Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Am J Alzheimers Dis Other Demen ; 38: 15333175231172283, 2023.
Article in English | MEDLINE | ID: mdl-37177903

ABSTRACT

BACKGROUND: The role of diastolic blood pressure (DBP) with cognitive functioning is under-explored in relation to levels of systolic blood pressure (SBP). METHODS: We studied 5466 participants from the National Health and Nutrition Examination Survey. Blood pressure was measured 3 times manually with a standardized sphygmomanometer and averaged. Cognitive functioning was measured using the digit symbol substitution test (DSST). RESULTS: Participants were 60 years or older, 55% female, and 81% non-Hispanic White. Most participants had a DBP between 70 to <80 mmHg (33.7%), between 60 to <70 mmHg (29.3%), or <60 mmHg (18.8%). From multivariable linear regression analyses, each 5 mmHg increment of DBP was associated with significantly higher DSST scores among individuals with SBP <120 only (ß: 0.56, 95% CI: 0.09, 1.03). CONCLUSIONS: Among older US adults, at non-elevated levels of SBP, higher DBP is associated with better cognitive performance.


Subject(s)
Hypertension , Humans , Adult , Female , Middle Aged , Aged , Male , Blood Pressure/physiology , Nutrition Surveys , Cognition/physiology
2.
Leukemia ; 21(9): 1964-70, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17581608

ABSTRACT

Patients with primary myelofibrosis (PMF) or post-polycythemia vera or post-essential thrombocythemia myelofibrosis (post-PV/ET MF) have limited therapeutic options. The farnesyltransferase-inhibitor tipifarnib inhibits in vitro proliferation of myeloid progenitors from such patients. In the current phase II clinical trial, single-agent oral tipifarnib (300 mg twice daily x 21 of 28 days) was given to 34 symptomatic patients with either PMF (n=28) or post-PV/ET MF (n=6). Median time to discontinuation of protocol therapy was 4.6 months; reasons for early termination (n=19; 56%) included disease progression (21%) and adverse drug effects (18%). Toxicities (>/=grade 3) included myelosuppression (n=16), neuropathy (n=2), fatigue (n=1), rash (n=1) and hyponatremia (n=1). Response rate was 33% for hepatosplenomegaly and 38% for transfusion-requiring anemia. No favorable changes occurred in bone marrow fibrosis, angiogenesis or cytogenetic status. Pre- and post-treatment patient sample analysis for in vitro myeloid colony growth revealed substantial reduction in the latter. Clinical response did not correlate with either degree of colony growth, measurable decrease in quantitative JAK2(V617F) levels or tipifarnib IC(50) values (median 11.8 nM) seen in pretreatment samples. The current study indicates both in vitro and in vivo tipifarnib activity in PMF and post-PV/ET MF.


Subject(s)
Antineoplastic Agents/administration & dosage , Polycythemia Vera/complications , Primary Myelofibrosis/drug therapy , Quinolones/administration & dosage , Thrombocythemia, Essential/complications , Aged , Aged, 80 and over , Anemia/drug therapy , Anemia/etiology , Antineoplastic Agents/adverse effects , Bone Marrow/pathology , Female , Humans , Janus Kinase 2/genetics , Male , Middle Aged , Polycythemia Vera/genetics , Primary Myelofibrosis/etiology , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Prospective Studies , Quinolones/adverse effects , Thrombocythemia, Essential/genetics , Treatment Outcome
3.
Leuk Res ; 31(11): 1503-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17397921

