1
|
Cvetković M, Arsenović I, Smiljanić M, Sobas M, Bogdanović A, Leković D. Neutrophil-to-lymphocyte and platelet-to-lymphocyte ratio as novel prognostic biomarkers in BCR-ABL negative myeloproliferative neoplasms. Ann Hematol 2024; 103:4545-4556. [PMID: 39331155 DOI: 10.1007/s00277-024-06023-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
Higher neutrophil-to-lymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) have been associated with increased risk of thrombosis, cardiovascular mortality, but their role in myeloproliferative neoplasms (MPN) remains unclear. We analyzed NLR and PLR as prognostic markers for thrombosis and overall survival (OS) in the study that included 461 consecutive MPN patients who were diagnosed from 2018 to 2022 at University center. Twenty age-matched patients without hematological disorder were used as controls. NLR and PLR were significantly increased in whole MPN group compared to controls. NLR was highest in PV > PMF > ET (p < 0.001) while PLR was highest in ET > PMF > PV (p < 0.001). Thrombosis occurrence during follow-up correlated with NLR, NLR ≥ 4.5, presence of ≥ 2 CV factors and previous thrombosis. Arterial thrombosis was associated with previous thrombosis, NLR and NLR ≥ 4.5. Similarly in venous thrombosis previous thrombosis was risk factor, together with NLR, NLR ≥ 4.5, PLR, but also secondary malignancy and female gender. In multivariate Cox model, most important factors for thrombosis development during follow-up were previous thrombosis, NLR ≥ 4.5 and PLR ≥ 500; for arterial thrombosis, NLR ≥ 4.5 and previous thrombosis; for venous thrombosis PLR ≥ 500 and previous thrombosis. Patients with pre-PMF had significantly higher NLR than ET patients. In multivariate Cox regression model, most important factors associated with survival were NLR ≥ 4.5 and PLR ≥ 500. This study highlights strong prognostic correlation of NLR ≥ 4.5 and PLR ≥ 500 with development of thrombosis and OS in MPN. Besides previous thrombosis, most important factor associated with development of arterial thrombosis is NLR ≥ 4.5 and for venous PLR ≥ 500. Our results revealed that NLR ≥ 4.5 could be used as additional marker to distinguish ET from prePMF.
Collapse
Affiliation(s)
- Mirjana Cvetković
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovića, Belgrade, 11000, Serbia.
| | - Isidora Arsenović
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovića, Belgrade, 11000, Serbia
| | - Mihailo Smiljanić
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovića, Belgrade, 11000, Serbia
| | - Marta Sobas
- Department of Hematology, Blood Neoplasms and Bone Marrow Transplantation, Wroclaw Medical University, Wroclaw, Poland
| | - Andrija Bogdanović
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovića, Belgrade, 11000, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Danijela Leković
- Clinic of Hematology, University Clinical Centre of Serbia, 2 Dr Koste Todorovića, Belgrade, 11000, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
2
|
Krecak I, Lekovic D, Arsenovic I, Bogdanovic A, Holik H, Zekanovic I, Moric Peric M, Lucijanic M. Systemic Inflammatory Index in Polycythemia Vera and Its Prognostic Implications. J Clin Med 2024; 13:4459. [PMID: 39124725 PMCID: PMC11313665 DOI: 10.3390/jcm13154459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/25/2024] [Accepted: 07/27/2024] [Indexed: 08/12/2024] Open
Abstract
Background: This study aimed to evaluate the clinical and prognostic associations of the systemic inflammatory index (SII) in polycythemia vera (PV) patients. SII integrates information on absolute neutrophil (ANC), lymphocyte (ALC), and platelet counts into one index (calculated as ANCxALC/platelet count) and was previously shown to predict thrombotic and mortality risks in the general population. Methods: A total of 279 PV patients treated in several hematologic centers in Croatia and Serbia was retrospectively evaluated. Results: The median SII for the overall cohort was 1960. Higher SII stratified at the specific cut-off points was significantly associated with shorter time to thrombosis (TTT; p = 0.004) driven by arterial thrombotic events, and shorter overall survival (OS; p < 0.001). Higher SII was able to refine the European Leukemia Net-defined high-risk patient subgroup for both thrombotic and survival risks, especially in individuals over 60 years of age. SII and all other evaluated CBC components and indices (leukocytes, ANC, ALC, platelets, neutrophil to lymphocyte ratio (NLR), and platelet to lymphocyte ratio (PLR)) demonstrated low-to-modest prognostic properties, whereas SII outperformed other parameters with respect to TTT and OS prognostications. Discussion: The presented results complement prior studies evaluating the prognostic performance of different CBC components for thrombotic and survival risk predictions and offer more options to personalize PV treatments.
Collapse
Affiliation(s)
- Ivan Krecak
- Department of Internal Medicine, General Hospital of Sibenik-Knin County, 22000 Sibenik, Croatia
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- University of Applied Sciences, 22000 Sibenik, Croatia
| | - Danijela Lekovic
- Clinic of Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Isidora Arsenovic
- Clinic of Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
| | - Andrija Bogdanovic
- Clinic of Hematology, University Clinical Center of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Hrvoje Holik
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, 35000 Slavonski Brod, Croatia
| | - Ivan Zekanovic
- Department of Internal Medicine, General Hospital Zadar, 23000 Zadar, Croatia
| | - Martina Moric Peric
- Department of Internal Medicine, General Hospital Zadar, 23000 Zadar, Croatia
| | - Marko Lucijanic
- Division of Hematology, University Hospital Dubrava, 10040 Zagreb, Croatia
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
- Department of Scientific Research and Translational Medicine, University Hospital Dubrava, 10040 Zagreb, Croatia
| |
Collapse
|
3
|
Gerds AT, Mesa R, Burke JM, Grunwald MR, Stein BL, Squier P, Yu J, Hamer-Maansson JE, Oh ST. Association between elevated white blood cell counts and thrombotic events in polycythemia vera: analysis from REVEAL. Blood 2024; 143:1646-1655. [PMID: 38142448 PMCID: PMC11103079 DOI: 10.1182/blood.2023020232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 12/05/2023] [Accepted: 12/07/2023] [Indexed: 12/26/2023] Open
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.
Collapse
Affiliation(s)
- Aaron T. Gerds
- Cleveland Clinic Taussig Cancer Institute, Cleveland, OH
| | - Ruben Mesa
- UT Health San Antonio MD Anderson Cancer Center, San Antonio, TX
| | | | | | - Brady L. Stein
- Northwestern University Feinberg School of Medicine, Chicago, IL
| | | | | | | | - Stephen T. Oh
- Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
4
|
Manan MR, Kipkorir V, Nawaz I, Waithaka MW, Srichawla BS, Găman AM, Diaconu CC, Găman MA. Acute myocardial infarction in myeloproliferative neoplasms. World J Cardiol 2023; 15:571-581. [PMID: 38058401 PMCID: PMC10696206 DOI: 10.4330/wjc.v15.i11.571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/21/2023] [Accepted: 11/13/2023] [Indexed: 11/23/2023] Open
Abstract
Myeloproliferative neoplasms (MPNs) are a heterogeneous group of hematologic malignancies characterized by an abnormal proliferation of cells of the myeloid lineage. Affected individuals are at increased risk for cardiovascular and thrombotic events. Myocardial infarction (MI) may be one of the earliest clinical manifestations of MPNs or may be a thrombotic complication that develops during the natural course of the disease. In the present review, we examine the epidemiology, pathogenesis, clinical presentation, and management of MI in MPNs based on the available literature. Moreover, we review potential biomarkers that could mediate the MI-MPNs crosstalk, from classical biochemical tests, e.g., lactate dehydrogenase, creatine kinase and troponins, to pro-inflammatory cytokines, oxidative stress markers, and clonal hematopoiesis.
