Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 88
Filter
1.
Blood ; 143(16): 1646-1655, 2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38142448

ABSTRACT

ABSTRACT: Polycythemia vera (PV) is a myeloproliferative neoplasm characterized by clonal proliferation of hematopoietic progenitor cells and is associated with an increased risk of thrombotic events (TEs). Established risk factors for TEs in patients with PV include advanced age, TE history, and elevated hematocrit. Although an association of TE with elevated white blood cell (WBC) counts has been suggested by retrospective studies, this relationship needs further validation. The prospective observational study of patients with polycythemia vera in US clinical practices (REVEAL) study collected prospective clinical data from 2510 patients with PV with a median follow-up of 44.7 months (range, 2-59 months) from enrollment. Using time-dependent covariate Cox proportional hazards models, blood counts were individually modeled with sex, age, disease duration, TE history at enrollment (baseline covariates), and treatment (time-dependent covariate). Analysis of 2271 participants identified 142 TEs in 106 patients. Significant associations with initial TE occurrence during the study period were observed for hematocrit level >45% (hazard ratio [HR], 1.84; 95% confidence interval [95% CI], 1.234-2.749; P = .0028) and WBCs >11 × 109/L (HR, 2.35; 95% CI, 1.598-3.465; P < .0001). Elevated WBC count was significantly associated with initial TE occurrence in both low-risk and high-risk PV. When hematocrit was controlled at ≤45%, WBC count >12 × 109/L was significantly associated with TE occurrence (HR, 1.95; 95% CI, 1.066-3.554; P = .0300). The results support incorporation of WBC count into PV risk stratification and studies of treatment strategies, and indicate the importance of controlling both hematocrit and WBC count in disease management. This trial was registered at www.clinicaltrials.gov as #NCT02252159.


Subject(s)
Polycythemia Vera , Thrombosis , Humans , Polycythemia Vera/drug therapy , Retrospective Studies , Prospective Studies , Thrombosis/etiology , Risk Factors , Leukocyte Count
2.
Br J Haematol ; 202(4): 879-882, 2023 08.
Article in English | MEDLINE | ID: mdl-37226361

ABSTRACT

Thrombotic thrombocytopenic purpura (TTP) is not uncommonly seen in pregnancy, either with the first episode or with the exacerbation of known disease. The management of TTP in pregnancy can be challenging if there is refractoriness to the use of therapeutic plasma exchange (TPE) and high-dose corticosteroids. Caplacizumab, a vWF-directed humanized antibody fragment, is approved for the treatment of acquired TTP but there is sparse data on its use in pregnant patients. Antenatal and peripartum haemorrhage is a theoretical concern with the use of the medication in the obstetric population. However, as options for treatment of TTP in the patients who have refractory disease are significantly limited, off-label use of caplacizumab to achieve disease control and prevent maternofetal morbidity and mortality is a reasonable consideration. This article described the successful use of caplacizumab in a pregnant patient with acquired TTP and the associated favourable outcome. The patient suffered an exacerbation following initial TPE and became refractory to both plasma exchange and high-dose corticosteroids. Off-label use of caplacizumab resulted in hematologic recovery and successful delivery of a healthy neonate. This case represents a contribution to the sparse literature on the use of this effective medication in an often-challenging clinical situation.


Subject(s)
Purpura, Thrombotic Thrombocytopenic , Pregnancy , Infant, Newborn , Humans , Female , Purpura, Thrombotic Thrombocytopenic/drug therapy , Hemorrhage/therapy , Plasma Exchange , Adrenal Cortex Hormones/therapeutic use , ADAMTS13 Protein
3.
Leuk Lymphoma ; 64(2): 292-299, 2023 02.
Article in English | MEDLINE | ID: mdl-36301740

ABSTRACT

The management of myelofibrosis has improved following approval of the JAK1/JAK2 inhibitor, ruxolitinib. This agent laid the foundation for JAK inhibitor therapy, yet limitations have included myelosuppression and other adverse events (skin cancer, weight gain, and infection), as well as loss of response. Recently, two additional JAK inhibitors were approved for use in myelofibrosis. Fedratinib can be used front-line and has demonstrated impressive responses as a salvage option after ruxolitinib loss of response. Previously, patients with severe thrombocytopenia had limited treatment options; approval of pacritinib offers an option to address splenomegaly and/or symptoms in these patients. A significant unmet need has been the treatment of anemia; momelotinib (not approved at the time of writing) has demonstrated spleen, symptom, and anemia responses. The possibility of having four approved options for myelofibrosis may be soon realized. This speaks to progress in the past decade, though achieving clinical and molecular remissions remain paramount.