ABSTRACT

We recently developed a modified Dupriez prognostic scoring system (PSS) that effectively discriminated between high-, intermediate-, and low-risk young patients (age < or =60 years) with primary myelofibrosis (PMF) based on the respective presence of none, one, or two or more of the following parameters: hemoglobin <10 g/dL, leukocyte count <4 or >30 x 10(9)L(-1), and platelet count <100 x 10(9)L(-1). The current study (n=129; median age, 52 years; 69 males) reveals, on multivariable analysis, that an absolute monocyte count of > or =1 x 10(9)L(-1) carries an independent predictive value (p=0.02), for an inferior survival, in addition to that provided by hemoglobin level (p=0.002), platelet count (0.02), and leukocyte count (p=0.16). The inclusion of the monocyte count as a fourth risk factor enabled the construction of a new and improved Mayo PSS; median survival was 173, 61, and 26 months in the absence of all four (low-risk), three (intermediate-risk), or two or less (high-risk) adverse features, respectively (p<0.0001). The independent prognostic value of monocytosis was validated in a separate database of 97 patients with PMF from another institution.


Subject(s)
Monocytes/pathology , Primary Myelofibrosis/pathology , Adolescent , Adult , Female , Humans , Male , Middle Aged , Prognosis
4.
Leukemia ; 20(10): 1800-8, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16871275

ABSTRACT

An activating point mutation in Janus kinase 2 (JAK2 V617F) was recently identified in myelofibrosis with myeloid metaplasia (MMM). To further elucidate the pathogenic significance, we examined the JAK2 mutation burden, phosphorylation of JAK2 substrates and neutrophil apoptotic resistance. Immunoblotting revealed phosphorylation of signal transducer and activator of transcription-3 (STAT3) in all four JAK2 with high V617F mutant allele burden and seven of eight with intermediate mutant allele burden, but only one of eight with wild-type JAK2 (P<0.001). In contrast, STAT5 phosphorylation was undetectable in patient MMM neutrophils; and phosphorylation of Akt and extracellular signal-regulated kinases (ERKs) failed to correlate with JAK2 mutation status. Apoptosis was lower in MMM neutrophils (median 41% apoptotic cells, n=50) compared to controls (median 66%, n=9) or other myeloproliferative disorder patients (median 53%, n=11; P=0.002). Apoptotic resistance in MMM correlated with anemia (P=0.01) and the JAK2-V617F (P=0.01). Indeed, apoptotic resistance was greatest in MMM neutrophils with high mutant allele burden (median 22% apoptosis, n=5) than with intermediate burden (median 39%, n=23) or wild-type JAK2 (median 47%, n=22; P=0.008). These results suggest that mutant JAK2 contributes to MMM pathogenesis by constitutively phosphorylating STAT3 and diminishing myeloid cell apoptosis.


Subject(s)
Apoptosis/physiology , Primary Myelofibrosis/metabolism , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , STAT3 Transcription Factor/metabolism , Alleles , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Janus Kinase 2 , Neutrophils/enzymology , Neutrophils/pathology , Phosphorylation , Point Mutation , Primary Myelofibrosis/genetics , Primary Myelofibrosis/pathology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism
5.
Leukemia ; 20(1): 29-34, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16281063

ABSTRACT

Absolute lymphocyte count (ALC) recovery postautologous stem cell transplantation is an independent predictor for survival in acute myelogenous leukemia (AML). The role of ALC recovery after induction chemotherapy (IC) in AML is unknown. We hypothesize that ALC recovery after IC has a direct impact on survival. We have now evaluated the impact of ALC recovery after IC on overall survival (OS) and leukemia-free survival (LFS) in 103 consecutive, newly diagnosed AML patients treated with standard IC and consolidation chemotherapy (CC) from 1998 to 2002. ALC recovery was studied at days 15 (ALC-15), 21 (ALC-21), 28 (ALC-28) after IC and before the first CC (ALC-CC). Superior OS and LFS at each time point were observed with an ALC-15, ALC-21, ALC-28, and ALC-CC > or = 500 cells/microl. Patients with an ALC > or = 500 cells/microl at all time points vs those who did not have superior OS and LFS (not reached vs 13 months, P<0.0001; and not reached vs 11 months, P<0.0001, respectively). Multivariate analysis demonstrated ALC > or = 500 cells/microl at all time points to be an independent prognostic factor for survival. Our data suggest a critical role of lymphocyte (immune) recovery on survival after IC in AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Lymphocyte Count , Adolescent , Adult , Aged , Aged, 80 and over , Disease-Free Survival , Female , Humans , Leukemia, Myeloid, Acute/diagnosis , Male , Middle Aged , Multivariate Analysis , Predictive Value of Tests , Prognosis , Survival Rate , Treatment Outcome
6.
Leukemia ; 31(2): 393-402, 2017 02.
Article in English | MEDLINE | ID: mdl-27479177