Collapse
Affiliation(s)
| | - Vincent Kipkorir
- Department of Human Anatomy and Physiology, University of Nairobi, Nairobi 00100, Kenya
| | - Iqra Nawaz
- Quaid-e-Azam Medical College, Bahawalpur 63100, Pakistan
| | | | - Bahadar Singh Srichawla
- Department of Neurology, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Amelia Maria Găman
- Department of Pathophysiology, University of Medicine and Pharmacy of Craiova, Craiova 200143, Romania
- Clinic of Hematology, Filantropia City Hospital, Craiova 200143, Romania
| | - Camelia Cristina Diaconu
- Department of Internal Medicine, Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest 050474, Romania
- Internal Medicine Clinic, Clinical Emergency Hospital of Bucharest, Bucharest 105402, Romania
| | - Mihnea-Alexandru Găman
- Faculty of Medicine, "Carol Davila" University of Medicine and Pharmacy, Bucharest 050474, Romania
- Department of Hematology, Center of Hematology and Bone Marrow Transplantation, Fundeni Clinical Institute, Bucharest 022328, Romania
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Romanian Academy, Bucharest 030304, Romania.
| |
Collapse
|
5
|
Krečak I, Zekanović I, Morić Perić M, Holik H, Coha B, Periša V, Lucijanić M. High plateletcrit may be associated with thrombotic risk in polycythemia vera. Int J Lab Hematol 2023; 45:799-801. [PMID: 37050866 DOI: 10.1111/ijlh.14073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 04/06/2023] [Indexed: 04/14/2023]
Affiliation(s)
- Ivan Krečak
- Department of Internal Medicine, General Hospital of Šibenik-Knin County, Šibenik, Croatia
- Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Ivan Zekanović
- Department of Internal Medicine, General Hospital Zadar, Zadar, Croatia
| | | | - Hrvoje Holik
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, Slavonski Brod, Croatia
- School of Medicine, University of Osijek, Osijek, Croatia
| | - Božena Coha
- Department of Internal Medicine, Dr. Josip Benčević General Hospital, Slavonski Brod, Croatia
| | - Vlatka Periša
- School of Medicine, University of Osijek, Osijek, Croatia
- Division of Hematology, University Hospital Osijek, Osijek, Croatia
| | - Marko Lucijanić
- Division of Hematology, University Hospital Dubrava, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
6
|
Gyabaah S, Ahmed EA, Adu-Gyamfi AA, Gyabaah FN, Bonsu AS, Addo AP, Opare Sem OK. Polycythemia vera disease profile in an African population-experience from a tertiary facility in Ghana. SAGE Open Med 2023; 11:20503121231187747. [PMID: 37529706 PMCID: PMC10387680 DOI: 10.1177/20503121231187747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/27/2023] [Indexed: 08/03/2023] Open
Abstract
Objectives The study describes the clinical and laboratory profile of the patients with polycythemia vera at Komfo Anokye Teaching Hospital in Kumasi, Ghana. Methods and design This was a retrospective hospital-based cohort study conducted from September 2020 to August 2022. Hematology clinic entry book was used to identify the patient's unique hospital code. Using these unique codes, retrospective data were collected using an Excel spreadsheet from the Hospital Lightwave health information management system (LHIMS) database. Results A total of 20 participants were recruited over the period of 2 years. The overall mean age was 51.53 ± 16.39 years. The hematological profile of the male participants revealed a mean hemoglobin of 18.25 ± 1.373 g/dl, mean hematocrit of 52 ± 3.47%, and a mean platelet of 345.5 ± 180.82. Comparatively, the mean hemoglobin, hematocrit, and platelet for the female participants were higher with figures of 19.26 ± 1.43 g/dl, 53 ± 3.61%, and 816 ± 935.32, respectively. Headache, tiredness, numbness, splenomegaly, and abnormal labs were the most common reasons why participants sought medical attention. Majority (60%) of the study participants had Janus Kinase 2 mutation. New-onset hypertension was identified in 45% of the study participants during follow-up. Thromboembolism was seen in 10% of the study population. Conclusion Polycythemia vera is an uncommon disease in Ghana mostly found in older males above 50 years. It is important to recognize it early to initiate therapy aimed at preventing common complications such as hypertension and thromboembolism. Polycythemia vera should be considered a differential diagnosis for patients with secondary hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ohene Kwaku Opare Sem
- Komfo Anokye Teaching Hospital, Kumasi, Ghana
- School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
7
|
Iizuka K, Morishita S, Nishizaki Y, Iizuka Y, Iriyama N, Ochiai T, Yanagisawa N, Yasuda H, Ando J, Gotoh A, Takei M, Hatta Y, Nakamura H, Nakayama T, Komatsu N. Von Willebrand factor activity levels are influenced by driver mutation status in polycythemia vera and essential thrombocythemia patients with well-controlled platelet counts. Eur J Haematol Suppl 2022; 109:779-786. [PMID: 36130908 DOI: 10.1111/ejh.13866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 01/19/2023]
Abstract
Von Willebrand factor ristocetin cofactor (VWF activity) and platelet count (PLT) are negatively correlated in patients with polycythemia vera (PV) and essential thrombocythemia (ET). However, VWF activity does not always normalize upon controlling PLT in those patients. To address this issue, we investigated the correlation between VWF activity and PLT in PV and ET patients. The negative correlation between VWF activity and PLT was stronger in calreticulin mutation-positive (CALR+) ET than in Janus kinase 2 mutation-positive (JAK2+) PV or ET groups. When PLT were maintained at a certain level (<600 × 109 /L), low VWF activity (<50%) was more frequently observed in JAK2+ PV patients than in JAK2+ ET (p=0.013) or CALR+ ET (p=0.013) groups, and in PV and ET patients with ≥50% JAK2+ allele burden than in those with allele burden <50% (p=0.015). High VWF activity (>150%) was more frequent in the JAK2+ ET group than in the CALR+ ET group (p=0.005), and often associated with vasomotor symptoms (p=0.002). This study suggests that some patients with JAK2+ PV or ET have VWF activity outside the standard range even with well-controlled PLT, and that the measurement of VWF activity is useful for assessing the risk of thrombosis and hemorrhage.
Collapse
Affiliation(s)
- Kazuhide Iizuka
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan.,Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan.,Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan.,Internal Medicine, Atami Tokoro Memorial Hospital, Shizuoka, Japan
| | - Soji Morishita
- Laboratory for the development of therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yuji Nishizaki
- Medical Technology Innovation Center, Juntendo University, Tokyo, Japan
| | - Yoshikazu Iizuka
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Noriyoshi Iriyama
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tomonori Ochiai
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Hajime Yasuda
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan
| | - Jun Ando
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan.,Department of Cell Therapy and Transfusion Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Akihiko Gotoh
- Department of Hematology, Tokyo Medical University, Tokyo, Japan
| | - Masami Takei
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Yoshihiro Hatta
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Hideki Nakamura
- Division of Hematology and Rheumatology, Department of Medicine, Nihon University School of Medicine, Tokyo, Japan
| | - Tomohiro Nakayama
- Division of Laboratory Medicine, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo, Japan
| | - Norio Komatsu
- Department of Hematology, Juntendo University School of Medicine, Tokyo, Japan.,Laboratory for the development of therapies against MPN, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Department of Advanced Hematology, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Pharmaessentia Japan KK, Tokyo, Japan
| |
Collapse
|
8
|
Rippel N, Tremblay D, Zubizarreta N, Podoltsev N, Gotlib J, Heaney M, Kuykendall A, O'Connell C, Shammo JM, Fleischman A, Kremyanskaya M, Hoffman R, Mesa R, Yacoub A, Mascarenhas J. Anagrelide for platelet-directed cytoreduction in polycythemia vera: Insights into utility and safety outcomes from a large multi-center database. Leuk Res 2022; 119:106903. [PMID: 35717689 DOI: 10.1016/j.leukres.2022.106903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 11/28/2022]
Abstract
Anagrelide (ANA) is a platelet-specific cytoreductive agent utilized in the guideline-directed management of high-risk essential thrombocythemia. In the context of polycythemia vera (PV), ANA is occasionally employed in clinical practice, although data has not consistently demonstrated a benefit to targeting a platelet goal as a therapeutic endpoint. The aim of the current study was to delineate the patterns of ANA use in PV, and to describe outcomes and toxicities. Within a multi-center cohort of 527 patients with PV, 48 received ANA (9 excluded for absent data). 27 (69.2%) had high-risk PV, 10 (25.6%) had prior thrombosis, and none had extreme thrombocytosis, acquired von Willebrand disease, and/or documented resistance to hydroxyurea. While ANA effectively lowered median platelet count, 43.5% of patients had an unresolved thrombocytosis at time of ANA discontinuation. Treatment-emergent adverse events-including headaches, cardiac palpitations and arrhythmias, nausea, vomiting and/or diarrhea-led to ANA discontinuation in 76.9% of patients. Further, three patients experienced arterial thromboses during a median duration of 27.5 months of ANA therapy. In conclusion, this study highlights ANA's restrictive tolerability profile which, compounded by the absence of clear advantage to strict platelet control in PV, suggests the use of ANA should be limited in this setting.
Collapse
Affiliation(s)
- Noa Rippel
- Department of Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicole Zubizarreta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai Tisch Cancer Institute, New York, NY, USA
| | - Nikolai Podoltsev
- Hematology Section, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jason Gotlib
- Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA
| | - Mark Heaney
- Columbia University Medical Center, New York, NY, USA
| | | | - Casey O'Connell
- Keck School of Medicine of University of Southern California, Los Angeles, CA, USA
| | - Jamile M Shammo
- Department of Internal Medicine, Division of Hematology/Oncology, Rush University Medical Center, Chicago, IL, USA
| | - Angela Fleischman
- Irvine Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA, USA
| | - Marina Kremyanskaya
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ruben Mesa
- Department of Hematology and Oncology, Mays MD Anderson Cancer Center at UT Health San Antonio, San Antonio, TX, USA
| | | | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
9
|
Lindgren M, Andréasson B, Samuelsson J, Pettersson H, Enblom-Larsson A, Ravn-Landtblom A, Scheding S, Bentham C, Ahlstrand E. Survival and risk of vascular complications in myelofibrosis-A population-based study from the Swedish MPN group. Eur J Haematol 2022; 109:336-342. [PMID: 35696444 DOI: 10.1111/ejh.13813] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/30/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVE To gain knowledge of underlying risk factors for vascular complications and their impact on life expectancy in myelofibrosis. METHODS From a cohort of 392 myelofibrosis patients registered in the Swedish MPN registry 58 patients with vascular complications during follow-up were identified. Patients with vascular complications were compared with both 1:1 matched controls and the entire myelofibrosis cohort to explore potential risk factors for vascular complications and their impact on survival. RESULTS Incidence of vascular complications was 2.8 events per 100 patient-years and the majority of complications were thrombotic. Patients with complications were significantly older and had lower hemoglobin when compared to the entire cohort. In the case-control analysis, no significant risk factor differences were observed. The major cause of death was vascular complications and median survival was significantly impaired in patients with vascular complications (48 months) compared to controls (92 months). Inferior survival in patients with vascular complications was found to be dependent on IPSS risk category in a Cox regression model. CONCLUSION Vascular complications have a considerable impact on survival in MF. At diagnosis, risk assessment by IPSS does not only predict survival but is also associated with the risk of vascular complications.
