Persistently enhanced platelet activation has been characterized in polycythemia vera (PV) and essential thrombocythemia (ET) and shown to contribute to a higher risk of both arterial and venous thrombotic complications. The incidence of major bleeding complications is also somewhat higher in PV and ET than in the general population. Although its efficacy and safety was assessed in just 1 relatively small trial in PV, low-dose aspirin is currently recommended in practically all PV and ET patients. Although for most patients with a thrombosis history the benefit/risk profile of antiplatelet therapy is likely to be favorable, in those with no such history this balance will depend critically on the level of thrombotic and hemorrhagic risks of the individual patient. Recent evidence for a chemopreventive effect of low-dose aspirin may tilt the balance of benefits and harm in favor of using aspirin more broadly, but the potential for additional benefits needs regulatory scrutiny and novel treatment guidelines. A clear pharmacodynamic rationale and analytical tools are available for a personalized approach to antiplatelet therapy in ET, and an improved regimen of low-dose aspirin therapy should be tested in a properly sized randomized trial.

Polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis are collectively classified as BCR-ABL 1–negative myeloproliferative neoplasms (MPNs). Chronic myeloid neoplasms share a common stem cell–derived clonal heritage of altered proliferation and differentiation, and their phenotypic diversity is attributed to differences in specific genetic rearrangements or mutation(s) that underlie the clonal myeloproliferation.1  Although considered relatively indolent diseases, MPNs are at lifelong enhanced risk of thrombosis, hemorrhage, and myelofibrotic or leukemic transformation.2-4  The aim of current therapy is to reduce the risk of thrombosis or hemorrhage in PV and ET, and to address the presenting major clinical issues in primary myelofibrosis. In particular, low-dose aspirin (81-100 mg/d), alone or in combination with phlebotomy and/or hydroxyurea, represents a consistent component of management in practically all PV and ET patients, independently of risk stratification.5,6  Inherent to these treatment recommendations are 2 distinct assumptions: (1) that the benefit/risk profile of antiplatelet therapy is consistently favorable in PV and ET, also for patients without prior thrombosis; and (2) that the same aspirin dose and dosing regimen is appropriate for both PV and ET and is comparable to that recommended in non-MPN, high-risk patients.

The aims of this perspective are to challenge these assumptions based on a critical review of the available data, to suggest a pharmacodynamic basis for individualized antiplatelet treatment, and to discuss the need for new randomized trials of antiplatelet agents in PV and ET.

Platelet activation, atherothrombosis, and venous thromboembolism

Enhanced platelet activation has been consistently demonstrated in PV and ET, by different groups and methods either in vivo7,8  or ex vivo9,10  suggesting a pathogenetic link between activated platelets and thrombotic complications. In particular, markedly enhanced urinary excretion of thromboxane (TX) A2 metabolites characterizes untreated ET and PV patients.7,8  TX metabolite excretion is a validated index of in vivo platelet activation.11  As shown in Figure 1, the urinary excretion rates of 11-dehydro-TXB2 reported in untreated ET7  and PV8  patients are at least comparable to unstable angina12  and higher than a variety of clinical settings at increased cardiovascular risk.13-19  Moreover, a role for circulating immature platelets as contributors to cardiovascular events and/or poor antiplatelet drug response has been emerging in both ET10  and non-MPN diseases.20,21  Interestingly, immature platelets are increased in ET and have been associated with a higher rate of thrombosis independently of thrombocythosis.10,22-25  Moreover, JAK2 V617F has been associated with a significantly higher number of immature platelets in ET.24,25 

Figure 1

Rate of TXB2 production in healthy subjects (A) and urinary excretion rates of 11-dehydro-TXB2 in clinical settings at high cardiovascular risk. (A) The metabolic fate of TXA2 in vivo and the calculated rate of its production in healthy subjects on the basis of TXB2 infusions and measurement of its major urinary metabolite.11,47  (B) Mean (± standard deviation) or median (interquartile range) urinary excretion rates of 11-dehydro-TXB2 in clinical settings characterized by high cardiovascular risk.7,8,12-19  CHD, coronary heart disease; PCI, percutaneous coronary intervention; T2DM, type 2 diabetes mellitus.

