSTRACT

Patients with secondary acute myeloid leukemia (AML) often escape inclusion in clinical trials and thus, population-based studies are crucial for its accurate characterization. In this first large population-based study on secondary AML, we studied AML with an antecedent hematological disease (AHD-AML) or therapy-related AML (t-AML) in the population-based Swedish Acute Leukemia Registry. The study included 3,363 adult patients of which 2,474 (73.6%) had de novo AML, 630 (18.7%) AHD-AML, and 259 (7.7%) t-AML. Secondary AML differed significantly compared to de novo AML with respect to age, gender, and cytogenetic risk. Complete remission (CR) rates were significantly lower but early death rates similar in secondary AML. In a multivariable analysis, AHD-AML (HR 1.51; 95% CI 1.26-1.79) and t-AML (1.72; 1.38-2.15) were independent risk factors for poor survival. The negative impact of AHD-AML and t-AML on survival was highly age dependent with a considerable impact in younger patients, but without independent prognostic value in the elderly. Although patients with secondary leukemia did poorly with intensive treatment, early death rates and survival were significantly worse with palliative treatment. We conclude that secondary AML in a population-based setting has a striking impact on survival in younger AML patients, whereas it lacks prognostic value among the elderly patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Breast Neoplasms/drug therapy , Leukemia, Myeloid, Acute/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Neoplasms, Second Primary/drug therapy , Palliative Care , Registries , Adult , Age Factors , Aged , Aged, 80 and over , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Female , Humans , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Lymphoma, Non-Hodgkin/mortality , Lymphoma, Non-Hodgkin/pathology , Male , Middle Aged , Neoplasms, Second Primary/mortality , Neoplasms, Second Primary/pathology , Prognosis , Remission Induction , Risk Factors , Survival Analysis , Sweden
14.
Clin Biochem ; 48(3): 122-4, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25526882

ABSTRACT

OBJECTIVES: Plasma or serum hyperviscosity in plasma cell dyscrasias (PCD) has been described as a risk factor for circulatory disturbances. Whole blood viscosity (WBV) would theoretically be a better biomarker but has not been studied in PCD. DESIGN AND METHODS: Plasma viscosity (PV) and WBV were measured in 89 subjects with PCD and in 60 healthy blood donors by free oscillation rheometry. A complete blood count was obtained using an automated hematology analyzer. Plasma proteins were quantitated by immunoturbidimetry. RESULTS: The reference intervals for men & women were 1.16-1.36 & 1.16-1.38 mPa for PV, and 4.9-6.3 & 4.4-6.2 mPa for WBV, respectively. Of the PCD patients, 71% had PV above the reference limit and 40% were above the WBV limit. Multivariate analysis showed that WBV was independently related to hematocrit, PV, concentration of the monoclonal protein (M-protein), plasma fibrinogen concentration and albumin concentration. This model accounted for 76% of the variance in WBV. When the same model was applied to PV, only the concentration of the M-protein was significantly related and the model accounted only for 20% of the variance in PV. CONCLUSION: PV cannot be used as a surrogate marker for WBV in PCD patients. Whole blood viscosity should replace plasma viscosity in patients with PCD.


Subject(s)
Blood Viscosity , Paraproteinemias/blood , Aged , Blood Donors , Cohort Studies , Erythrocyte Volume , Female , Fibrinogen/metabolism , Hematocrit , Humans , Immunoglobulin A/blood , Immunoglobulin G/blood , Immunoglobulin M/blood , Male , Multivariate Analysis , Plasma/metabolism , Reference Values
15.
Eur J Haematol ; 94(5): 419-23, 2015 May.
Article in English | MEDLINE | ID: mdl-25200361

ABSTRACT

Unsuccessful cytogenetics (UC) in patients with acute myeloid leukaemia (AML) treated on different SWOG trials was recently reported to be associated with increased age and dismal outcome. To ascertain whether this holds true also in unselected patients with AML, we retrieved all cytogenetic reports in cases from the population-based Swedish AML Registry. Between 1997 and 2006, 1737 patients below 80 yr of age without myelosarcoma or acute promyelocytic leukaemia received intensive treatment. The frequencies of UC and unperformed cytogenetics (UPC) were 2.1% and 20%, respectively. The early death rates differed between the cytogenetic subgroups (P = 0.006) with the highest rates in patients with UC (14%) and UPC (12%) followed by high-risk (HR) AML, intermediate risk (IR) and standard risk (SR) cases successfully karyotyped (8.6%, 5.9%, and 5.8%, respectively). The complete remission rate was lower in UC and UPC and HR compared with the other risk groups (P < 0.001). The overall five-year survival rates were 25% for UC and 22% for UPC, whereas the corresponding frequencies for SR, IR and HR AML patients without UC and UPC were 64%, 31% and 15%, respectively. In conclusion, lack of cytogenetic data translates into a poor prognosis.