Collapse
Affiliation(s)
- Marie Lindgren
- Department of Medicine, Kalmar County Hospital, Kalmar, Sweden.,Faculty of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Björn Andréasson
- Division of Hematology, Department of Medicine, NU Hospital Group, Uddevalla, Sweden
| | - Jan Samuelsson
- Department of Hematology, University Hospital Linköping, Linköping, Sweden
| | - Helna Pettersson
- Division of Hematology, Department of Medicine, NU Hospital Group, Uddevalla, Sweden
| | - Anneli Enblom-Larsson
- Department of Public Health and Clinical Medicine, Luleå Research Unit, Umeå University, Umeå, Sweden
| | - Anna Ravn-Landtblom
- Department of Medicine, Karolinska Institute and Department of Medicine, Division of Hematology, Stockholm South Hospital, Stockholm, Sweden
| | - Stefan Scheding
- Division of Molecular Hematology, Department of Laboratory Medicine, Lund Stem Cell Center, Lund University, Lund, Sweden.,Department of Hematology, Skåne University Hospital, Lund, Sweden
| | | | - Erik Ahlstrand
- Department of Medicine, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| |
Collapse
|
10
|
Clinical insights into the origins of thrombosis in myeloproliferative neoplasms. Blood 2021; 137:1145-1153. [PMID: 33237986 DOI: 10.1182/blood.2020008043] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 11/18/2020] [Indexed: 02/06/2023] Open
Abstract
Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs), polycythemia vera, essential thrombocythemia, and primary myelofibrosis, are hematopoietic stem cell disorders that are defined by activating mutations in signal transduction pathways and are characterized clinically by the overproduction of platelets, red blood cells, and neutrophils, significant burden of disease-specific symptoms, and high rates of vascular events. The focus of this review is to critically reevaluate the clinical burden of thrombosis in MPNs, to review the clinical associations among clonal hematopoiesis, JAK2V617F burden, inflammation, and thrombosis, and to provide insights into novel primary and secondary thrombosis-prevention strategies.
Collapse
|
11
|
Benevolo G, Vassallo F, Urbino I, Giai V. Polycythemia Vera (PV): Update on Emerging Treatment Options. Ther Clin Risk Manag 2021; 17:209-221. [PMID: 33758507 PMCID: PMC7981161 DOI: 10.2147/tcrm.s213020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 02/21/2021] [Indexed: 12/13/2022] Open
Abstract
Polycythemia Vera (PV) is a chronic myeloproliferative neoplasm characterized by exuberant red cell production leading to a broad range of symptoms that compromise quality of life and productivity of patients. PV reduces survival expectation, primarily due to thrombotic events, transformation to blast phase and post-PV myelofibrosis or to development of second cancers, which are associates with poor prognosis. Current therapeutic first line recommendations based on risk adapted classification divided patients into two groups, according to age (< or >60 years) and presence of prior thrombotic events. Low-risk patients (age <60 years and no prior history of thrombosis) should be treated with aspirin (81-100 mg/d) and phlebotomy, to maintain hematocrit <45%. High-risk patients (age >60 years and/or prior history of thrombosis), in addition to aspirin and phlebotomies, should receive cytoreductive therapy in order to reduce thrombotic risk. Nowadays hydroxyurea still remains the cytoreductive agent of first choice, reserving Interferon to young patients or childbearing women. During the last years, ruxolitinib emerged as a new treatment in PV patients, as second line therapy: it appeared especially effective in patients with severe pruritus, symptomatic splenomegaly, or post-PV myelofibrosis symptoms. Currently, in PV treatment, several molecules have been tested or are under investigation. At present, the drug that has shown the most encouraging results is givinostat.
Collapse
Affiliation(s)
- Giulia Benevolo
- Division of Haematology, Città della Salute e della Scienza, Turin, Italy
| | - Francesco Vassallo
- Division of Haematology, Città della Salute e della Scienza, Turin, Italy
| | - Irene Urbino
- Division of Haematology, Città della Salute e della Scienza, Turin, Italy
| | - Valentina Giai
- Division of Haematology, Città della Salute e della Scienza, Turin, Italy
| |
Collapse
|
12
|
Tremblay D, Kosiorek HE, Dueck AC, Hoffman R. Evaluation of Therapeutic Strategies to Reduce the Number of Thrombotic Events in Patients With Polycythemia Vera and Essential Thrombocythemia. Front Oncol 2021; 10:636675. [PMID: 33665170 PMCID: PMC7921696 DOI: 10.3389/fonc.2020.636675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 12/30/2020] [Indexed: 12/13/2022] Open
Abstract
Thrombosis is the largest contributor to morbidity and mortality in patients with polycythemia vera (PV) and essential thrombocythemia (ET). Our understanding of the risk factors and pathophysiology of thrombosis in PV and ET patients is developing, including recent insights into the role of aberrant platelet-neutrophil interactions, JAK2 mutated endothelial cells and the pro-thrombotic inflammatory milieu. To date, few available therapies have demonstrated the ability to reduce the thrombotic burden in patients with these diseases. Although numerous therapeutic agents have been investigated in both PV and ET patients, few studies are designed to assess their impact on thrombotic events. In this review, we first describe the burden of thrombosis in patients with these myeloproliferative neoplasms (MPNs) and briefly explore their pathophysiologic mechanisms. We then critically assess and summarize the evidence behind currently available therapies with attention toward thrombotic endpoints. Finally, we describe a path forward for clinical research in MPNs that involves surrogate endpoint validation, biomarker development, and clinical trial design strategies in order to accurately assess reduction of thrombotic events when evaluating novel therapies.
Collapse
Affiliation(s)
- Douglas Tremblay
- Hematology/Oncology Section, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Heidi E. Kosiorek
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, United States
| | - Amylou C. Dueck
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, United States
| | - Ronald Hoffman
- Hematology/Oncology Section, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
13
|
Falanga A, Marchetti M, Schieppati F. Prevention and Management of Thrombosis in BCR/ABL-Negative Myeloproliferative Neoplasms. Hamostaseologie 2021; 41:48-57. [PMID: 33588455 DOI: 10.1055/a-1334-3259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are clonal disorders of the hematopoietic stem cell. Classical BCR/ABL-negative MPNs include polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF). Thrombotic events are a major cause of morbidity and mortality in these patients. Pathogenesis of blood clotting activation involves various abnormalities of platelets, erythrocytes, and leukocytes, as well as dysfunctions of endothelial cells. Patients with MPN can be stratified in "high risk" or "low risk" of thrombosis according to established risk factors. ET and PV clinical management is highly dependent on the patient's thrombotic risk, and a risk-oriented management strategy to treat these diseases is strongly recommended. In this review, we give an overview of risk factors, pathogenesis, and thrombosis prevention and treatment in MPN.
Collapse
Affiliation(s)
- Anna Falanga
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy.,University of Milano-Bicocca, Department of Medicine and Surgery, Monza, Italy
| | - Marina Marchetti
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| | - Francesca Schieppati
- Department of Immunohematology and Transfusion Medicine, Hospital Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
14
|
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.