Figure 1

Rate of TXB2 production in healthy subjects (A) and urinary excretion rates of 11-dehydro-TXB2 in clinical settings at high cardiovascular risk. (A) The metabolic fate of TXA2 in vivo and the calculated rate of its production in healthy subjects on the basis of TXB2 infusions and measurement of its major urinary metabolite.11,47  (B) Mean (± standard deviation) or median (interquartile range) urinary excretion rates of 11-dehydro-TXB2 in clinical settings characterized by high cardiovascular risk.7,8,12-19  CHD, coronary heart disease; PCI, percutaneous coronary intervention; T2DM, type 2 diabetes mellitus.

Close modal

Incidence of arterial thrombosis and venous thromboembolism.

The pathophysiology and incidence of platelet-mediated microvascular disturbances in PV and ET has been reviewed elsewhere26  and will not be addressed here.

The true incidence of major arterial thrombosis (ie, fatal and nonfatal myocardial infarction [MI] and ischemic stroke) and venous thromboembolism (ie, deep vein thrombosis [DVT] or pulmonary embolism [PE]) in BCR-ABL 1–negative MPNs is difficult to estimate because of several confounding factors across different studies. Most reports included major thrombotic events together with microvascular disturbances (migraine, erythromelalgia), transient ischemic attacks (TIA), and superficial venous thrombosis. Moreover, the incidence of events is rarely expressed as percent per year, and the majority of the cohorts include patients with different cytoreductive strategies, with or without previous thrombosis and/or antiplatelet treatments (mainly aspirin). In addition, the recent revisions of diagnostic criteria27,28  render comparisons across different patient cohorts quite problematic.

The incidence of major thrombosis in PV and ET derives mainly from observational and retrospective studies2-4,29-37  (Tables 1 and 2). In reviewing these studies, we only considered those that allowed estimating the annualized incidence of thrombotic and/or hemorrhagic events. Thromboses involve arteries in approximately two-thirds of cases in both disorders. The incidence of major and minor arterial and venous thromboses ranged from 1.1% to 4.9% per year in PV, and from 1.3% to 6.6% per year in ET (Table 1). Such figures vary mainly according to age and previous thrombosis, which are the 2 main factors consistently identified as predictors of thrombosis and currently used for risk stratification (Table 2).

The largest prospective data for PV and ET derive from the European Collaboration on Low-Dose Aspirin in Polycythemia Vera (ECLAP) study4  and from the Primary Thrombocythemia-1 (PT-1) trial,32  respectively. The ECLAP cohort included 1638 PV patients monitored independently of their inclusion in the randomized trial. In this cohort, the incidence of thrombosis was 4.9% per year, ranging from 2.5% to 10.9% according to age and previous thrombosis4  (Table 2). In the PT-1 trial, 809 high-risk ET patients were randomized to hydroxyurea (n = 404, 69% with previous thrombosis) or anagrelide (n = 405, 71% with previous thrombosis) on a background of aspirin.32  The overall incidence of thrombosis was 2.6% per year, with more venous thromboses in the hydroxyurea arm and more TIA in the anagrelide arm. Based on indirect comparison of different studies, the rate of first major and minor thrombosis in PV seems higher than in ET, independently of age (Table 2).2-4,29-41  Nevertheless, the incidence appears similar when only major nonfatal arterial (MI, ischemic stroke) and venous thromboembolism (DVT, PE) are considered: major arterial thrombosis ranged between 0.4% and 1.7% per year in PV and 0.6% and 1.3% in ET (Table 1); major venous thromboembolism ranged between 0.3% and 1.5% per year in PV and 0.2% and 1.5% in ET (Table 1). Moreover, the rate of cardiovascular death has been reported between 0.43% and 0.72% per year in PV2-4  and 0.47% per year in ET.3 

In the general population of the Western countries, the annual incidence of major venous thromboembolism is between 0.1% and 0.2%.42  In patients with PV43  or ET30  not treated with aspirin, the incidence of DVT and/or PE seems ∼10-fold higher, being 1.5% and 1.0% per year, respectively (Table 1). In the prospective randomized ECLAP trial43  and in the PT-1 trial,32  the PV and ET patients receiving aspirin had an incidence of major venous thromboembolism of 0.5% and 0.6% per year, respectively.