Subject(s)
Primary Myelofibrosis , Humans , Primary Myelofibrosis/diagnosis , Primary Myelofibrosis/drug therapy , Protein Kinase Inhibitors/adverse effects , Janus Kinase 2/genetics , Pyrazoles/adverse effects , Nitriles/therapeutic use
4.
J Natl Compr Canc Netw ; 20(9): 1033-1062, 2022 09.
Article in English | MEDLINE | ID: mdl-36075392

ABSTRACT

The classic Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) consist of myelofibrosis, polycythemia vera, and essential thrombocythemia and are a heterogeneous group of clonal blood disorders characterized by an overproduction of blood cells. The NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for MPN were developed as a result of meetings convened by a multidisciplinary panel with expertise in MPN, with the goal of providing recommendations for the management of MPN in adults. The Guidelines include recommendations for the diagnostic workup, risk stratification, treatment, and supportive care strategies for the management of myelofibrosis, polycythemia vera, and essential thrombocythemia. Assessment of symptoms at baseline and monitoring of symptom status during the course of treatment is recommended for all patients. This article focuses on the recommendations as outlined in the NCCN Guidelines for the diagnosis of MPN and the risk stratification, management, and supportive care relevant to MF.


Subject(s)
Myeloproliferative Disorders , Polycythemia Vera , Primary Myelofibrosis , Thrombocythemia, Essential , Adult , Humans , Medical Oncology , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/therapy , Polycythemia Vera/diagnosis , Primary Myelofibrosis/diagnosis , Thrombocythemia, Essential/diagnosis
5.
Curr Hematol Malig Rep ; 16(5): 483-489, 2021 10.
Article in English | MEDLINE | ID: mdl-34499329

ABSTRACT

PURPOSE OF REVIEW: The management of myelofibrosis is risk-adapted when considering transplant eligibility and symptom-directed, prioritizing the most burdensome symptoms for the patient. Unfortunately, myelofibrosis-anemia is common, multifactorial in its origin, and impactful regarding prognosis. While clinical trials are advised, not all patients have convenient access, and therefore, hematologists should be aware of the data supporting the use of conventional agents such as erythropoietin-stimulating agents, steroid treatments (danazol and prednisone), and immunomodulatory drugs (thalidomide and lenalidomide). This review summarizes the conventional approach to treating myelofibrosis-anemia and highlights recent data from 3 novel agents that are under phase 3 evaluation. RECENT FINDINGS: Momelotonib is a JAK1/2 and ACVR1 inhibitor that has demonstrated not only improvements in splenomegaly and symptoms, but also amelioration of anemia on the SIMPLIFY 1 and 2 clinical trial program. This may occur through suppression of hepcidin production. Luspatercept promotes late-stage hematopoiesis, and the phase 2 study has shown promise in ameliorating anemia as a monotherapy, and especially in combination with ruxolitinib. Finally, CP-0160, a BET inhibitor, has shown efficacy as an anemia-directed agent, when used as monotherapy and in combination. This agent reduces cytokine production and promotes erythroid differentiation. Results have been presented for patients previously treated with JAK inhibitors, as well as those who were naïve to JAK inhibitor therapy. Safety and effectiveness are reviewed for both conventional and selected novel agents used in the treatment of MF-anemia. A practical approach to treatment is presented, and data from ASH 2020 are presented.