ABSTRACT

NS-018 is a Janus-activated kinase 2 (JAK2)-selective inhibitor, targeting the JAK-signal transducer and activator of transcription (STAT) pathway that is deregulated in myelofibrosis. In this phase I, dose-escalation portion of a phase I/II study, patients with myelofibrosis received oral NS-018 in continuous 28-day cycles. The primary study objective was to evaluate safety, tolerability and clinically active dose of NS-018. Forty-eight patients were treated; 23 (48%) had previously received a JAK inhibitor (JAKi). The most common drug-related adverse events were thrombocytopenia (27%)/anemia (15%) for hematologic events, and dizziness (23%)/nausea (19%) for non-hematologic events. Once daily NS-018 at 300 mg was chosen as the phase II study dose based on improved tolerability compared with higher doses. A ⩾50% reduction in palpable spleen size was achieved in 56% of patients (47% of patients with prior JAKi treatment), and improvements were observed in myelofibrosis-associated symptoms. Bone marrow fibrosis grade (local assessment) improved from baseline in 11/30 evaluable patients (37%) after 3 cycles of NS-018. JAK2 allele burden was largely unchanged. Changes in cytokine/protein levels were noted after 4 weeks of treatment. NS-018 reached peak plasma concentration in 1-2 h and did not accumulate with multiple dosing. NS-018 will be assessed in patients with previous JAKi exposure in the phase II portion.


Subject(s)
Antineoplastic Agents/therapeutic use , Janus Kinase 2/antagonists & inhibitors , Molecular Targeted Therapy , Primary Myelofibrosis/drug therapy , Protein Kinase Inhibitors/therapeutic use , Adult , Aged , Aged, 80 and over , Alleles , Antineoplastic Agents/pharmacology , Biomarkers , Bone Marrow/pathology , Female , Follow-Up Studies , Humans , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Male , Middle Aged , Neoplasm Grading , Phenotype , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/metabolism , Primary Myelofibrosis/therapy , Protein Kinase Inhibitors/pharmacology , Retreatment , Treatment Outcome
7.
Leukemia ; 17(5): 849-55, 2003 May.
Article in English | MEDLINE | ID: mdl-12750696

ABSTRACT

R115777 is an orally bioavailable farnesyltransferase inhibitor (FTI) that has displayed encouraging activity in patients with acute myeloid leukemia. To determine whether R115777 might exert similar activity in myelofibrosis with myeloid metaplasia (MMM), we evaluated its effects on circulating myeloid progenitor cells from patients with MMM (n=25) using in vitro colony-forming assays. The median R115777 concentrations that inhibited colony formation by 50% were 34 and 2.7 nM for myeloid and megakaryocytic colonies from MMM patients, respectively. Progenitors from normal controls and patients with other myeloproliferative disorders demonstrated similar sensitivity. Since the ras polypeptides are one putative target of FTIs, the potential role of ras effectors was examined by incubating parallel progenitor assays with the phosphatidyl-inositol-3 (PI-3) kinase inhibitor LY294002 and the mitogen-activated protein kinase 1 inhibitor PD98059. MMM progenitor colonies (n=7) were highly sensitive to LY294002 but not to PD98059, implying that the PI-3 kinase pathway may be critical for survival and proliferation of these cells. In addition to indicating that MMM progenitors are sensitive to clinically achievable R115777 concentrations in vitro, these results provide a potential explanation for the thrombocytopenia observed with R115777 during the treatment of other hematologic malignancies.