Collapse
|
15
|
Lu Y, Zhuoga C, Jin H, Zhu F, Zhao Y, Ding Z, He S, Du A, Xu J, Luo J, Sun Y. Characteristics of acute ischemic stroke in hospitalized patients in Tibet: a retrospective comparative study. BMC Neurol 2020; 20:380. [PMID: 33087079 PMCID: PMC7576739 DOI: 10.1186/s12883-020-01957-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 10/14/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Numerous studies on acute ischemic stroke (AIS) have been conducted at low-altitude regions, and the related findings have been used to guide clinical management. However, corresponding studies at high altitude are few. This study aimed to analyse the clinical characteristics of AIS patients at high-altitude regions through a hospital-based comparative study between Tibet and Beijing. METHODS This study included the diagnoses of AIS patients from People's Hospital of Tibet Autonomous Region (PHOTAR) and Peking University First Hospital (PUFH) between 1 January 2014 and 31 December 2017, where data including patient demographics, treatment time, onset season, risk factors, infarction location, laboratory data, image examination results, treatments, and AIS subtype were collected and compared. Continuous and categorical variables were analysed with a two-sample t-test or Wilcoxon rank sum test and chi-square test, respectively. Significant risk factors were examined with binary logistic regression analysis. RESULTS In total, 236 and 1021 inpatients from PHOTAR and PUFH were included, respectively. The PHOTAR patients were younger than the PUFH patients (P < 0.001). Young adult stroke, erythrocytosis, and hyperhomocysteinemia were more frequent in PHOTAR patients (all P < 0.001). Other vascular risk factors, including hypertension, diabetes mellitus, hyperlipidaemia, smoking and alcohol consumption history, were less prevalent in PHOTAR patients than in PUFH patients. The rate of intravenous thrombolysis and the rate of within intravenous thrombolysis window time were also lower in PHOTAR patients (both P < 0.001). The PHOTAR group also tended to have anterior circulation infarction. Erythrocytosis and hyperhomocysteinemia were independent risk factors in PHOTAR, and young adults accounted for a larger proportion of stroke cases. CONCLUSION In Tibet, AIS patients were relatively younger, and anterior circulation infarctions were more common. Erythrocytosis and hyperhomocysteinemia may contribute to these differences. Here, young adult stroke also accounted for a higher proportion, and this may be associated with erythrocytosis. Our findings present the first hospital-based comparative study in Tibet and may contribute to policies for stroke prevention in this region.
Collapse
Affiliation(s)
- Yuxuan Lu
- Department of Neurology, Peking University First Hospital, No. 8, Xishiku St, Xicheng District, Beijing, 100034, China
| | - Cidan Zhuoga
- Department of Neurology, People's Hospital of Tibet Autonomous Region, Lhasa, 850000, Tibet, China
| | - Haiqiang Jin
- Department of Neurology, Peking University First Hospital, No. 8, Xishiku St, Xicheng District, Beijing, 100034, China
| | - Feiqi Zhu
- Cognitive Impairment Ward of Neurology Department, The Third Affiliated Hospital of Shenzhen University Medical College, Shenzhen, 518005, China
| | - Yuhua Zhao
- Department of Neurology, People's Hospital of Tibet Autonomous Region, Lhasa, 850000, Tibet, China
| | - Zhijie Ding
- Department of Neurology, People's Hospital of Tibet Autonomous Region, Lhasa, 850000, Tibet, China
| | - Shihua He
- Department of Neurology, People's Hospital of Tibet Autonomous Region, Lhasa, 850000, Tibet, China
| | - Ailian Du
- Department of Neurology, Tongren Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200050, China
| | - Jun Xu
- Department of Cognitive Neurology, China National Clinical Research Center for Neurological Diseases (NCRC-ND), Beijing Tiantan Hospital, Capital Medical University, Beijing, 100070, China
| | - Jingjing Luo
- Department of Neurology, Peking University First Hospital, No. 8, Xishiku St, Xicheng District, Beijing, 100034, China.
| | - Yongan Sun
- Department of Neurology, Peking University First Hospital, No. 8, Xishiku St, Xicheng District, Beijing, 100034, China.
| |
Collapse
|
16
|
Current management strategies for polycythemia vera and essential thrombocythemia. Blood Rev 2020; 42:100714. [DOI: 10.1016/j.blre.2020.100714] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/02/2019] [Accepted: 05/11/2020] [Indexed: 02/06/2023]
|
17
|
Warny M, Helby J, Birgens HS, Bojesen SE, Nordestgaard BG. Arterial and venous thrombosis by high platelet count and high hematocrit: 108 521 individuals from the Copenhagen General Population Study. J Thromb Haemost 2019; 17:1898-1911. [PMID: 31309714 DOI: 10.1111/jth.14574] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 07/11/2019] [Indexed: 08/31/2023]
Abstract
BACKGROUND It is unclear whether high platelet count or high hematocrit predict risk of thrombosis in individuals from the general population. OBJECTIVES We tested the hypothesis that individuals from the general population with high platelet count or high hematocrit have high risk of arterial and venous thrombosis. METHODS We prospectively followed 108 521 individuals from The Copenhagen General Population Study for a median of 8 years. Platelet count and blood hematocrit were measured at study entry. RESULTS AND CONCLUSION Multivariable adjusted hazard ratios for individuals with platelet counts in the top 5 percentiles (>398 × 109 /L) vs in the 25th-75th percentiles (231-316 × 109 /L) were 1.77 (95% confidence interval [CI], 1.38-2.24) for arterial thrombosis in the brain (38 and 26 events/10 000 person-years) and 0.82 (95%, 0.61-1.11) for arterial thrombosis in the heart (23 and 28 events/10 000 person-years). For individuals with hematocrit values in the top 5 percentiles (women/men: >45/>48%) vs the 25th-75th percentiles (women/men: 38.1-42/41.1-45%), hazard ratios were 1.27 (95% CI, 0.91-1.75) for arterial thrombosis in the brain (40 and 26 events/10 000 person-years) and 1.46 (95% CI, 1.06-2.00) for arterial thrombosis in the heart (43 and 25 events/10 000 person-years). Neither high platelet count nor high hematocrit was associated with risk of venous thromboembolism. When excluding individuals with myeloproliferative neoplasia from the main analyses, results on risk of thrombosis were similar. In this prospective study, high platelet counts were associated with 1.8-fold risk of arterial thrombosis in the brain, whereas high hematocrit was associated with 1.5-fold risk of arterial thrombosis in the heart.
Collapse
Affiliation(s)
- Marie Warny
- Department of Hematology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Jens Helby
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Department of Internal Medicine, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Henrik S Birgens
- Department of Hematology, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stig E Bojesen
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| | - Børge G Nordestgaard
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
- Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
18
|
Dewarrat N, Kaiser J, Baud D, Alberio L, Gavillet M. Pregnancy with Chuvash Polycythaemia and Other Congenital Erythrocytosis. Acta Haematol 2019; 143:69-72. [PMID: 31167179 DOI: 10.1159/000499861] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 03/22/2019] [Indexed: 11/19/2022]
Abstract
This original report describes the management of a pregnant woman with congenital erythrocytosis (Chuvash polycythaemia) and reviews the scarce data available in the literature. Therapy consisted of low-dose aspirin and phlebotomies to maintain haematocrit <50% while monitoring iron stores to avoid severe deficiency detrimental to the foetus. Despite normal initial foetal growth, the pregnancy was complicated by preterm birth due to chorioamnionitis. The placenta showed no signs of thrombotic events. The published reports cover 13 pregnancies in 8 patients, showing 1 first-trimester miscarriage, 5 infants with intrauterine growth restriction and/or preterm birth and 1 maternal thrombotic event. These cases were managed with phlebotomies, low-dose aspirin and/or low-molecular-weight heparin, although inconsistently.
Collapse
Affiliation(s)
- Natacha Dewarrat
- Service and Central Laboratory of Haematology, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Julie Kaiser
- Service and Central Laboratory of Haematology, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Haematology Department, Institute Central des Hôpitaux (ICH), Sion, Switzerland
| | - David Baud
- Materno-Foetal and Obstetrics Research Unit, Department "Femme-Mère-Enfant," University Hospital, Lausanne, Switzerland
| | - Lorenzo Alberio
- Service and Central Laboratory of Haematology, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Mathilde Gavillet
- Service and Central Laboratory of Haematology, Department of Oncology, University Hospital of Lausanne (CHUV), Lausanne, Switzerland,
| |
Collapse
|
19
|
Mochizuki T, Tanabe K, Saito R, Ota H, Yamamoto Y, Saeki Y, Ohdan H. Perioperative management of polycythemia vera with advanced gastric cancer: A case report. Int J Surg Case Rep 2019; 58:224-227. [PMID: 31112910 PMCID: PMC6527945 DOI: 10.1016/j.ijscr.2019.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/27/2019] [Accepted: 04/08/2019] [Indexed: 11/11/2022] Open
Abstract
We report a case involving operation for gastric cancer after treatment for PV. Control of WBC and Plt counts during the perioperative period led to good results. Perioperative management for PV is important for complication-free surgery. Careful follow up should be performed for gastric cancer and PV recurrence.
Introduction Polycythemia vera (PV) is a chronic myeloproliferative disorder with generally good prognosis. However, thrombotic and cardiovascular complications are among the leading causes of death in patients with PV and sufficiently effective management strategies are yet to be established. We report a case involving operation for gastric cancer in a patient being treated for PV. Presentation of case A 73-year-old man was diagnosed as PV on the basis of abnormal laboratory data eight years previously. Three months ago, he was referred to our department for anorexia and melena. The preoperative diagnosis was advanced gastric cancer, Stage IIB. To avoid perioperative thrombotic and hemorrhagic events, low-dose aspirin administration was continued with an additional dose of hydroxyurea. Emergent control was necessary because of the severity of gastric cancer symptoms, including pyloric stenosis and anemia. Distal gastrectomy with D2 lymph node resection was performed. The postoperative course was satisfactory, and the patient is currently doing well without any signs of recurrence or hematological complications. Discussion We described the successful management of a patient being treated for PV who underwent gastrectomy. As mentioned earlier, thrombotic complications and hemorrhage during the perioperative period are the major risk factors in patients with PV. In this case, control of white blood cell and platelet counts during the perioperative period led to good results. Conclusions Perioperative management for PV is important for complication-free surgery. Careful follow up should be performed for gastric cancer and PV recurrence.