Subject(s)
Anemia/drug therapy , Anemia/etiology , Hematinics/therapeutic use , Immunologic Factors/therapeutic use , Primary Myelofibrosis/complications , Steroids/therapeutic use , Clinical Trials, Phase III as Topic , Disease Management , Drug Discovery , Humans , Primary Myelofibrosis/drug therapy
6.
Hepatology ; 74(5): 2735-2744, 2021 11.
Article in English | MEDLINE | ID: mdl-34021505

ABSTRACT

BACKGROUND AND AIMS: Extrahepatic portal vein occlusion (EHPVO) from portal vein thrombosis is a rare condition associated with substantial morbidity and mortality. The purpose of this study is to investigate the efficacy of transjugular intrahepatic portosystemic shunts (TIPS) for the treatment of chronic EHPVO, cavernomatosis, and mesenteric venous thrombosis in adults without cirrhosis who are refractory to standard-of-care therapy. APPROACH AND RESULTS: Thirty-nine patients with chronic EHPVO received TIPS. Laboratory parameters and follow-up were assessed at 1, 3, 6, 12, and 24 months, and every 6 months thereafter. Two hepatologists adjudicated symptom improvement attributable to mesenteric thrombosis and EHPVO before/after TIPS. Kaplan-Meier was used to assess primary and overall TIPS patency, assessing procedural success. Adverse events, radiation exposure, hospital length-of-stay and patency were recorded. Cavernoma was present in 100%, with TIPS being successful in all cases using splenic, mesenteric, and transhepatic approaches. Symptom improvement was noted in 26 of 30 (87%) at 6-month follow-up. Twelve patients (31%) experienced TIPS thrombosis. There were no significant long-term laboratory adverse events or deaths. At 36 months, freedom from primary TIPS thrombosis was 63%; following secondary interventions, overall patency was increased to 81%. CONCLUSIONS: TIPS in chronic, noncirrhotic EHPVO with cavernomas and mesenteric venous thrombosis is technically feasible and does not adversely affect liver function. Most patients demonstrate subjective and objective benefit from TIPS. Improvement in patency rates are needed with proper timing of adjuvant anticoagulation.


Subject(s)
Anticoagulants/administration & dosage , Mesenteric Ischemia/therapy , Portal Vein/surgery , Portasystemic Shunt, Transjugular Intrahepatic/adverse effects , Venous Thrombosis/therapy , Adult , Aged , Chronic Disease/therapy , Combined Modality Therapy/methods , Feasibility Studies , Female , Humans , Male , Middle Aged , Portal Vein/pathology , Retrospective Studies , Treatment Outcome , Vascular Patency
8.
Cancer Discov ; 11(6): 1398-1410, 2021 06.
Article in English | MEDLINE | ID: mdl-33579786

ABSTRACT

The myeloproliferative neoplasms (MPN) frequently progress to blast phase disease, an aggressive form of acute myeloid leukemia. To identify genes that suppress disease progression, we performed a focused CRISPR/Cas9 screen and discovered that depletion of LKB1/Stk11 led to enhanced in vitro self-renewal of murine MPN cells. Deletion of Stk11 in a mouse MPN model caused rapid lethality with enhanced fibrosis, osteosclerosis, and an accumulation of immature cells in the bone marrow, as well as enhanced engraftment of primary human MPN cells in vivo. LKB1 loss was associated with increased mitochondrial reactive oxygen species and stabilization of HIF1α, and downregulation of LKB1 and increased levels of HIF1α were observed in human blast phase MPN specimens. Of note, we observed strong concordance of pathways that were enriched in murine MPN cells with LKB1 loss with those enriched in blast phase MPN patient specimens, supporting the conclusion that STK11 is a tumor suppressor in the MPNs. SIGNIFICANCE: Progression of the myeloproliferative neoplasms to acute myeloid leukemia occurs in a substantial number of cases, but the genetic basis has been unclear. We discovered that loss of LKB1/STK11 leads to stabilization of HIF1a and promotes disease progression. This observation provides a potential therapeutic avenue for targeting progression.This article is highlighted in the In This Issue feature, p. 1307.