In a meta-analysis of 6 primary prevention aspirin trials in ∼95 000 participants, the control annual rate of any major coronary event and ischemic stroke was 0.34% and 0.36%, respectively.44  Similar control rates (ie, 0.3% and 0.4% per year, respectively) have been reported in a meta-analysis of randomized trials of cyclooxygenase (COX)–2 inhibitors and traditional nonsteroidal anti-inflammatory drugs involving ∼145 000 participants.45  In the control arm of the ECLAP trial in PV patients, the annual rate of myocardial infarction and ischemic stroke was 0.2% and 1.1%, respectively.43 

Incidence of recurrent arterial and venous thromboembolism.

The risk of recurrence after a first thrombosis is difficult to estimate because the published cohorts included patients with and without a previous event.2-4,30-37  There is only one retrospective study investigating the incidence of recurrent thrombosis in 235 PV and 259 ET patients after a first thrombotic event.29  Microvascular events, except for TIA, were not included as a primary outcome. Overall, thrombosis recurred in 166 patients (33.6%), corresponding to 5.6% per year; recurrence was 6.0% and 5.3% per year in PV and ET, respectively, with a 1.7-fold increased risk in patients aged >60 years.29 

Efficacy and safety of aspirin

Aspirin prevents fatal and nonfatal major vascular events by ∼25% in non-MPN patients at high cardiovascular risk because of a previous vascular event.44  Recently, the efficacy of low-dose aspirin in the secondary prevention of venous thromboembolism after vitamin K antagonists has been reported in non-MPN patients.46  Low-dose aspirin increases the risk of major extracranial (largely gastrointestinal) bleeds by ∼1.5- to 2.0-fold in randomized trials and observational studies.44,47  Because the clinical development of aspirin as an antithrombotic agent was not industry driven, a wide range of doses and dosing regimens were tested by independent studies, from as low as 30 mg to as high as 1500 mg daily.47  Lower doses (75-150 mg) were as effective as higher doses and associated with reduced gastrointestinal toxicity.47 

Primary prophylaxis.

In the ECLAP trial, 518 PV patients, 90% without previous thrombosis, were randomized to 100 mg aspirin daily or a placebo.43  Aspirin nonsignificantly reduced the risk of a combined primary end point including nonfatal MI, stroke, or cardiovascular death (relative risk, 0.41; 95% confidence interval [CI], 0.15-1.15; P = .08), but the inclusion of major venous thromboembolism in a combined coprimary end point led to a statistically significant risk reduction (relative risk, 0.40; 95% CI, 0.18-0.91; P = .02).43  This apparent protection appears larger than in non-MPN patients44 ; however, the ECLAP trial was terminated prematurely because of the slow recruitment rate, enrolling approximately one-third of the planned population. Thus, the apparent benefit of antiplatelet therapy remains of substantial statistical uncertainty in this setting. In the whole prospective ECLAP cohort (including both randomized and observational studies), 38% of patients had a prior thrombosis, and antiplatelet therapy reduced the incidence of cardiovascular events by 28%4  (ie, an effect size comparable to non-MPN patients).44  Based on these results, primary prophylaxis with low-dose (81-100 mg) aspirin is currently recommended in PV.5,6,48 

No randomized trials have directly assessed the efficacy and safety of low-dose aspirin in ET. Microvascular disturbances typical of ET, such as erythromelalgia, TIA, and migraine-like symptoms, have been reported to be extremely sensitive to aspirin in small-sized studies.49,50  An early trial randomized 114 ET patients to hydroxyurea (n = 56, 54% with previous thrombosis) or observation (no myelosuppression, n = 58, 38% with previous thrombosis), and 70% of the patients in each arm received antiplatelet agents (300 mg aspirin or 500 mg ticlopidine daily).51  In the hydroxyurea group, 2 patients (3.6%) experienced MI and ischemic stroke, whereas in the control group, 14 (24%) had major or minor thromboses, but only 2 patients (3.4%) had major thromboses (ie, ischemic stroke and DVT).51 

A recent retrospective study in low-risk ET patients (<60 years, without thrombosis and cytoreduction), 198 on antiplatelet drugs only (mainly 100 mg aspirin daily) and 102 under careful observation, showed a similar incidence of thrombosis with or without aspirin (2.1% vs 1.7% per year, respectively; P = .6).40 

Secondary prophylaxis.