Subject(s)
Alkyl and Aryl Transferases/antagonists & inhibitors , Antineoplastic Agents/pharmacology , Hematopoietic Stem Cells/drug effects , Primary Myelofibrosis/blood , Quinolones/pharmacology , Adult , Aged , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Cell Division/drug effects , Cell Survival/drug effects , Cells, Cultured , Chromones/pharmacology , Colony-Forming Units Assay , Enzyme Inhibitors/pharmacology , Erythrocyte Count , Farnesyltranstransferase , Female , Flavonoids/pharmacology , GPI-Linked Proteins , Humans , Male , Membrane Glycoproteins , Mesothelin , Middle Aged , Morpholines/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Polycythemia Vera/blood , Polycythemia Vera/drug therapy , Polycythemia Vera/pathology , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/pathology , Proteins/metabolism , Thrombocythemia, Essential/blood , Thrombocythemia, Essential/drug therapy , Thrombocythemia, Essential/pathology
8.
Blood Rev ; 14(3): 121-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10986148

ABSTRACT

Myelofibrosis with myeloid metaplasia (MMM) is a collective term that describes the related disorders AMM, PPMM, and PTMM. The chronic myeloid disorders include chronic myeloid leukemia, polycythemia vera, essential thrombocythemia, and agnogenic myeloid metaplasia (myelofibrosis). These disorders display varying propensities for pathologic enlargement of the spleen which can lead to mechanical discomfort, hypercatabolic symptoms, anemia, thrombocytopenia, and portal hypertension. Splenectomy has been found to be of little benefit in the early stages of chronic myeloid leukemia. Similarly, the benefit of splenectomy in advanced cases is limited to symptomatic palliation and treatment of delayed engraftment after allogeneic bone marrow transplantation. Although polycythemia vera and essential thrombocythemia are also characterized by splenomegaly, splenectomy is not considered a therapeutic option in the absence of transformation of the disease into myelofibrosis with myeloid metaplasia. Splenectomy has been studied most in myelofibrosis with myeloid metaplasia. Although there is no clear survival advantage to splenectomy in this disorder, the surgical procedure can result in substantial palliation of mechanical discomfort, hypercatabolic symptoms, portal hypertension, and anemia. However, the procedure is associated with an approximately 9% mortality rate, and the postsplenectomy occurrence of extreme thrombocytosis, hepatomegaly, and leukemic transformation is of major concern.


Subject(s)
Leukemia, Myelogenous, Chronic, BCR-ABL Positive/surgery , Primary Myelofibrosis/surgery , Splenectomy , Humans , Primary Myelofibrosis/complications
9.
Mayo Clin Proc ; 73(1): 37-45, 1998 Jan.
Article in English | MEDLINE | ID: mdl-9443676

ABSTRACT

In this article, we describe pulmonary hypertension in two men (31 and 43 years of age) with human immunodeficiency virus (HIV) infection who were examined at Mayo Clinic Rochester. Among 88 reported cases (including the two current ones) of HIV- or acquired immunodeficiency syndrome (AIDS)-associated pulmonary hypertension, 61% were male; the age range was 2 to 56 years (mean, 32). Dyspnea was the usual initial symptom. Of the 74 patients in whom pulmonary artery pressure was recorded or calculated by echocardiography, systolic pressures ranged from 49 to 118 mm Hg (mean, 68). Of the 33 cases in which lung tissue was evaluated microscopically, 28 (85%) were of the plexogenic variant of pulmonary arterial hypertension. Of the other five cases examined histologically, three consisted of thrombotic pulmonary arteriopathy (one was due to recurrent thromboembolism, and the other two were due to in situ thrombosis), and two were of pulmonary venoocclusive disease. No correlation existed between either CD4 counts or a history of pulmonary infections and the development of pulmonary hypertension. In 15 of the 88 patients (17%), confounding factors for hypertensive pulmonary vascular disease were present, including coexisting liver disease in 13 and coagulation abnormalities in 2. In 83% of the patients, the development of pulmonary hypertension seems to have been related primarily to the chronic HIV infection. Pulmonary hypertension was more rapidly progressive in patients with HIV or AIDS than in those with primary pulmonary hypertension; the reported time intervals between onset of symptoms and diagnosis were 6 months and 30 months, respectively. The 1-year survival rate for patients with HIV and pulmonary hypertension was 51%, based on the follow-up data compiled from the 63 patients in whom it was described; this compares with a 1-year survival rate of 68% for patients with primary pulmonary hypertension. Death was considered a direct consequence of pulmonary hypertension in 29 (76%) of the 38 fatal cases.