Collapse
Affiliation(s)
- Tetsuya Mochizuki
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biochemical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Kazuaki Tanabe
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biochemical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan.
| | - Ryusuke Saito
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biochemical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hiroshi Ota
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biochemical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yuji Yamamoto
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biochemical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Yoshihiro Saeki
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biochemical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biochemical & Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| |
Collapse
|
20
|
Tavares RS, Nonino A, Pagnano KBB, Nascimento ACKVD, Conchon M, Fogliatto LM, Funke VAM, Bendit I, Clementino NCD, Chauffaille MDLLF, Bernardo WM, Santos FPDS. Guideline on myeloproliferative neoplasms: Associacão Brasileira de Hematologia, Hemoterapia e Terapia Cellular: Project guidelines: Associação Médica Brasileira - 2019. Hematol Transfus Cell Ther 2019; 41 Suppl 1:1-73. [PMID: 31248788 PMCID: PMC6630088 DOI: 10.1016/j.htct.2019.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 03/20/2019] [Indexed: 12/22/2022] Open
Affiliation(s)
| | - Alexandre Nonino
- Instituto Hospital de Base do Distrito Federal (IHBDF), Brasília, DF, Brazil
| | | | | | | | | | | | - Israel Bendit
- Hospital Das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil
| | | | | | - Wanderley Marques Bernardo
- Hospital Das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, SP, Brazil; Associação Médica Brasileira (AMB), São Paulo, SP, Brazil
| | | |
Collapse
|
21
|
Gordeuk VR, Key NS, Prchal JT. Re-evaluation of hematocrit as a determinant of thrombotic risk in erythrocytosis. Haematologica 2019; 104:653-658. [PMID: 30872370 PMCID: PMC6442963 DOI: 10.3324/haematol.2018.210732] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 01/28/2019] [Indexed: 12/20/2022] Open
Abstract
Here we critically evaluate the role of elevated hematocrit as the principal determinant of thrombotic risk in polycythemia and erythrocytosis, defined by an expansion of red cell mass. Since red cell volume determination is no longer readily available, in clinical practice, polycythemia and erythrocytosis are defined by elevated hemoglobin and hematocrit. Thrombosis is common in Chuvash erythrocytosis and polycythemia vera. Although the increased thrombotic risk is assumed to be due to the elevated hematocrit and an associated increase in blood viscosity, thrombosis does not accompany most types of erythrocytosis. We review studies indicating that the occurrence of thrombosis in Chuvash erythrocytosis is independent of hematocrit, that the thrombotic risk is paradoxically increased by phlebotomy in Chuvash erythrocytosis, and that, when compared to chemotherapy, phlebotomy is associated with increased thrombotic risk in polycythemia vera. Inherited and environmental causes that lead to polycythemia and erythrocytosis are accompanied by diverse cellular changes that could directly affect thrombotic risk, irrespective of the elevated hematocrit. The pressing issue in these disorders is to define factors other than elevated hematocrit that determine thrombotic risk. Defining these predisposing factors in polycythemia and erythrocytosis should then lead to rational therapies and facilitate development of targeted interventions.
Collapse
Affiliation(s)
- Victor R Gordeuk
- Division of Hematology and Oncology, University of Illinois at Chicago, IL
| | - Nigel S Key
- Division of Hematology-Oncology and UNC Hemophilia and Thrombosis Center, UNC, Chapel Hill, NC
| | - Josef T Prchal
- Division of Hematology and Hematologic Malignancies, University of Utah and Huntsman Cancer Center, Salt Lake City, UT, USA
| |
Collapse
|
22
|
McMullin MFF, Mead AJ, Ali S, Cargo C, Chen F, Ewing J, Garg M, Godfrey A, Knapper S, McLornan DP, Nangalia J, Sekhar M, Wadelin F, Harrison CN. A guideline for the management of specific situations in polycythaemia vera and secondary erythrocytosis: A British Society for Haematology Guideline. Br J Haematol 2019; 184:161-175. [PMID: 30426472 PMCID: PMC6519221 DOI: 10.1111/bjh.15647] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
| | - Adam J. Mead
- MRC Molecular Haematology UnitMRC Weatherall Institute of Molecular MedicineRadcliffe Department of MedicineUniversity of OxfordOxfordUK
| | - Sahra Ali
- Castle Hill HospitalHull and East Yorkshire Hospitals NHS TrustHullUK
| | | | - Frederick Chen
- The Royal London HospitalBart's Health NHS TrustLondonUK
| | - Joanne Ewing
- Birmingham Heart of England NHS Foundation TrustBirminghamUK
| | - Mamta Garg
- University Hospital of Leicester NHS TrustLeicester (BSH representative)UK
| | - Anna Godfrey
- Department of Haematology and Haematopathology and Oncology Diagnostic ServiceCambridge University Hospitals NHS Foundation TrustCambridgeUK
| | | | | | | | - Mallika Sekhar
- Royal Free London NHS Foundation TrustUniversity College London HospitalLondonUK
| | | | | | | |
Collapse
|
23
|
McMullin MF, Harrison CN, Ali S, Cargo C, Chen F, Ewing J, Garg M, Godfrey A, S SK, McLornan DP, Nangalia J, Sekhar M, Wadelin F, Mead AJ. A guideline for the diagnosis and management of polycythaemia vera. A British Society for Haematology Guideline. Br J Haematol 2019; 184:176-191. [PMID: 30478826 DOI: 10.1111/bjh.15648] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
| | | | - Sahra Ali
- Castle Hill Hospital, Hull and East Yorkshire Hospitals NHS Trust, Hull, UK
| | | | - Frederick Chen
- The Royal London Hospital, Bart's Health NHS Trust, London, UK
| | - Joanne Ewing
- Birmingham Heart of England NHS Foundation Trust, Birmingham, UK
| | - Mamta Garg
- University Hospital of Leicester NHS Trust, Leicester (BSH representative), UK
| | - Anna Godfrey
- Department of Haematology and Haematopathology and Oncology Diagnostic Service, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | | | | | | | | | - Adam J Mead
- MRC Weatherall, Institute of Molecular Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
24
|
|
25
|
Evidence- and consensus-based recommendations for phlebotomy in polycythemia vera. Leukemia 2018; 32:2077-2081. [PMID: 29955128 DOI: 10.1038/s41375-018-0199-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 04/13/2018] [Accepted: 04/25/2018] [Indexed: 01/11/2023]
|
26
|
Kamiunten A, Shide K, Kameda T, Sekine M, Kubuki Y, Ito M, Toyama T, Kawano N, Marutsuka K, Maeda K, Takeuchi M, Kawano H, Sato S, Ishizaki J, Akizuki K, Tahira Y, Shimoda H, Hidaka T, Yamashita K, Matsuoka H, Shimoda K. Thrombohemorrhagic events, disease progression, and survival in polycythemia vera and essential thrombocythemia: a retrospective survey in Miyazaki prefecture, Japan. Int J Hematol 2018; 107:681-688. [DOI: 10.1007/s12185-018-2428-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/21/2018] [Accepted: 02/21/2018] [Indexed: 12/16/2022]
|
27
|
Thrombosis in Philadelphia negative classical myeloproliferative neoplasms: a narrative review on epidemiology, risk assessment, and pathophysiologic mechanisms. J Thromb Thrombolysis 2018; 45:516-528. [DOI: 10.1007/s11239-018-1623-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
28
|
Mangaonkar AA, Hoversten KP, Gangat N. Prognostic risk model for patients with high-risk polycythemia vera and essential thrombocythemia. Expert Rev Hematol 2018; 11:247-252. [PMID: 29313725 DOI: 10.1080/17474086.2018.1426455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Polycythemia Vera (PV) and Essential Thrombocythemia (ET) are the most frequent Philadelphia chromosome-negative myeloproliferative neoplasms, the other entity being myelofibrosis. Management of patients with PV and ET is fraught with difficulties as they have an inherent tendency to cause thrombotic and hemorrhagic events. There are no curative treatment options, therefore it is important that a risk-adapted treatment approach is applied. Areas covered: This review discusses existing literature about prognosis in PV and ET, and addresses critical aspects related to defining 'high-risk' disease. In addition to the traditional risk factors such as age and prior thrombotic history, we discuss the prognostic impact of additional parameters such as cardiovascular risk factors, white blood cell count, karyotype and gene mutations. Expert commentary: We use age>60 years, presence of JAK2 mutation and a prior thrombotic history as the principle determinants of 'high-risk' for thrombosis in PV and ET, dividing the patients into very-low, low, intermediate and high-risk disease. Typically, low-risk patients are treated either with observation or aspirin alone. High-risk patients require cytoreductive therapies, along with aspirin and/or systemic anticoagulation. Intermediate-risk patients are treated on a case-by-case basis. Further, we aim to maintain a hematocrit <45% with aggressive phlebotomy in patients with PV.