The effects of cytoreduction and antithrombotic prophylaxis have been investigated in the aforementioned cohort including only PV and ET patients with previous thrombosis.29  By multivariate analysis, cytoreduction was associated with a statistically sign1ificant risk reduction in the entire cohort (hazard ratio [HR] = 0.53; 95% CI, 0.38-0.73), which was confirmed in the patients with a first arterial thrombosis (HR = 0.47; 95% CI, 0.31-0.70); in patients with a first venous event, the risk reduction associated with cytoreduction was not statistically significant (HR = 0.66; 95% CI, 0.38-1.13). In patients with a first arterial thrombosis, antiplatelet agents were associated with a nonsignificant risk reduction (HR = 0.67; 95% CI, 0.41-1.08), whereas in patients with a first venous event, both antiplatelet drugs and vitamin K antagonists were associated with significantly lower risk of recurrence (HR = 0.42; 95% CI, 0.22-0.77; and HR = 0.32; 95% CI, 0.15-0.64, respectively).29 

Safety.

Aspirin was initially discouraged in PV after the Polycythemia Vera Study Group 05 trial,52  which randomized 166 patients to phlebotomy plus 300 mg aspirin thrice daily and 75 mg dipyridamole thrice daily or 32P. No hemorrhagic complications occurred in the 32P arm, whereas 7.2% of patients in the antiplatelet arm experienced bleeding, most likely reflecting the high aspirin dose.

In patients with MPN, the overall incidence of major hemorrhages (both intracranial and gastrointestinal) ranges between 0.3% and 0.8% per year (Table 1). The rate of intracranial bleeding has been reported between 0.03% and 0.2% per year; however, in prospective studies the rate of intracranial bleeding among the PV43  or ET32  aspirin-treated patients has been consistently reported as 0.2% per year (Table 1), which seems higher than that observed in the aspirin-allocated participants in primary and secondary prevention trials (0.04%).44  The incidence of major gastrointestinal bleeding has been reported between 0% and 0.3% per year in the absence of aspirin treatment,30,43  but in the cohorts where aspirin has been administrated to >70% of patients, the incidence of major gastrointestinal bleeding was between 0.3% and 1.2% per year (Table 1). In primary prevention trials, the annual rate of major gastrointestinal bleeding was 0.07% and 0.1% per year in control and aspirin-treated participants, respectively.44  However, any indirect comparison of the data obtained in MPN patients and non-MPN individuals is weakened by the relatively small number of MPN patients investigated and by the high variability of the baseline risk of upper gastrointestinal bleeding depending on age and prior gastrointestinal history (Figure 2).47 

Figure 2

Estimated rates of upper gastrointestinal complications in men, according to age and the presence or absence of a history of such complications and regular treatment with low-dose aspirin. The solid lines connecting each pair of yellow and red symbols depict the absolute excess of complications related to aspirin therapy. Adapted from Patrono et al47  with permission.

Figure 2

Estimated rates of upper gastrointestinal complications in men, according to age and the presence or absence of a history of such complications and regular treatment with low-dose aspirin. The solid lines connecting each pair of yellow and red symbols depict the absolute excess of complications related to aspirin therapy. Adapted from Patrono et al47  with permission.

Close modal

In retrospective PV cohorts, fatal bleeding accounted for 3.1% of all deaths,2  with 0.07% per year incidence.3  In the ECLAP cohort, the incidence of fatal bleeding was 0.15% per year, independently of antiplatelet treatment.4  In patients with ET, the annual rate of fatal bleeding was 0.02% in a retrospective study (57% of patients on aspirin)3  and 0.3% in a prospective study (all patients on aspirin).32 