Subject(s)
HIV Infections/complications , Hypertension, Pulmonary/virology , Adult , HIV Infections/pathology , HIV Infections/physiopathology , Humans , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Male , Pulmonary Artery/pathology
10.
Cancer Genet Cytogenet ; 95(2): 173-7, 1997 Jun.
Article in English | MEDLINE | ID: mdl-9169037

ABSTRACT

In a prospective study of 93 consecutive untreated patients with B-cell chronic lymphocytic leukemia, we examined the clinical relevance of surface immunoglobulin (sIg) heavy chain (HC) and light chain (LC) isotypes, CD11c or CD25 expression, and the presence of trisomy 12 by fluorescence in situ hybridization (FISH). Careful morphologic evaluation was performed to exclude patients with other forms of chronic lymphoid leukemias, including mantle cell lymphoma, prolymphocytic leukemia, and leukemia phase of lymphoma. In addition, clonally restricted sIg and CD5 surface determinant were expressed in all patients. Clinical presentation, including blood cell counts, clinical stage, and organomegaly, did not correlate with any of the measured variables. After a median follow-up period of 3 years, the particular HC or LC isotype or CD11c expression did not correlate with either disease progression or treatment-free survival. However, trisomy 12 and CD25 expressions were both associated with accelerated disease progression and a shorter treatment-free survival time. Our results confirm the adverse prognostic significance of trisomy 12 expression in chronic lymphocytic leukemia and suggest that CD25 expression may have an unfavorable clinical impact.


Subject(s)
Chromosomes, Human, Pair 12 , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Trisomy , Adult , Aged , Aged, 80 and over , Female , Humans , Immunophenotyping , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Male , Middle Aged
11.
Leuk Lymphoma ; 42(5): 901-11, 2001.
Article in English | MEDLINE | ID: mdl-11697645

ABSTRACT

Myelofibrosis with myeloid metaplasia (MMM) is a chronic myeloproliferative disorder in which the accumulation and growth of circulating myeloid progenitors in the spleen lead to pathologic enlargement of the organ with resulting mechanical discomfort, hypercatabolic symptoms, anemia, thrombocytopenia, and portal hypertension. Medical therapy and splenic irradiation may be of benefit in certain patients, yet many may still require splenectomy to palliate their symptoms. Although there is no clear survival advantage to splenectomy in MMM, the procedure can result in substantial palliation of symptoms. However, the surgical procedure is associated with an approximately 9% mortality rate, and the postsplenectomy occurrence of extreme thrombocytosis, hepatomegaly, and leukemic transformation is of major concern. The management of splenomegaly and the role of splenectomy in MMM are discussed in this review.