Subject(s)
Chromosome Aberrations , Karyotyping/statistics & numerical data , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Registries , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/therapeutic use , Female , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Prognosis , Remission Induction , Retrospective Studies , Risk , Survival Analysis , Sweden
16.
Leuk Lymphoma ; 55(12): 2769-77, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24524339

ABSTRACT

We conducted a randomized phase III trial to compare the efficacy and safety of two purine analogs, cladribine and fludarabine, with high-dose chlorambucil, in patients with previously untreated chronic lymphocytic leukemia (CLL). Between 1997 and 2004, 223 patients with CLL were randomly assigned to cladribine, fludarabine or chlorambucil, for six cycles of therapy with frequent health-related quality of life assessments. There was no statistical difference for the primary endpoint of overall response with cladribine (70%), fludarabine (67%) and chlorambucil (59%), or complete remission (12%, 7% and 8%), respectively. However, the median progression-free survival (25, 10, 9 months) and median time to second treatment (40, 22, 21 months) were superior with cladribine. There was no significant difference in overall survival (96, 82 and 91 months), nor in toxicity or HRQoL assessments. Monotherapy with cladribine gives superior PFS and longer response duration than fludarabine and chlorambucil as first-line treatment of CLL.


Subject(s)
Antineoplastic Agents/therapeutic use , Chlorambucil/therapeutic use , Cladribine/therapeutic use , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Vidarabine/analogs & derivatives , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Chlorambucil/administration & dosage , Chlorambucil/adverse effects , Cladribine/administration & dosage , Cladribine/adverse effects , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Male , Middle Aged , Neoplasm Staging , Quality of Life , Retreatment , Treatment Outcome , Vidarabine/administration & dosage , Vidarabine/adverse effects , Vidarabine/therapeutic use
17.
J Neurol Neurosurg Psychiatry ; 85(10): 1116-21, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24554104

ABSTRACT

BACKGROUND: Autologous haematopoietic stem cell transplantation (HSCT) is a viable option for treatment of aggressive multiple sclerosis (MS). No randomised controlled trial has been performed, and thus, experiences from systematic and sustained follow-up of treated patients constitute important information about safety and efficacy. In this observational study, we describe the characteristics and outcome of the Swedish patients treated with HSCT for MS. METHODS: Neurologists from the major hospitals in Sweden filled out a follow-up form with prospectively collected data. Fifty-two patients were identified in total; 48 were included in the study and evaluated for safety and side effects; 41 patients had at least 1 year of follow-up and were further analysed for clinical and radiological outcome. In this cohort, 34 patients (83%) had relapsing-remitting MS, and mean follow-up time was 47 months. RESULTS: At 5 years, relapse-free survival was 87%; MRI event-free survival 85%; expanded disability status scale (EDSS) score progression-free survival 77%; and disease-free survival (no relapses, no new MRI lesions and no EDSS progression) 68%. Presence of gadolinium-enhancing lesions prior to HSCT was associated with a favourable outcome (disease-free survival 79% vs 46%, p=0.028). There was no mortality. The most common long-term side effects were herpes zoster reactivation (15%) and thyroid disease (8.4%). CONCLUSIONS: HSCT is a very effective treatment of inflammatory active MS and can be performed with a high degree of safety at experienced centres.