Extreme thrombocytosis has been associated with increased bleeding, although the platelet threshold remains controversial (between 750 and 1500 × 109/L) and the relationship is likely nonlinear.53  This phenomenon has been attributed to acquired von Willebrand syndrome.54,55  However, in 99 ET patients aged <60 years and with extreme thrombocytosis at diagnosis, the incidence of major thrombosis was 0.15% and 0.13% per year with and without cytoreduction, respectively, with a major bleeding rate of 0.05% and 0.04% per year, thus challenging the association of thrombocytosis with bleeding.56  Aspirin use was comparable in the 2 groups (44% and 54%, respectively), but information concerning bleeding among the aspirin-treated patients is lacking.56  Moreover, in a large retrospective cohort of 891 ET patients,36  the annual rate of major bleeding was 0.79%, and, on multivariate analysis, thrombocytosis >1000 × 109/L did not enhance bleeding risk, whereas aspirin increased the risk by 3.1-fold independently of the platelet count. In aspirin-naive patients, the incidence of major bleeding was 0.46% and 0.49% per year in patients below and above 1000 × 109 platelets per L, respectively; in aspirin-treated patients, the incidence was 0.95% and 1.08% per year below and above the same threshold, respectively.36  In contrast, in a cohort of low-risk ET patients the incidence of major bleeding in those receiving aspirin or not was 1.26% and 0.6% per year, respectively (P = .09), and only thrombocytosis >800 × 109/L was independently associated with increased bleeding.40  In another retrospective study including aspirin-treated ET patients, bleeding risk was doubled in association with thrombocytosis >1000 × 109/L but was not affected by aspirin.37  Finally, in the ET patients recruited in the PT-1 high-risk trial, platelet count at diagnosis could not predict future bleedings.53  The analysis of 21 887 longitudinal blood counts collected from 776 aspirin-treated ET patients enrolled to 1 of the 3 PT-1 studies confirmed the lack of predictivity of platelet count at diagnosis. However, during follow-up the risk of major bleeding approximately doubled for platelet counts between 750 and 1000 × 109/L and increased exponentially beyond 1000 × 109/L.53 

How to assess the adequacy of platelet COX-1 inhibition and aspirin “resistance”

The mechanism of action of low-dose aspirin in preventing atherothrombosis relies on the irreversible acetylation of a single serine residue near the active site of platelet prostaglandin (PG) H–synthase 1, hampering the access of arachidonic acid (AA) to the catalytic site of COX activity and inhibiting the subsequent biosynthesis of TXA2.47  In spite of aspirin’s short half-life (20 minutes in the human circulation), blockade of platelet COX-1 activity lasts for the entire life span due to the limited platelet capacity for new protein synthesis, thus allowing once-daily dosing. Moreover, aspirin acetylates a variable fraction of COX isozymes in the bone marrow megakaryocytes57  and proplatelets, as suggested by a 24- to 48-hour delay between aspirin withdrawal and reappearance of TXA2 biosynthesis in peripheral platelets (Figure 3).58-60  Thus, under normal conditions, a 24-hour dosing interval of a short-lived drug is ensured by a favorable combination of the irreversible inhibition of a slowly renewable drug target (platelet COX-1) and an effect on progenitors, leading to a new platelet progeny with a largely nonfunctioning enzyme throughout the dosing interval (Figure 4).25 

Figure 3

Recovery of serum TXB2 after aspirin withdrawal in healthy subjects. Following aspirin withdrawal in 48 aspirin-treated healthy subjects, the recovery of platelet COX-1 activity shows a 24- to 48-hour delay before linear increase of serum TXB2. This delay is likely due to the degree of acetylation of bone marrow megakaryocytes and proplatelets. Adapted from Santilli et al60  with permission.

Figure 3

Recovery of serum TXB2 after aspirin withdrawal in healthy subjects. Following aspirin withdrawal in 48 aspirin-treated healthy subjects, the recovery of platelet COX-1 activity shows a 24- to 48-hour delay before linear increase of serum TXB2. This delay is likely due to the degree of acetylation of bone marrow megakaryocytes and proplatelets. Adapted from Santilli et al60  with permission.