Subject(s)
Primary Myelofibrosis/surgery , Splenectomy/adverse effects , Disease Management , Humans , Palliative Care , Primary Myelofibrosis/complications , Splenectomy/mortality , Splenomegaly/etiology , Splenomegaly/surgery , Treatment Outcome
12.
Leukemia ; 28(8): 1657-65, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24451410

ABSTRACT

Synergistic molecular vulnerabilities enhancing hypomethylating agents in myeloid malignancies have remained elusive. RNA-interference drug modifier screens identified antiapoptotic BCL-2 family members as potent 5-Azacytidine-sensitizing targets. In further dissecting BCL-XL, BCL-2 and MCL-1 contribution to 5-Azacytidine activity, siRNA silencing of BCL-XL and MCL-1, but not BCL-2, exhibited variable synergy with 5-Azacytidine in vitro. The BCL-XL, BCL-2 and BCL-w inhibitor ABT-737 sensitized most cell lines more potently compared with the selective BCL-2 inhibitor ABT-199, which synergized with 5-Azacytidine mostly at higher doses. Ex vivo, ABT-737 enhanced 5-Azacytidine activity across primary AML, MDS and MPN specimens. Protein levels of BCL-XL, BCL-2 and MCL-1 in 577 AML patient samples showed overlapping expression across AML FAB subtypes and heterogeneous expression within subtypes, further supporting a concept of dual/multiple BCL-2 family member targeting consistent with RNAi and pharmacologic results. Consequently, silencing of MCL-1 and BCL-XL increased the activity of ABT-199. Functional interrogation of BCL-2 family proteins by BH3 profiling performed on patient samples significantly discriminated clinical response versus resistance to 5-Azacytidine-based therapies. On the basis of these results, we propose a clinical trial of navitoclax (clinical-grade ABT-737) combined with 5-Azacytidine in myeloid malignancies, as well as to prospectively validate BH3 profiling in predicting 5-Azacytidine response.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Azacitidine/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Proto-Oncogene Proteins c-bcl-2/physiology , Biphenyl Compounds/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Cell Line, Tumor , Humans , Myeloid Cell Leukemia Sequence 1 Protein/antagonists & inhibitors , Myeloid Cell Leukemia Sequence 1 Protein/physiology , Myeloproliferative Disorders/drug therapy , Nitrophenols/pharmacology , Piperazines/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , RNA Interference , Sulfonamides/pharmacology , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/physiology
15.
Leuk Suppl ; 1(Suppl 1): S2-7, 2012 May.
Article in English | MEDLINE | ID: mdl-27175229

ABSTRACT

The myeloproliferative neoplasm, myelofibrosis (MF), has only one therapeutic intervention that is potentially curative in these individuals, specifically that of allogeneic stem cell transplantation (ASCT). ASCT has been utilized up to this juncture, primarily in younger individuals with higher risk disease. There is more limited data on outcomes in individuals over the age of 60 years. The choice of an individualized therapeutic intervention for a patient with MF is a very complex issue and is dependent on several factors. The first factor being their overall prognosis with their illness (which can vary from a median of 2 years in high-risk patients to over 10 years in low-risk patients) and the potential impact of a therapeutic intervention not only on survival but also on quality of life. Current available therapies have been strictly palliative for disease-associated anemia and/or splenomegaly. At present, we have a new generation of inhibitors of JAK2 (Ruxolitinib, CYT387, SB1518, TG101348, with others in development), which have been shown to improve splenomegaly, improve symptomatic burden of illness and improve quality of life. In addition, these inhibitors of JAK2 may have an impact on the natural history of MF, but confirmation of the presence and degree of this impact is still pending. Clinical availability of JAK2 inhibitors may alter the timing of transplant in marginal transplant candidates (that is, those over the age of 60), may have a role preceding ASCT to improve spleen size and performance status before transplant and might be frontline therapy in intermediate and high-risk patients who are not candidates for ASCT.