Subject(s)
AMP-Activated Protein Kinases/genetics , Genes, Tumor Suppressor , Leukemia, Myeloid, Acute/genetics , Animals , Disease Models, Animal , Disease Progression , Mice , Mice, Inbred C57BL , Mutation , Myeloproliferative Disorders/genetics
9.
J Natl Compr Canc Netw ; 18(9): 1248-1269, 2020 09.
Article in English | MEDLINE | ID: mdl-32886902

ABSTRACT

Eosinophilic disorders and related syndromes represent a heterogeneous group of neoplastic and nonneoplastic conditions, characterized by more eosinophils in the peripheral blood, and may involve eosinophil-induced organ damage. In the WHO classification of myeloid and lymphoid neoplasms, eosinophilic disorders characterized by dysregulated tyrosine kinase (TK) fusion genes are recognized as a new category termed, myeloid/lymphoid neoplasms with eosinophilia and rearrangement of PDGFRA, PDGFRB or FGFR1 or with PCM1-JAK2. In addition to these aforementioned TK fusion genes, rearrangements involving FLT3 and ABL1 genes have also been described. These new NCCN Guidelines include recommendations for the diagnosis, staging, and treatment of any one of the myeloid/lymphoid neoplasms with eosinophilia (MLN-Eo) and a TK fusion gene included in the 2017 WHO Classification, as well as MLN-Eo and a FLT3 or ABL1 rearrangement.


Subject(s)
Eosinophilia , Myeloproliferative Disorders , Neoplasms , Eosinophilia/diagnosis , Eosinophilia/genetics , Humans , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/therapy , Oncogene Proteins, Fusion/genetics
10.
Bone ; 141: 115559, 2020 12.
Article in English | MEDLINE | ID: mdl-32730929

ABSTRACT

BACKGROUND: Two weekly infusions of ferric carboxymaltose (FCM) are commonly prescribed for treatment of iron-deficiency anemia. However, administration of FCM increases intact levels of fibroblast growth factor 23 (FGF23), which causes hypophosphatemia due to renal phosphate wasting, calcitriol deficiency and secondary hyperparathyroidism. The adverse effects of FCM on mineral metabolism and bone health emerged from case reports and secondary analyses of trials. Data on these safety signals with FCM in clinical practice are limited because markers of mineral and bone metabolism are not routinely checked. METHODS: To obtain real-world experience with effects of FCM on mineral and bone metabolism, we conducted a prospective observational study of 16 women who were managed at a single-center hematology clinic for iron-deficiency anemia. From October 2016 to February 2018, all participants received two weekly infusions of FCM at a hematology infusion clinic. We hypothesized that FCM would decrease phosphate, increase intact FGF23 (iFGF23), and decrease c-terminal FGF23 (cFGF23). Secondary outcomes were changes in hemoglobin, iron indices, urine fractional excretion of phosphate (FePi), parathyroid hormone (PTH), calcitriol, calcium, osteocalcin, and bone-specific alkaline phosphatase (BAP). FCM was administered at weeks zero and one, and we measured laboratory values at weeks zero, one, two, and five of the study. We used linear mixed models to analyze the significance of the changes in laboratory values over time. RESULTS: After two FCM infusions, nearly all (14 of 16) participants developed hypophosphatemia. iFGF23 increased, cFGF23 decreased, and phosphate decreased significantly from week zero to week two (iFGF23 increased by +134.0% [40.6, 305.8], p < 0.001; cFGF23 decreased by -516.3% [-1332.7, -142.7], p = 0.002; phosphate decreased by -49.8 ± 15.4%, p < 0.001). There was also a significant increase in FePi, PTH, and BAP and a significant decrease in calcitriol and calcium from week zero to week two. There was no significant change in osteocalcin during this time period. iFGF23, but not PTH, was independently associated with decreased phosphate. iFGF23 was also significantly associated with decrease in calcitriol from week zero to week two. Elevation in BAP suggests disordered bone mineralization in response to FCM therapy. CONCLUSION: In this prospective observational study of women with iron deficiency anemia, two FCM infusions significantly altered markers of bone mineralization and mineral metabolism. The results suggest that FCM should be used cautiously in the treatment of iron-deficiency anemia.