Close modal
Figure 4

Model of altered aspirin pharmacodynamics in ET. Under conditions of normal megakaryopoiesis, low-dose aspirin acetylates COX isozymes in both circulating platelets and bone marrow megakaryocytes, and only negligible amounts of unacetylated enzymes are resynthesized within the 24-hour dosing interval. This pharmacodynamic pattern is associated with virtually complete suppression of platelet TXA2 production in peripheral blood throughout the dosing interval. Under conditions of abnormal megakaryopoiesis, an accelerated rate of COX-isozyme resynthesis is biologically plausible in bone marrow megakaryocytes, accompanied by faster release of immature platelets with unacetylated enzyme(s) during the aspirin dosing interval, and in particular between 12 and 24 hours after dosing. This pharmacodynamic pattern is associated with incomplete suppression of platelet TXA2 production in peripheral blood and time-dependent recovery of TXA2-dependent platelet function during the 24-hour dosing interval. Immunohistochemistry panels depict megakaryocytes from an ET patient stained for COX-1 and from a normal subject stained for COX-2, and peripheral washed platelets from an ET patient stained for COX-2. Reprinted from Pascale et al25  with permission.

Figure 4

Model of altered aspirin pharmacodynamics in ET. Under conditions of normal megakaryopoiesis, low-dose aspirin acetylates COX isozymes in both circulating platelets and bone marrow megakaryocytes, and only negligible amounts of unacetylated enzymes are resynthesized within the 24-hour dosing interval. This pharmacodynamic pattern is associated with virtually complete suppression of platelet TXA2 production in peripheral blood throughout the dosing interval. Under conditions of abnormal megakaryopoiesis, an accelerated rate of COX-isozyme resynthesis is biologically plausible in bone marrow megakaryocytes, accompanied by faster release of immature platelets with unacetylated enzyme(s) during the aspirin dosing interval, and in particular between 12 and 24 hours after dosing. This pharmacodynamic pattern is associated with incomplete suppression of platelet TXA2 production in peripheral blood and time-dependent recovery of TXA2-dependent platelet function during the 24-hour dosing interval. Immunohistochemistry panels depict megakaryocytes from an ET patient stained for COX-1 and from a normal subject stained for COX-2, and peripheral washed platelets from an ET patient stained for COX-2. Reprinted from Pascale et al25  with permission.

Close modal

The platelet pharmacology of aspirin has been described best by the ex vivo synthesis of TXA2 during whole blood clotting, measured as its stable hydrolysis product, TXB2, in serum (Figure 5).61  This relatively simple assay of aspirin pharmacodynamics characterized the cumulative nature of platelet COX-1 inactivation upon repeated daily dosing and demonstrated saturability of the antiplatelet effect at low doses,62  in the same dose range later shown to be clinically effective.63  High residual serum TXB2 has been identified as a predictor of poor outcome in aspirin-treated patients presenting for cardiac catheterization.64 

Figure 5

Principle and kinetics of the serum TXB2 ex vivo assay. (A) The chain of enzymatic reactions triggered by whole blood clotting in vitro at 37°C. Thrombin generated during blood clotting activates platelet phospholipases A2 (PLA2), which releases AA from membrane phospholipids. AA is the substrate of the sequential action of COX-1 and TX synthase, resulting in TXA2 generation. Because of chemical instability of its oxane ring, TXA2 is rapidly hydrolyzed to the chemically stable, biologically inactive hydration product, TXB2, which can be measured in serum with high sensitivity and specificity. (B) The kinetics of TXB2 production during whole blood clotting at 37°C and based on data from Patrono et al.61 

Figure 5

Principle and kinetics of the serum TXB2 ex vivo assay. (A) The chain of enzymatic reactions triggered by whole blood clotting in vitro at 37°C. Thrombin generated during blood clotting activates platelet phospholipases A2 (PLA2), which releases AA from membrane phospholipids. AA is the substrate of the sequential action of COX-1 and TX synthase, resulting in TXA2 generation. Because of chemical instability of its oxane ring, TXA2 is rapidly hydrolyzed to the chemically stable, biologically inactive hydration product, TXB2, which can be measured in serum with high sensitivity and specificity. (B) The kinetics of TXB2 production during whole blood clotting at 37°C and based on data from Patrono et al.61 