Subject(s)
Brain/surgery , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/mortality , Multiple Sclerosis/surgery , Transplantation, Autologous/adverse effects , Transplantation, Autologous/mortality , Adolescent , Adult , Brain/pathology , Child , Disability Evaluation , Disease-Free Survival , Female , Follow-Up Studies , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Multiple Sclerosis/mortality , Multiple Sclerosis/pathology , Neuroimaging , Recurrence , Sweden , Treatment Outcome , Young Adult
18.
J Clin Oncol ; 30(29): 3633-9, 2012 Oct 10.
Article in English | MEDLINE | ID: mdl-22965953

ABSTRACT

PURPOSE: APR-246 (PRIMA-1MET) is a novel drug that restores transcriptional activity of unfolded wild-type or mutant p53. The main aims of this first-in-human trial were to determine maximum-tolerated dose (MTD), safety, dose-limiting toxicities (DLTs), and pharmacokinetics (PK) of APR-246. PATIENTS AND METHODS: APR-246 was administered as a 2-hour intravenous infusion once per day for 4 consecutive days in 22 patients with hematologic malignancies and prostate cancer. Acute myeloid leukemia (AML; n = 7) and prostate cancer (n = 7) were the most frequent diagnoses. Starting dose was 2 mg/kg with dose escalations up to 90 mg/kg. RESULTS: MTD was defined as 60 mg/kg. The drug was well tolerated, and the most common adverse effects were fatigue, dizziness, headache, and confusion. DLTs were increased ALT/AST (n = 1), dizziness, confusion, and sensory disturbances (n = 2). PK showed little interindividual variation and were neither dose nor time dependent; terminal half-life was 4 to 5 hours. Tumor cells showed cell cycle arrest, increased apoptosis, and upregulation of p53 target genes in several patients. Global gene expression analysis revealed changes in genes regulating proliferation and cell death. One patient with AML who had a p53 core domain mutation showed a reduction of blast percentage from 46% to 26% in the bone marrow, and one patient with non-Hodgkin's lymphoma with a p53 splice site mutation showed a minor response. CONCLUSION: We conclude that APR-246 is safe at predicted therapeutic plasma levels, shows a favorable pharmacokinetic profile, and can induce p53-dependent biologic effects in tumor cells in vivo.


Subject(s)
Genes, p53/drug effects , Hematologic Neoplasms/drug therapy , Prostatic Neoplasms/drug therapy , Quinuclidines/therapeutic use , Aged , Aged, 80 and over , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Infusions, Intravenous , Male , Maximum Tolerated Dose , Middle Aged , Molecular Targeted Therapy/methods , Patient Safety , Pharmacogenetics , Prospective Studies , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Quinuclidines/adverse effects , Quinuclidines/pharmacology , Risk Factors , Sensitivity and Specificity , Treatment Outcome , Tumor Cells, Cultured/drug effects
20.
Blood ; 118(20): 5573-82, 2011 Nov 17.
Article in English | MEDLINE | ID: mdl-21960591

ABSTRACT

Cytogenetically normal acute myeloid leukemia (CN-AML) compose between 40% and 50% of all adult acute myeloid leukemia (AML) cases. In this clinically diverse group, molecular aberrations, such as FLT3-ITD, NPM1, and CEBPA mutations, recently have added to the prognostic accuracy. Aberrant DNA methylation is a hallmark of cancer, including AML. We investigated in total 118 CN-AML samples in a test and a validation cohort for genome-wide promoter DNA methylation with Illumina Methylation Bead arrays and compared them with normal myeloid precursors and global gene expression. IDH and NPM1 mutations were associated with different methylation patterns (P = .0004 and .04, respectively). Genome-wide methylation levels were elevated in IDH-mutated samples (P = .006). We observed a negative impact of DNA methylation on transcription. Genes targeted by Polycomb group (PcG) proteins and genes associated with bivalent histone marks in stem cells showed increased aberrant methylation in AML (P < .0001). Furthermore, high methylation levels of PcG target genes were independently associated with better progression-free survival (odds ratio = 0.47, P = .01) and overall survival (odds ratio = 0.36, P = .001). In summary, genome-wide methylation patterns show preferential methylation of PcG targets with prognostic impact in CN-AML.


Subject(s)
Chromatin/genetics , DNA Methylation/physiology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Stem Cells/physiology , Adolescent , Adult , Aged , Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Female , Genome-Wide Association Study/methods , Genome-Wide Association Study/standards , Histones/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Nucleophosmin , Polycomb-Group Proteins , Prognosis , Promoter Regions, Genetic/physiology , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reproducibility of Results , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...