Subject(s)
Anemia, Iron-Deficiency , Female , Ferric Compounds , Fibroblast Growth Factor-23 , Humans , Maltose/analogs & derivatives , Minerals
11.
Blood Adv ; 4(9): 1965-1973, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32384540

ABSTRACT

Allogeneic hematopoietic cell transplantation (HCT) is the only curative therapy for myelofibrosis (MF). In this large multicenter retrospective study, overall survival (OS) in MF patients treated with allogeneic HCT (551 patients) and without HCT (non-HCT) (1377 patients) was analyzed with Cox proportional hazards model. Survival analysis stratified by the Dynamic International Prognostic Scoring System (DIPSS) revealed that the first year of treatment arm assignment, due to upfront risk of transplant-related mortality (TRM), HCT was associated with inferior OS compared with non-HCT (non-HCT vs HCT: DIPSS intermediate 1 [Int-1]: hazard ratio [HR] = 0.26, P < .0001; DIPSS-Int-2 and higher: HR, 0.39, P < .0001). Similarly, in the DIPSS low-risk MF group, due to upfront TRM risk, OS was superior with non-HCT therapies compared with HCT in the first-year post treatment arm assignment (HR, 0.16, P = .006). However, after 1 year, OS was not significantly different (HR, 1.38, P = .451). Beyond 1 year of treatment arm assignment, an OS advantage with HCT therapy in Int-1 and higher DIPSS score patients was observed (non-HCT vs HCT: DIPSS-Int-1: HR, 2.64, P < .0001; DIPSS-Int-2 and higher: HR, 2.55, P < .0001). In conclusion, long-term OS advantage with HCT was observed for patients with Int-1 or higher risk MF, but at the cost of early TRM. The magnitude of OS benefit with HCT increased as DIPSS risk score increased and became apparent with longer follow-up.


Subject(s)
Hematopoietic Stem Cell Transplantation , Primary Myelofibrosis , Humans , Primary Myelofibrosis/therapy , Prognosis , Retrospective Studies , Transplantation, Homologous
12.
Curr Hematol Malig Rep ; 15(4): 261-267, 2020 08.
Article in English | MEDLINE | ID: mdl-32399765

ABSTRACT

PURPOSE OF REVIEW: Thrombocytosis is common to all myeloproliferative neoplasms (MPN), including essential thrombocythemia (ET), polycythemia vera (PV), and myelofibrosis. Despite the traditionally held belief amongst many clinicians that thrombocytosis correlates with thrombosis risk, there is little evidence in the literature to support that claim. Herein we critically analyze the literature to better understand the relationship between thrombocytosis and risk of thrombosis in MPN. RECENT FINDINGS: Both retrospective and prospective studies argue against associations between thrombocytosis and risk of thrombosis in patients with ET and PV. Rather, most studies suggest that the presence of extreme thrombocytosis is instead associated with an increased risk of hemorrhagic events, a paradoxical phenomenon with important clinical implications. Thrombosis risk has a multifactorial set of etiologies in MPNs. While qualitative abnormalities of the platelets may contribute, associations between platelet quantity and thrombosis risk are weak in MPN patients.


Subject(s)
Blood Platelets/metabolism , Hemorrhage/etiology , Hemostasis , Myeloproliferative Disorders/complications , Thrombocytosis/etiology , Thrombosis/etiology , Female , Hemorrhage/blood , Humans , Middle Aged , Myeloproliferative Disorders/blood , Prognosis , Risk Assessment , Risk Factors , Thrombocytosis/blood , Thrombosis/blood
13.
Clin Lymphoma Myeloma Leuk ; 20(4): 219-225, 2020 04.
Article in English | MEDLINE | ID: mdl-32063527

ABSTRACT

BACKGROUND: Polycythemia vera (PV) is associated with increased blood cell counts, risk of thrombosis, and symptoms including fatigue and pruritus. National guidelines support the use of hydroxyurea (HU) in high-risk patients or those with some other clinical indication for cytoreduction. PATIENTS AND METHODS: REVEAL is a prospective, observational study designed to collect data pertaining to demographics, disease burden, clinical management, patient-reported outcomes, and health care resource utilization of patients with PV in the United States. In this analysis, HU treatment patterns and outcomes were assessed from 6 months prior to enrollment to the time of discontinuation, death, or data cutoff. RESULTS: Of the 1381 patients who received HU for ≥ 3 months, the median HU exposure was 23.6 months (range, 3.1-38.5 months). The most common maximum daily HU doses were 1000 mg (30.6%) and 500 mg (30.1%); only 6.4% received ≥ 2 g/d HU. Approximately one-third (32.3%) of patients had dose adjustments, 23.8% had dose interruptions, and 257 (18.6%) discontinued HU. The most common reasons for HU discontinuations and interruptions were adverse events/intolerance (37.1% and 54.5%, respectively) and lack of efficacy (35.5% and 22.1%, respectively). Of those who received HU for ≥ 3 months, 57.1% had hematocrit values > 45% on ≥ 1 occasion, 33.1% continued to receive phlebotomies, and 27.4% had uncontrolled myeloproliferation. CONCLUSION: The results of this analysis emphasize the need for active management of patients with PV with appropriate HU dose titration to maintain blood count control while monitoring for signs and symptoms of HU intolerance.