Collapse
Affiliation(s)
| | | | - Naseema Gangat
- a Department of Hematology , Mayo Clinic , Rochester , MN , USA
| |
Collapse
|
29
|
|
30
|
Tefferi A, Vannucchi AM, Barbui T. Polycythemia vera treatment algorithm 2018. Blood Cancer J 2018; 8:3. [PMID: 29321547 PMCID: PMC5802495 DOI: 10.1038/s41408-017-0042-7] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 07/27/2017] [Accepted: 07/27/2017] [Indexed: 12/18/2022] Open
Abstract
Recently reported mature survival data have confirmed the favorable prognosis in polycythemia vera (PV), with an estimated median survival of 24 years, in patients younger than age 60 years old. Currently available drugs for PV have not been shown to prolong survival or alter the natural history of the disease and are instead indicated primarily for prevention of thrombosis. Unfortunately, study endpoints that are being utilized in currently ongoing clinical trials in PV do not necessarily target clinically or biologically relevant outcomes, such as thrombosis, survival, or morphologic remission, and are instead focused on components of disease palliation. Even more discouraging has been the lack of critical appraisal from "opinion leaders", on the added value of newly approved drugs. Keeping these issues in mind, at present, we continue to advocate conservative management in low-risk PV (phlebotomy combined with once- or twice-daily aspirin therapy) and include cytoreductive therapy in "high-risk" patients; in the latter regard, our first, second, and third line drugs of choice are hydroxyurea, pegylated interferon-α and busulfan, respectively. In addition, it is reasonable to consider JAK2 inhibitor therapy, in the presence of protracted pruritus or markedly enlarged splenomegaly shown to be refractory to the aforementioned drugs.
Collapse
Affiliation(s)
- Ayalew Tefferi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Alessandro M Vannucchi
- Department of Experimental and Clinical Medicine, CRIMM, Center Research and Innovation of Myeloproliferative Neoplasms, Azienda Ospedaliera Universitaria Careggi, University of Florence, Florence, Italy
| | - Tiziano Barbui
- Research Foundation, Papa Giovanni XXIII Hospital, Bergamo, Italy
| |
Collapse
|
31
|
Gao C, Yang X, Li J, Wang W, Hou J, Li H, Tan X, Shi J, Fu Y, Zhou J. Role of erythrocytes and platelets in the hypercoagulable status in polycythemia vera through phosphatidylserine exposure and microparticle generation. Thromb Haemost 2017; 109:1025-32. [DOI: 10.1160/th12-11-0811] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2012] [Accepted: 02/24/2013] [Indexed: 02/02/2023]
Abstract
SummaryThe development of thrombosis in polycythaemia vera (PV) involves multifactorial processes including pathological activation of blood cells. Release of microparticles (MPs) by activated cells in diseases is associated with thrombotic risk, but relatively few data are available in PV. The aim of the present study was to investigate the increase in MP release and exposure of phosphatidylserine (PS) on the outer membrane of MP-origin cells in patients with PV, and to analyse their procoagulant activity (PCA). PS-positive MPs and cells were detected by flow cytometry, while PCA was assessed with clotting time and purified coagulation complex assays. We found that PV patients had elevated circulating lactadherin+ MPs, which mostly originating from erythrocytes, platelets, granulocytes, and endothelial cells, as well as increased PS exposing erythrocytes/platelets as compared to secondary polycythaemia patients or healthy controls. These PS-bearing MPs and cells were highly procoagulant. Moreover, lactadherin competed factor V and VIII to PS and inhibited about 90% of the detected PCA in a dose-response manner while anti-TF antibody did no significant inhibition. Treatment with hydroxyurea is associated with a decrease in PS exposure and lactadherin+ MP release of erythrocytes/platelets. Our data demonstrate that PV patients are characterised by increased circulating procoagulant MPs and PS exposing erythrocytes/platelets, which could contribute to the hypercoagulable state in these patients.
Collapse
|
32
|
Abstract
Red blood cells (RBCs) have historically been considered passive bystanders in thrombosis. However, clinical and epidemiological studies have associated quantitative and qualitative abnormalities in RBCs, including altered hematocrit, sickle cell disease, thalassemia, hemolytic anemias, and malaria, with both arterial and venous thrombosis. A growing body of mechanistic studies suggests that RBCs can promote thrombus formation and enhance thrombus stability. These findings suggest that RBCs may contribute to thrombosis pathophysiology and reveal potential strategies for therapeutically targeting RBCs to reduce thrombosis.
Collapse
|
33
|
Analysis of thrombosis and bleeding complications in patients with polycythemia vera: a Turkish retrospective study. Int J Hematol 2016; 105:70-78. [DOI: 10.1007/s12185-016-2105-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 09/26/2016] [Accepted: 09/26/2016] [Indexed: 12/20/2022]
|
34
|
Rajnics P, Kellner Á, Karádi É, Moizs M, Bödör C, Király P, Marosvári D, Andrikovics H, Egyed M. Increased Lipocalin 2 level may have important role in thrombotic events in patients with polycythemia vera and essential thrombocythemia. Leuk Res 2016; 48:101-6. [DOI: 10.1016/j.leukres.2016.04.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 03/25/2016] [Accepted: 04/27/2016] [Indexed: 01/08/2023]
|
35
|
Vaddi K, Verstovsek S, Kiladjian JJ. Ruxolitinib: a targeted treatment option for patients with polycythemia vera. BLOOD AND LYMPHATIC CANCER-TARGETS AND THERAPY 2016; 6:7-19. [PMID: 31360077 PMCID: PMC6467337 DOI: 10.2147/blctt.s101185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
Polycythemia vera (PV) is a chronic myeloproliferative neoplasm characterized by erythrocytosis and the presence of Janus kinase (JAK) 2V617F or similar mutations. This review summarizes the pathophysiology of PV, the challenges associated with traditional treatment options, and the scientific rationale and supportive clinical evidence for targeted therapy with ruxolitinib. Accumulating evidence indicates that activating mutations in JAK2 drive the PV disease state. Traditional PV treatment strategies, including aspirin, phlebotomy, and cytoreductive agents such as hydroxyurea, provide clinical benefits for some but not all patients and may not adequately treat PV-related symptoms. Furthermore, traditional treatment approaches are associated with potential side effects that may limit their usage and lead some patients to discontinue the treatment. Ruxolitinib is an orally available small-molecule tyrosine kinase inhibitor that is a potent and selective inhibitor of JAK1/JAK2. Ruxolitinib is approved in the US for patients with PV with an inadequate response or intolerance to hydroxyurea and in Europe for adults with PV who are resistant to or intolerant of hydroxyurea. In the Phase III RESPONSE registration trial, ruxolitinib was superior to the best available therapy in patients with PV who were resistant to or intolerant of hydroxyurea in controlling hematocrit levels, reducing spleen volume, and improving PV-related symptoms and quality-of-life measures. The most common nonhematologic adverse events in ruxolitinib-treated patients were headache, diarrhea, pruritus, and fatigue in the RESPONSE trial; hematologic adverse events were primarily grade 1 or 2. In the Phase IIIb nonregistration RELIEF trial, there were nonsignificant trends toward an improved symptom control in patients with PV on a stable hydroxyurea dose who were generally well controlled but reported disease-associated symptoms and switched to ruxolitinib vs those who continued hydroxyurea therapy. Updated treatment guidelines will be important for educating physicians about the role of ruxolitinib in the treatment of patients with PV.
Collapse
Affiliation(s)
- Kris Vaddi
- Drug Discovery, Incyte Corporation, Wilmington, DE,
| | - Srdan Verstovsek
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jean-Jacques Kiladjian
- Clinical Investigations Center, Hôpital Saint-Louis et Université Paris Diderot, Paris, France
| |
Collapse
|
36
|
Choi CW, Bang SM, Jang S, Jung CW, Kim HJ, Kim HY, Kim SJ, Kim YK, Park J, Won JH. Guidelines for the management of myeloproliferative neoplasms. Korean J Intern Med 2015; 30:771-88. [PMID: 26552452 PMCID: PMC4642006 DOI: 10.3904/kjim.2015.30.6.771] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/30/2015] [Indexed: 01/04/2023] Open
Abstract
Polycythemia vera, essential thrombocythemia, and primary myelofibrosis are collectively known as 'Philadelphia-negative classical myeloproliferative neoplasms (MPNs).' The discovery of new genetic aberrations such as Janus kinase 2 (JAK2) have enhanced our understanding of the pathophysiology of MPNs. Currently, the JAK2 mutation is not only a standard criterion for diagnosis but is also a new target for drug development. The JAK1/2 inhibitor, ruxolitinib, was the first JAK inhibitor approved for patients with intermediate- to high-risk myelofibrosis and its effects in improving symptoms and survival benefits were demonstrated by randomized controlled trials. In 2011, the Korean Society of Hematology MPN Working Party devised diagnostic and therapeutic guidelines for Korean MPN patients. Subsequently, other genetic mutations have been discovered and many kinds of new drugs are now under clinical investigation. In view of recent developments, we have revised the guidelines for the diagnosis and management of MPN based on published evidence and the experiences of the expert panel. Here we describe the epidemiology, new genetic mutations, and novel therapeutic options as well as diagnostic criteria and standard treatment strategies for MPN patients in Korea.