16.
Leukemia ; 25(2): 301-4, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21052089

ABSTRACT

In a previous study, we reported on the safety and efficacy of low-dose (0.5 mg) pomalidomide and prednisone and pomalidomide alone (2 mg/day), for the treatment of anemia associated with myelofibrosis (MF). The current study examined the value of low-dose pomalidomide alone. The main eligibility criterion was transfusion-dependency or hemoglobin <10 gm per 100 ml. Anemia response was assessed by International Working Group criteria. Pomalidomide (0.5 mg/day) was given to 58 patients (median age 68 years); 46 (79%) were transfusion-dependent and 42 were JAK2V617F positive. Anemia response was documented only in the presence of JAK2V617F (24 vs 0%; P=0.03) but was not further affected by mutant allele burden (P=0.39); 9 of the 10 anemia responders became transfusion independent. Anemia response in JAK2V617F-positive patients was predicted by the presence of pomalidomide-induced basophilia in the first month of therapy (38 vs 6%; P=0.02) or absence of marked splenomegaly (38 vs 11%; P=0.05). A total of 14 (58%) of 24 patients with a platelet count of ≤ 100 × 10(9) cells/l experienced a >50% increment in platelet count. There were no spleen responses. Grade 3 or 4 thrombocytopenia/neutropenia occurred in 2%/0% of patients. Low-dose pomalidomide is effective in the treatment of anemia associated with JAK2V617F-positive MF; response is predicted by early drug-induced basophilia.


Subject(s)
Anemia/drug therapy , Primary Myelofibrosis/drug therapy , Thalidomide/analogs & derivatives , Aged , Aged, 80 and over , Anemia/pathology , Anemia/therapy , Basophils/pathology , Erythrocyte Transfusion , Female , Humans , Janus Kinase 2/genetics , Male , Middle Aged , Mutation , Primary Myelofibrosis/pathology , Primary Myelofibrosis/therapy , Thalidomide/administration & dosage
17.
Leukemia ; 22(3): 474-86, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18185525

ABSTRACT

Optimal clinical management of patients with primary myelofibrosis and post-essential thrombocythemia/polycythemia vera myelofibrosis is a challenge, given the typically advanced age of presentation and variability of the disease course and prognosis. Current medical therapeutic options have not demonstrated an impact on the disease course, which exceeds the palliation of disease-related extramedullary hematopoiesis and alleviation of cytopenias. In contrast, allogeneic stem cell transplantation (SCT) can lead to 'cure' but is limited due to patient's age or comorbidities. Currently, in patients, who are reasonable candidates, SCT (frequently with a reduced intensity conditioning regimen) is employed for intermediate- to high-risk disease. Current pharmaco-medical therapy is used as a bridge to transplant, or instead of transplant in poor transplant candidates. Pathogenetic insights, especially the discovery of the Janus kinase (JAK)2(V617F) mutation, have ushered in a host of new potential therapeutic agents that may augment the role of medical therapy. Similarly, the boundaries of transplantation continue to alter with strategies that decrease conditioning-related toxicity, improved antimicrobial prophylaxis and decreased graft-versus-host disease. The potential for continued improvements in both medical and transplant therapy suggests that for the immediate future the optimal choices for an individual patient will remain potentially volatile and present complex decisions.


Subject(s)
Cytostatic Agents/therapeutic use , Disease Management , Hematopoietic Stem Cell Transplantation , Primary Myelofibrosis/therapy , Adult , Androgens/therapeutic use , Clinical Trials as Topic/statistics & numerical data , Cytostatic Agents/adverse effects , Drugs, Investigational/therapeutic use , Erythropoietin/therapeutic use , Etanercept , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunoglobulin G/therapeutic use , Janus Kinase 2/genetics , Middle Aged , Myeloablative Agonists/administration & dosage , Myeloablative Agonists/adverse effects , Myeloablative Agonists/therapeutic use , Palliative Care , Polycythemia Vera/drug therapy , Polycythemia Vera/surgery , Primary Myelofibrosis/drug therapy , Primary Myelofibrosis/genetics , Primary Myelofibrosis/surgery , Prognosis , Receptors, Tumor Necrosis Factor/therapeutic use , Risk , Thrombocythemia, Essential/drug therapy , Thrombocythemia, Essential/surgery , Transplantation Conditioning/adverse effects , Transplantation Conditioning/methods , Treatment Outcome
18.
Leukemia ; 22(4): 756-61, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18216871