Subject(s)
Hydroxyurea/administration & dosage , Polycythemia Vera/blood , Polycythemia Vera/drug therapy , Adult , Aged , Aged, 80 and over , Blood Cell Count , Female , Humans , Longitudinal Studies , Male , Middle Aged , Prospective Studies , United States
14.
Phlebology ; 35(6): 402-408, 2020 Jul.
Article in English | MEDLINE | ID: mdl-31821779

ABSTRACT

OBJECTIVES: To examine the prescribing patterns and outcomes of antithrombotic regimens after venous stent placement. METHODS: A total of 87 patients who received inferior vena cava or iliofemoral venous stents were included in the study. A retrospective review was performed to determine the antithrombotic regimens and the subsequent rates of in-stent restenosis, stent thrombosis, and bleeding. RESULTS: The prescribing patterns of specific antithrombotic regimens were highly variable. In-stent restenosis and stent thrombosis events were observed in 13 of 63 patients (21%) with available follow-up imaging, while major bleeding events were noted in 6 of 87 patients (7%). Triple therapy appeared to reduce the odds of in-stent restenosis/ stent thrombosis when compared to dual antiplatelet therapy (OR = 0.07, P = 0.01). CONCLUSIONS: Substantial variability exists in antithrombotic therapy following venous stenting at our institution. This study demonstrated a reduction of in-stent restenosis/thrombosis events when utilizing triple therapy compared to antiplatelet-only regimens. However, larger prospective trials are needed to more accurately determine the relative risks and benefits of each antithrombotic regimen.


Subject(s)
Fibrinolytic Agents/pharmacology , Stents , Venous Thrombosis/drug therapy , Adult , Aged , Constriction, Pathologic , Female , Hemorrhage , Humans , Iliac Vein , Male , Middle Aged , Platelet Aggregation Inhibitors/pharmacology , Retrospective Studies , Tomography, X-Ray Computed , Vena Cava, Inferior/physiopathology , Young Adult
15.
Hematology Am Soc Hematol Educ Program ; 2019(1): 397-406, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31808903

ABSTRACT

Thrombotic and hemorrhagic complications are prevalent in patients with essential thrombocythemia, polycythemia vera, and myelofibrosis. Given the impact on morbidity and mortality, reducing the risk of thrombosis and/or hemorrhage is a major therapeutic goal. Historically, patients have been risk stratified on the basis of traditional factors, such as advanced age and thrombosis history. However, multiple factors contribute to the thrombotic tendency, including gender, mutational profile, inflammatory stress, and abnormal cell adhesion. Management includes cardiovascular risk reduction and use of antiplatelet therapy, depending on myeloproliferative neoplasm subtype and mutational status. Anticoagulation is a mainstay of therapy for those with venous thrombosis, but practice patterns remain heterogeneous. Cytoreduction is indicated for higher-risk patients, but efficacy may depend on the involved vascular bed. Management of special situations, such as unusual site thrombosis, bleeding, the perioperative period, and pregnancy, are especially challenging. In this article, risk factors and treatment strategies for myeloproliferative neoplasm thrombosis and bleeding, including special situations, are reviewed. Insights gained from recent studies may lead to the development of a more precise risk classification and tailored therapy.