Collapse
Affiliation(s)
- Chul Won Choi
- Division of Oncology-Hematology, Department of Internal Medicine, Korea University Guro Hospital, Seoul, Korea
| | - Soo-Mee Bang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Seongsoo Jang
- Department of Laboratory Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Chul Won Jung
- Division of Hematology/Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee-Jin Kim
- Department of Laboratory Medicine & Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Ho Young Kim
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang, Korea
| | - Soo-Jeong Kim
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Yeo-Kyeoung Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Hwasun Hospital, Hwasun, Korea
| | - Jinny Park
- Division of Hematology-Oncology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Korea
| | - Jong-Ho Won
- Division of Hematology-Oncology, Department of Internal Medicine, Soonchunhyang University College of Medicine, Seoul, Korea
| |
Collapse
|
37
|
Stein BL, Oh ST, Berenzon D, Hobbs GS, Kremyanskaya M, Rampal RK, Abboud CN, Adler K, Heaney ML, Jabbour EJ, Komrokji RS, Moliterno AR, Ritchie EK, Rice L, Mascarenhas J, Hoffman R. Polycythemia Vera: An Appraisal of the Biology and Management 10 Years After the Discovery of JAK2 V617F. J Clin Oncol 2015; 33:3953-60. [PMID: 26324368 DOI: 10.1200/jco.2015.61.6474] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Polycythemia vera (PV) is a chronic myeloproliferative neoplasm that is associated with a substantial symptom burden, thrombohemorrhagic complications, and impaired survival. A decade after the seminal discovery of an activating mutation in the tyrosine kinase JAK2 in nearly all patients with PV, new treatment options are finally beginning to emerge, necessitating a critical reappraisal of the underlying pathogenesis and therapeutic modalities available for PV. Herein, we comprehensively review clinical aspects of PV including diagnostic considerations, natural history, and risk factors for thrombosis. We summarize recent studies delineating the genetic basis of PV, including their implications for evolution to myelofibrosis and secondary acute myeloid leukemia. We assess the quality of evidence to support the use of currently available therapies, including aspirin, phlebotomy, hydroxyurea, and interferon. We analyze recent studies evaluating the safety and efficacy of JAK inhibitors, such as ruxolitinib, and evaluate their role in the context of other available therapies for PV. This review provides a framework for practicing hematologists and oncologists to make rational treatment decisions for patients with PV.
Collapse
Affiliation(s)
- Brady L Stein
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Stephen T Oh
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Dmitriy Berenzon
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Gabriela S Hobbs
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Marina Kremyanskaya
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Raajit K Rampal
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Camille N Abboud
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Kenneth Adler
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Mark L Heaney
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Elias J Jabbour
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rami S Komrokji
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Alison R Moliterno
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ellen K Ritchie
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Lawrence Rice
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - John Mascarenhas
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ronald Hoffman
- Brady L. Stein, Northwestern Feinberg University School of Medicine, Chicago, IL; Stephen T. Oh and Camille Abboud, Washington University School of Medicine, St Louis, MO; Dmitriy Berenzon, Wake Forest University School of Medicine, Winston Salem, NC; Gabriela S. Hobbs, Massachusetts General Hospital, Boston, MA; Marina Kremyanskaya, John Mascarenhas, and Ronald Hoffman, Mount Sinai School of Medicine; Raajit K. Rampal, Memorial Sloan Kettering Cancer Center; Mark L. Heaney, Columbia University Medical Center; Ellen K. Ritchie, Cornell University School of Medicine, New York, NY; Kenneth Adler, Regional Cancer Care Associates, Morristown, NJ; Elias J. Jabbour, MD Anderson Cancer Center; Lawrence Rice, Cornell Houston Methodist Hospital, Houston, TX; Rami S. Komrokji, Moffitt Cancer Center, Tampa, FL; and Alison R. Moliterno, Johns Hopkins University School of Medicine, Baltimore, MD.
| |
Collapse
|
38
|
Harrison CN, McLornan DP, Francis YA, Woodley C, Provis L, Radia DH. How We Treat Myeloproliferative Neoplasms. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2015; 15 Suppl:S19-S26. [PMID: 26297273 DOI: 10.1016/j.clml.2015.02.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 02/06/2015] [Accepted: 02/26/2015] [Indexed: 06/04/2023]
Abstract
The present report focuses on management strategies for the myeloproliferative neoplasm according to the structure and processes we use within our center, a large tertiary unit in central London. The standard procedures for achieving an accurate diagnosis and risk stratification and therapeutic strategies for these diseases with a detailed focus on contentious areas are discussed. In the 9 years after the description of the Janus kinase 2 mutation, this field has altered quite radically in several aspects. For example, a new therapeutic paradigm exists, especially for myelofibrosis. We share how our unit has adapted to these changes.
Collapse
Affiliation(s)
- Claire N Harrison
- Department of Haematology, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom.
| | - Donal P McLornan
- Department of Haematology, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom
| | - Yvonne A Francis
- Department of Haematology, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom
| | - Claire Woodley
- Department of Haematology, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom
| | - Lizzie Provis
- Pharmacy Service, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom
| | - Deepti H Radia
- Department of Haematology, Guy's and St. Thomas' National Health Service Foundation Trust, London, United Kingdom
| |
Collapse
|
39
|
Piccin A, Steurer M, Mitterer M, Blöchl EM, Marcheselli L, Pusceddu I, Marabese A, Bertozzi I, Corvetta D, Randi ML, Elli E, Pogliani EM, Veneri D, Perbellini O, Krampera M, Pacquola E, Gottardi M, Tiribelli M, Guella A, Innella B, Vivaldi P, De Biasi E, Sancetta R, Rocconi R, Bassan R, Gherlinzoni F, Pizzolo G, Gastl G, Cortelazzo S. Role of blood cells dynamism on hemostatic complications in low-risk patients with essential thrombocythemia. Intern Emerg Med 2015; 10:451-60. [PMID: 25585678 DOI: 10.1007/s11739-015-1186-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2014] [Accepted: 12/30/2014] [Indexed: 10/24/2022]
Abstract
Patients with essential thrombocythemia (ET) aged less than 60 years, who have not suffered a previous vascular event (low-risk patients), may develop thrombotic or hemorrhagic events. So far, it has not been possible to identify useful markers capable of predicting which of these patients are more likely to develop an event and therefore who needs to be treated. In the present study, we analysed the relationship between vascular complications and longitudinal blood counts of 136 low-risk ET patients taken over a sustained period of time (blood cells dynamism). After a median follow-up of 60 months, 45 out of 136 patients (33%) suffered 40 major thrombotic and 5 severe hemorrhagic complications. A total number of 5,781 blood counts were collected longitudinally. Thrombotic and hemorrhagic events were studied together (primary endpoint) but also separately (thrombotic alone = secondary endpoint; hemorrhagic alone = tertiary endpoint). The primary endpoint showed no significant association between platelet and WBC count at diagnosis and risk of any event (platelet, p = 0.797; WBC, p = 0.178), while Hb at baseline did show an association (p = 0.024). In the dynamic analysis with Cox regression model, where the blood count values were studied by time of follow-up, we observed that the risk for Hb was 1.49 (95% CI 1.13-1.97) for every increase of 1 g/dL, and that this risk then marginally decreased during follow-up. WBC was associated with an increased risk at baseline for every increase of 1 × 10(9)/L (hazard ratio (HR) 1.07, 95% CI 1.01-1.13, p = 0.034), the risk was stable during follow-up (HR 0.95, p = 0.187 at 60 months). Also, for each increment at baseline of 100 × 10(9) platelets/L, HR was increased by 1.08 (95% CI 0.97-1.22, p = 0.159) and decreases during follow-up. In conclusion, this study is the first to evaluate in ET low-risk patients, the risk of developing a thrombotic/hemorrhagic event considering blood counts over time. Overall our study shows that the risk changes over time. For example, the risk associated with WCC is not linear as previously reported. An interesting new finding is that PLT and even Hb contribute to the risk of developing vascular events. Future treatments should take into consideration these findings and aim to control all parameters over time. We believe this early study may help develop a dynamic analysis model to predict thrombosis in the single patient. Further studies are now warranted to further validate our findings.