Close modal

The relationship between inhibition of serum TXB2 and AA-dependent platelet function assays (ie, AA-induced optical aggregation), Verify-Now Aspirin, and urinary TXA2 metabolites (11-dehydro-TXB2) is strikingly nonlinear (Figure 6).60,65  Thus, platelet COX-1 activity must be nearly completely (∼99%) suppressed to fully inhibit in vivo platelet activation.60,65  Given this relationship, platelet function assays, including urinary 11-dehydro-TXB2, are poorly sensitive to and do not linearly reflect the degree of COX-1 inactivation.25,66  Moreover, many platelet function assays often used to measure aspirin response, are unrelated to its mechanism of action, display poor agreement,60,67,68  and give variable results upon repeated measurements.60,69  This methodological background helps explain the aspirin “resistance” phenomenon, usually defined as lower-than-expected response to aspirin in heterogeneous studies, without an objective assessment of compliance.70  Not surprisingly, the incidence of resistance ranges from 1% up to 65%, is assay-dependent, fluctuates over time, and has uncertain clinical significance.70  The International Society on Thrombosis and Haemostasis defined this phenomenon as “variability in laboratory test response,” rather than “resistance” to aspirin and recommended against monitoring aspirin response by standard aggregation.71  Aspirin “resistance” has been considered in ET.72 

Figure 6

Correlations between biochemical and functional assays of platelet response to aspirin in healthy subjects and patients with ET. Individual percent inhibition values are depicted from healthy volunteers (n = 48) during aspirin treatment (100 mg daily) and following its withdrawal. The nonlinear relationships between percent inhibition of serum TXB2 and Verify-Now Aspirin (A), AA-induced platelet aggregation (B), and urinary 11-dehydro-TXB2 (C). (D) The relationship between absolute values of serum TXB2 and Verify-Now Aspirin measurements performed in 15 ET patients during different aspirin regimens. The dotted line represents the best fitting of the experimental data (logarithmic relation between the 2 variables: y = 40ln[x] + 378, R2 = 0.43; n = 75). Each point in the graph represents the median value for each aspirin regimen, and the horizontal and vertical bars indicate the correspondent interquartile range (25th to 75th percentile). bid, twice daily; EC, enteric coated aspirin; od, once daily. Reprinted from Santilli et al60  and Pascale et al25  with permission.

Figure 6

Correlations between biochemical and functional assays of platelet response to aspirin in healthy subjects and patients with ET. Individual percent inhibition values are depicted from healthy volunteers (n = 48) during aspirin treatment (100 mg daily) and following its withdrawal. The nonlinear relationships between percent inhibition of serum TXB2 and Verify-Now Aspirin (A), AA-induced platelet aggregation (B), and urinary 11-dehydro-TXB2 (C). (D) The relationship between absolute values of serum TXB2 and Verify-Now Aspirin measurements performed in 15 ET patients during different aspirin regimens. The dotted line represents the best fitting of the experimental data (logarithmic relation between the 2 variables: y = 40ln[x] + 378, R2 = 0.43; n = 75). Each point in the graph represents the median value for each aspirin regimen, and the horizontal and vertical bars indicate the correspondent interquartile range (25th to 75th percentile). bid, twice daily; EC, enteric coated aspirin; od, once daily. Reprinted from Santilli et al60  and Pascale et al25  with permission.

Close modal

Measurements of serum TXB2 were instrumental in characterizing the inhibition of platelet COX-1 activity by low-dose aspirin in PV,73  providing the rationale for testing its efficacy and safety in this setting.43  Similarly to PV, patients with ET display enhanced TXA2 biosyntheses in vivo.7 However, the same once-daily regimen of 100 mg aspirin shown effective in PV was unable to fully inhibit platelet TXA2 production in ∼80% of ET patients.74  The residual platelet COX (both COX-1 and COX-2) activity was fully suppressed by adding aspirin to whole blood in vitro,74  thus demonstrating that platelet COX isozymes are not “resistant” to aspirin.74  Additional studies demonstrated that aspirin-insensitive TXA2 biosynthesis in ET is explained by accelerated renewal of the drug target.25  The abnormal megakaryopoiesis that characterizes ET accounts for shorter-lasting antiplatelet effects of low-dose aspirin through faster renewal of platelet COX-1, as modeled in Figure 4.25 

Is there a need for “tailored” antiplatelet therapy in MPN?