Subject(s)
Budd-Chiari Syndrome , Hematologic Neoplasms , Hemorrhage , Myeloproliferative Disorders , von Willebrand Diseases , Adult , Budd-Chiari Syndrome/diagnosis , Budd-Chiari Syndrome/genetics , Budd-Chiari Syndrome/therapy , Female , Hematologic Neoplasms/diagnosis , Hematologic Neoplasms/genetics , Hematologic Neoplasms/therapy , Hemorrhage/diagnosis , Hemorrhage/genetics , Hemorrhage/therapy , Humans , Myeloproliferative Disorders/diagnosis , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/therapy , von Willebrand Diseases/diagnosis , von Willebrand Diseases/genetics , von Willebrand Diseases/therapy
16.
J Blood Med ; 10: 359-371, 2019.
Article in English | MEDLINE | ID: mdl-31695542

ABSTRACT

Polycythemia vera is a Philadelphia-negative chronic myeloproliferative neoplasm, characterized by erythrocytosis, which is unique, compared to essential thrombocytosis and primary myelofibrosis. Though longevity can usually be expected, vascular morbidity is associated with this condition, as well as a propensity to evolve into myelofibrosis (post-PV MF) and acute myeloid leukemia. In addition, patients can have a pronounced symptom burden. Herein, contributors to the symptomatic burden, as well as the thrombotic and transformative tendencies are reviewed. From a symptom perspective, some are explained by cytokine release, others by microvascular complications, whereas certain symptoms can herald disease evolution. Thrombosis has multifactorial contributors, including but not limited to gender, and inflammatory stress; investigators have recently hypothesized that microparticles and Neutrophil Extracellular Trap Formations may add to thrombotic burden. Finally, we examine the progression to post-PV MF as well as leukemic transformation, highlighting well-established risk factors including age and leukocytosis, certain treatments, and the presence of "non-driver" mutations.

17.
Blood ; 134(22): 1902-1911, 2019 11 28.
Article in English | MEDLINE | ID: mdl-31778549

ABSTRACT

Thrombotic and hemorrhagic complications are prevalent in patients with essential thrombocythemia, polycythemia vera, and myelofibrosis. Given the impact on morbidity and mortality, reducing the risk of thrombosis and/or hemorrhage is a major therapeutic goal. Historically, patients have been risk stratified on the basis of traditional factors, such as advanced age and thrombosis history. However, multiple factors contribute to the thrombotic tendency, including gender, mutational profile, inflammatory stress, and abnormal cell adhesion. Management includes cardiovascular risk reduction and use of antiplatelet therapy, depending on myeloproliferative neoplasm subtype and mutational status. Anticoagulation is a mainstay of therapy for those with venous thrombosis, but practice patterns remain heterogeneous. Cytoreduction is indicated for higher-risk patients, but efficacy may depend on the involved vascular bed. Management of special situations, such as unusual site thrombosis, bleeding, the perioperative period, and pregnancy, are especially challenging. In this article, risk factors and treatment strategies for myeloproliferative neoplasm thrombosis and bleeding, including special situations, are reviewed. Insights gained from recent studies may lead to the development of a more precise risk classification and tailored therapy.


Subject(s)
Anticoagulants/therapeutic use , Budd-Chiari Syndrome , Hematologic Neoplasms , Hemorrhage , Myeloproliferative Disorders , von Willebrand Diseases , Adult , Budd-Chiari Syndrome/blood , Budd-Chiari Syndrome/drug therapy , Budd-Chiari Syndrome/etiology , Budd-Chiari Syndrome/genetics , Female , Hematologic Neoplasms/blood , Hematologic Neoplasms/complications , Hematologic Neoplasms/drug therapy , Hematologic Neoplasms/genetics , Hemorrhage/blood , Hemorrhage/drug therapy , Hemorrhage/etiology , Hemorrhage/genetics , Humans , Male , Myeloproliferative Disorders/blood , Myeloproliferative Disorders/complications , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Sex Factors , von Willebrand Diseases/blood , von Willebrand Diseases/drug therapy , von Willebrand Diseases/etiology , von Willebrand Diseases/genetics
18.
Blood Cancer J ; 9(9): 74, 2019 09 11.
Article in English | MEDLINE | ID: mdl-31511492

ABSTRACT

Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by ineffective clonal hematopoiesis, splenomegaly, bone marrow fibrosis, and the propensity for transformation to acute myeloid leukemia. The discovery of mutations in JAK2, CALR, and MPL have uncovered activated JAK-STAT signaling as a primary driver of MF, supporting a rationale for JAK inhibition. However, JAK inhibition alone is insufficient for long-term remission and offers modest, if any, disease-modifying effects. Given this, there is great interest in identifying mechanisms that cooperate with JAK-STAT signaling to predict disease progression and rationally guide the development of novel therapies. This review outlines the latest discoveries in the biology of MF, discusses current clinical management of patients with MF, and summarizes the ongoing clinical trials that hope to change the landscape of MF treatment.