Collapse
Affiliation(s)
- Andrea Piccin
- Department of Haematology, San Maurizio Regional Hospital, Bolzano, South Tyrol, Italy,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Mesa RA, Scherber RM, Geyer HL. Reducing symptom burden in patients with myeloproliferative neoplasms in the era of Janus kinase inhibitors. Leuk Lymphoma 2015; 56:1989-99. [PMID: 25644746 DOI: 10.3109/10428194.2014.983098] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Philadelphia chromosome-negative myeloproliferative neoplasms (MPNs) are clonal stem cell-derived malignancies that include primary myelofibrosis, polycythemia vera and essential thrombocythemia and are characterized by dysregulated Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling. Clinical manifestations include splenomegaly, cytopenias and/or systemic inflammation. Patients have a heterogeneous symptom profile that includes fatigue, loss of appetite, pruritus and night sweats, which significantly impact quality of life (QoL) and lead to poor survival outcomes. With the introduction of JAK inhibitors, improvement in disease-related symptoms has emerged as a realistic expectation of therapy and an integral measure of clinical efficacy. The JAK1/JAK2 inhibitor ruxolitinib is approved for the treatment of myelofibrosis and is currently under clinical development for polycythemia vera. Ruxolitinib has demonstrated significant reductions in symptom burden, with consequent improvements in QoL measures. With the potential to improve QoL, recognition of the impact and burden of symptoms on patients with MPNs is critical.
Collapse
|
41
|
Connelly-Smith LS, Linenberger ML. Therapeutic Apheresis for Patients with Cancer. Cancer Control 2015; 22:60-78. [DOI: 10.1177/107327481502200109] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Affiliation(s)
- Laura S. Connelly-Smith
- Seattle Cancer Care Alliance, School of Medicine, University of Washington, Seattle, Washington
- Division of Hematology, School of Medicine, University of Washington, Seattle, Washington
| | - Michael L. Linenberger
- Seattle Cancer Care Alliance, School of Medicine, University of Washington, Seattle, Washington
- Division of Hematology, School of Medicine, University of Washington, Seattle, Washington
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
42
|
Abstract
Thrombotic and cardiovascular events are among the leading causes of death for patients with polycythemia vera (PV), and thrombosis history is a key criterion for patient risk stratification and treatment strategy. Little is known, however, about mechanisms of thrombogenesis in patients with PV. This report provides an overview of thrombogenesis pathophysiology in patients with PV and elucidates the roles of conventional and nonconventional thrombotic risk factors. In addition to several conventional risk factors for thrombosis, clinical data have implicated increased hematocrit and red blood cell adhesiveness, activated platelets, leukocytosis, and elevated JAK2(V617F) allele burden in patients with PV. Furthermore, PV-related inflammation may exacerbate thrombogenesis through varied mechanisms, including endothelial damage, inhibition of natural anticoagulant pathways, and secretion of procoagulant factors. These findings suggest a direct link between myeloproliferation and thrombogenesis in PV, which is likely to provide new opportunities for targeted antithrombotic interventions aimed at decreasing PV-related morbidity and mortality.
Collapse
|
43
|
Abstract
Abstract
Thrombocytosis has a large number of potential underlying causes, but the dominant group of hematological conditions for consideration in this setting are the myeloproliferative neoplasms (MPNs). In this chapter, we consider several key linked questions relating to the management of thrombocytosis in MPNs and discuss several issues. First, we discuss the differential diagnosis of thrombocytosis, which myeloid disorders to consider, and practical approaches to the discrimination of each individual MPN from other causes. Second, there have been several major advances in our understanding of the molecular biology of these conditions and we discuss how these findings are likely to be practically applied in the future. Third, we consider whether there is evidence that thrombocytosis contributes to the complications known to be associated with MPN: thrombosis, hemorrhage and transformation to leukemia and myelofibrosis. Last, we review current ideas for risk stratification and management of essential thrombocythemia and polycythemia vera as the 2 entities within the MPN family that are most frequently associated with thrombocytosis.
Collapse
|
44
|
Abstract
Polycythemia vera (PV) is a chronic myeloproliferative neoplasm associated with JAK2 mutations (V617F or exon 12) in almost all cases. The World Health Organization has defined the criteria for diagnosis, but it is still unclear which parameter (hemoglobin or hematocrit) is the most reliable for demonstrating increased red cell volume and for monitoring response to therapy; also, the role of bone marrow biopsy is being revisited. PV is associated with reduced survival because of cardiovascular complications and progression to post-PV myelofibrosis or leukemia. Criteria for risk-adapted treatment rely on the likelihood of thrombosis. Controlled trials have demonstrated that incidence of cardiovascular events is reduced by sustained control of hematocrit with phlebotomies (low-risk patients) and/or cytotoxic agents (high-risk patients) and antiplatelet therapy with aspirin. Hydroxyurea and interferon may be used as first-line treatments, whereas busulfan is reserved for patients that are refractory or resistant to first-line agents. However, there is no evidence that therapy improves survival, and the significance of reduction of JAK2 mutated allele burden produced by interferon is unknown. PV is also associated with a plethora of symptoms that are poorly controlled by conventional therapy. This article summarizes my approach to the management of PV in daily clinical practice.
Collapse
|
45
|
Hernández-Boluda JC, Gómez M. Target hematologic values in the management of essential thrombocythemia and polycythemia vera. Eur J Haematol 2014; 94:4-11. [PMID: 24814134 DOI: 10.1111/ejh.12381] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/16/2014] [Indexed: 12/24/2022]
Abstract
Treatment of essential thrombocythemia (ET) and polycythemia vera (PV) is aimed at preventing vascular complications, which are the main cause of morbidity and mortality in these diseases. Over the years, clinical trials have demonstrated that the incidence of thrombosis and bleeding can be reduced by controlling the blood cell counts, but the target hematological levels have varied across the studies. In this article, we review the evidence supporting the use of predefined target hematologic values for the management of ET and PV in routine clinical practice. At present, the recommended target hematocrit in PV is below 45%, regardless of the patients' risk profile. Concerning platelet counts, no direct correlation has been demonstrated with thrombotic risk in either ET or PV. Thus, although cytoreductive treatment reduces the rate of vascular complications in high-risk patients, no particular threshold of the platelet counts has been shown to be more protective against thrombosis. Extreme thrombocytosis is a risk factor for bleeding, particularly when aspirin or anagrelide are given. Leukocytosis at baseline or during follow-up appears to be a risk factor for thrombosis, mostly in high-risk patients. However, the clinical benefit of strictly controlling this parameter is not yet established. Finally, standardized definitions of response to cytoreductive treatment in ET and PV have recently been published. Nevertheless, they have been produced to compare the efficacy of new therapies in clinical trials, whereas its relevance in clinical practice has been questioned in retrospective studies showing that such response definitions do not correlate with the patients' clinical outcome.
Collapse
|
46
|
JAK2V617F homozygosity drives a phenotypic switch in myeloproliferative neoplasms, but is insufficient to sustain disease. Blood 2014; 123:3139-51. [DOI: 10.1182/blood-2013-06-510222] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Key Points
JAK2V617F homozygosity drives a phenotypic switch between myeloproliferative neoplasms. JAK2V617F homozygosity is insufficient to sustain clonal expansion.
Collapse
|
47
|
Vannucchi AM, Guglielmelli P, Pieri L, Antonioli E, Bosi A. Treatment options for essential thrombocythemia and polycythemia vera. Expert Rev Hematol 2014; 2:41-55. [DOI: 10.1586/17474086.2.1.41] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
48
|
Assi TB, Baz E. Current applications of therapeutic phlebotomy. BLOOD TRANSFUSION = TRASFUSIONE DEL SANGUE 2014; 12 Suppl 1:s75-83. [PMID: 24120605 PMCID: PMC3934278 DOI: 10.2450/2013.0299-12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 05/20/2013] [Indexed: 12/19/2022]
Affiliation(s)
- Tarek Bou Assi
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Centre, Beirut, Lebanon
| | - Elizabeth Baz
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Centre, Beirut, Lebanon
| |
Collapse
|
49
|
Tam CS, Nazha A, Verstovsek S. Pharmacotherapy of polycythemia vera. Expert Opin Orphan Drugs 2013. [DOI: 10.1517/21678707.2013.854164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
50
|
Could hypoxia increase the prevalence of thrombotic complications in polycythemia vera? Blood Coagul Fibrinolysis 2013; 24:311-6. [PMID: 23392352 DOI: 10.1097/mbc.0b013e32835bfdb9] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Thromboses represent a major cause of morbidity and mortality in polycythemia vera but the contributing mechanisms are not fully described. To evaluate whether environmental conditions such as altitude/hypoxia could impact thrombosis history, we retrospectively analyzed thrombosis history in 71 polycythemia vera patients living at an elevation of 5000 feet or more in the Salt Lake City (SLC) area and 166 polycythemia vera patients living near sea level in the Baltimore (BLM) area. The SLC cohort was older with a longer disease duration. No significant differences in type of anticoagulation therapy or prothrombotic factors were present between the two cohorts. After adjusting for age, sex and disease duration, SLC patients experienced an estimated 3.9-fold increase in the odds of a history of thrombosis compared with BLM patients (95% confidence interval 1.8-7.6; P=0.0004). A history of a cardiovascular event was present in 58% of the SLC patients compared with 27% of the BLM patients (P<0.0001). Before diagnosis, thrombosis occurred in 18 and 4% of the SLC and BLM groups, respectively (P=0.003). No correlation between the JAK2 allele burden and thrombosis was observed in this study. This retrospective study suggests that even moderate hypoxia associated with 5000 feet elevation should be considered as an independent prothrombotic risk factor. This observation needs to be confirmed by prospective studies.
Collapse
|