ABSTRACT

The clinical relevance of JAK2V617F allele burden in primary myelofibrosis (PMF) has not been previously studied. Bone marrow-derived DNA from 199 patients with PMF was subjected to qualitative (n=199) and quantitative (n=129) analysis for V617F. Mutational frequency was 58% and median mutant allele burden ratio in V617F-positive patients was 29% (range, 1-74%). Multivariable analysis identified older age, platelet count > or =100 x 10(9) l(-1) and peripheral blood blast percentage <3% as being associated with a positive mutational status. The mere presence of the mutation did not affect the incidence of thrombosis (P=0.78), overall survival (P=0.22) or leukemia-free survival (P=0.5). The 129 patients with allele burden information were divided into four groups: V617F-negative (n=53) and V617F-positive with mutant allele burden in the lower quartile (n=19), middle quartiles (n=38) or upper quartile (n=19) range. Kaplan-Meier plots revealed significantly shortened overall (P=0.0008) and leukemia-free (P=0.01) survival for the lower quartile, but not for upper quartile allele burden group; independent prognostic relevance was validated by multivariable analysis. We conclude that low V617F allele burden in PMF might indicate the presence of an overriding V617F-negative clone that confers a more aggressive disease phenotype.


Subject(s)
Janus Kinase 2/genetics , Mutation, Missense , Primary Myelofibrosis/genetics , Primary Myelofibrosis/mortality , Adult , Age Factors , Aged , Aged, 80 and over , Alleles , Bone Marrow Examination , Disease-Free Survival , Female , Gene Frequency , Humans , Male , Middle Aged , Platelet Count , Primary Myelofibrosis/epidemiology , Survival Rate
20.
Blood ; 95(7): 2226-33, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10733489

ABSTRACT

In a 20-year period, 223 patients (median age, 64.8 years) with myelofibrosis with myeloid metaplasia (MMM) had therapeutic splenectomy at our institution. Primary indications for surgery were transfusion-dependent anemia (45.3%), symptomatic splenomegaly (39. 0%), portal hypertension (10.8%), and severe thrombocytopenia (4.9%). Operative mortality and morbidity rates were 9% and 31%, respectively. The 203 survivors of surgery had a median postsplenectomy survival time (PSS) of 27 months (range, 0-155). Among preoperative variables, thrombocytopenia (platelet count less than 100 x 10(9/)L) and nonhypercellular bone marrow were identified as independent risk factors for decreased PSS. Durable remissions in constitutional symptoms, transfusion-dependent anemia, portal hypertension, and severe thrombocytopenia were achieved in 67%, 23%, 50%, and 0% of the patients, respectively. Histologic or cytogenetic features of bone marrow obtained before splenectomy did not predict a response in cytopenias. After splenectomy, substantial enlargement of the liver and marked thrombocytosis occurred in 16.1% and 22.0% of the patients, respectively. The thrombocytosis was associated with an increased risk of perioperative thrombosis and decreased PSS. The rate of blast transformation (BT) was 16.3%, and the risk of BT was higher in the presence of increased spleen mass and preoperative thrombocytopenia. However, the PSS of patients with BT was not significantly different from that of patients without BT. We conclude that presplenectomy thrombocytopenia in MMM may be a surrogate for advanced disease and is associated with an increased risk of BT and inferior PSS. However, the development of BT after splenectomy may not affect overall survival and does not undermine the palliative role of the procedure for the other indications.


Subject(s)
Primary Myelofibrosis/surgery , Splenectomy , Adolescent , Adult , Aged , Aged, 80 and over , Anemia/etiology , Female , Hepatomegaly , Humans , Hypertension, Portal/etiology , Lymphocyte Activation , Male , Middle Aged , Postoperative Complications , Primary Myelofibrosis/complications , Primary Myelofibrosis/mortality , Splenomegaly , Survival Rate , Thrombocytopenia/etiology , Thrombocytosis
SELECTION OF CITATIONS
SEARCH DETAIL