Subject(s)
Gene Amplification/genetics , Janus Kinase 2/antagonists & inhibitors , Primary Myelofibrosis/genetics , Protein Kinase Inhibitors/therapeutic use , Female , History, 21st Century , Humans , Male , Primary Myelofibrosis/pathology , Protein Kinase Inhibitors/pharmacology
19.
Cancer Treat Res ; 179: 159-178, 2019.
Article in English | MEDLINE | ID: mdl-31317487

ABSTRACT

The myeloproliferative neoplasms (MPNs) are clonal stem cell-derived diseases. This chapter focuses on the subcategory of Philadelphia (Ph) chromosome-negative classical MPNs, polycythemia vera (PV), essential thrombocythemia (ET), primary myelofibrosis (PMF). These MPNs are associated with both microvascular and macrovascular thrombosis, which may occur in the venous and arterial circulation. Erythrocytosis, leukocytosis, and increased JAK2V617F allele burden are known to be risk factors. In this chapter, we review the thrombotic and hemostatic manifestations of the Philadelphia (Ph) chromosome-negative classical MPNs, including the clinical manifestations, the pathophysiology, as well as management.


Subject(s)
Myeloproliferative Disorders/complications , Thrombosis , Bone Marrow Neoplasms/complications , Bone Marrow Neoplasms/genetics , Hemostasis/physiology , Humans , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/physiopathology , Philadelphia Chromosome , Polycythemia Vera/complications , Primary Myelofibrosis/complications , Thrombocythemia, Essential/complications , Thrombosis/diagnosis , Thrombosis/etiology , Thrombosis/physiopathology , Thrombosis/therapy
20.
Clin Lymphoma Myeloma Leuk ; 19(9): 579-584.e1, 2019 09.
Article in English | MEDLINE | ID: mdl-31303457

ABSTRACT

BACKGROUND: Approximately 50% of patients with polycythemia vera (PV) have PV-related symptoms at diagnosis; these symptoms might develop or worsen with time. Symptoms have been shown to negatively affect quality of life and interfere with daily activities. To our knowledge, an analysis to evaluate the relationship between blood count control and symptoms has not been published. PATIENTS AND METHODS: The Prospective Observational Study of Patients with Polycythemia Vera in US Clinical Practices (REVEAL; NCT02252159) is a multicenter, noninterventional, nonrandomized prospective observational study of patients with PV in the United States. Patients included were required to have a complete blood count result within 30 days before completing the at-enrollment Myeloproliferative Neoplasm Self-Assessment Form Total Symptom Score (MPN-SAF TSS). Symptom severity was compared between those who had blood count control versus those who did not. RESULTS: At the time of enrollment, 1714 patients (94.5%) were being managed with cytoreductive therapy; 468 patients (25.8%) had complete hematologic remission (CHR), 1614 patients (89.0%) had ≥1 controlled blood count, and 1122 patients (61.9%) had ≥2 controlled blood counts. Mean MPN-SAF TSSs were similar across patients in different blood count control groups. Fatigue was the most frequently reported symptom. The severity of individual symptoms, except those of pruritus and night sweats, was not affected by CHR or the number of blood counts that were controlled. CONCLUSION: Symptom burden in patients with PV can persist despite control of blood counts, which suggests some discordance between laboratory values and symptom burden. Consequently, regular monitoring of symptom burden should be factored into the assessment of disease control.


Subject(s)
Blood Cell Count , Polycythemia Vera/blood , Polycythemia Vera/diagnosis , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Disease Management , Female , Humans , Male , Middle Aged , Polycythemia Vera/complications , Polycythemia Vera/therapy , Prospective Studies , Severity of Illness Index , Symptom Assessment , Treatment Outcome , United States , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...