Current risk stratification in PV and ET is designed to estimate the likelihood of thrombotic complications: high-risk is defined by age ≥60 years or thrombosis history; low-risk is defined by age <60 years and no thrombosis history.5,6,48  Cardiovascular risk factors are not taken under consideration in formal risk categorization in the current clinical practice. Very recently, a prognostic score for thrombosis including some of the traditional cardiovascular risk factors (smoking habits defined as “active tobacco use,” hypertension, diabetes) and the presence of JAK2 V617F, besides age and thrombosis history, has been developed,75  but further prospective studies are needed to validate such a score. Similarly, the risk of bleeding complications is evaluated only with respect to acquired von Willebrand syndrome, especially in the presence of extreme thrombocytosis (platelets >1000 × 109/L).6  In the 2012 update on diagnosis, risk stratification, and management of PV and ET, low-dose aspirin (81 mg/d; range 40-100 mg/d) is recommended in all patients, irrespective of risk category, with the notable exception of patients with PV or ET and extreme thrombocytosis, if ristocetin cofactor activity is <30%.6  Risk-adapted therapy considers cytoreductive therapy (eg, hydroxyurea, interferon-α) according to individual disease and risk category.

As noted above, the randomized evidence for the efficacy and safety of low-dose aspirin in MPN is limited to 1 clinical trial in PV43  with inadequate statistical power to detect differences in major bleeding complications because of its relatively small sample size. Thus, whether the potential benefits of reduced thrombotic risk demonstrated in PV also apply to ET, and whether the balance of benefits and risk of bleeding complications of antiplatelet therapy is favorable in all clinical scenarios, is currently unknown. Extrapolation from randomized trials of low-dose aspirin in people without MPN44  may provide useful information, particularly for patients without thrombosis history, inasmuch as most traditional cardiovascular risk factors appear to influence both the risk of atherothrombotic and hemorrhagic complications.44  In the Antithrombotic Trialists’ meta-analysis of 6 primary prevention trials, low-dose aspirin reduced the risk of major vascular events by approximately one-tenth largely due to a reduction in nonfatal MI.44  On the other hand, low-dose aspirin roughly doubled the risk of major extracranial hemorrhages. When considering age (as a continuous variable) and prior gastrointestinal history as risk factors, the absolute excess of upper gastrointestinal complications due to aspirin can vary ∼100-fold when comparing the lowest to the highest risk category (Figure 2).47  Therefore, we suggest that when considering low-dose aspirin for primary prevention, the bleeding risk profile of the individual patient should be carefully examined along the lines depicted in Figure 2.

Finally, the recommendation of the same aspirin dose range (40-100 mg) and dosing regimen (once daily) for ET patients as for non-MPN patients implies assuming similar antiplatelet pharmacodynamics. We have shown that the duration of the antiplatelet effect of low-dose aspirin is in fact shortened in the majority of aspirin-treated ET patients, most likely reflecting accelerated renewal of the drug target.25  Inadequate suppression of platelet TXA2 production during the 24-hour dosing interval is largely overcome by a twice-daily regimen of low-dose aspirin.25  Thus, we suggest testing for the adequacy of platelet COX-1 inactivation by 2 repeated measurements of serum TXB2, a validated surrogate marker of efficacy,76  performed 24 hours after a witnessed administration of low-dose aspirin in ET patients treated for at least 2 weeks with a conventional regimen. A serum TXB2 concentration consistently above 4.0 ng/mL, the upper limit of values observed in aspirin-treated healthy subjects,60  should warrant considering a twice-daily regimen of aspirin administration, although the clinical effectiveness of such a strategy remains to be tested.

Is the chemopreventive effect of aspirin of any relevance to the balance of benefits and risks in MPN?

Long-term follow-up of patients randomized in trials of daily aspirin vs control for cardiovascular prevention has shown that low-dose aspirin reduces the incidence and mortality due to colorectal cancer after a delay of 8-10 years77,78  and reduces deaths due to several other common cancers after 5-15 years’ delay.79  The lowest doses used in these trials (75-100 mg once daily) appeared as effective as higher doses (300-1200 mg daily). Subsequently, a pooled analysis of 6 primary prevention trials of daily low-dose aspirin use (75-100 mg) found a similar reduction (HR = 0.76; 95% CI, 0.66-0.88; P = .0003) in overall cancer incidence occurring after ≥3 years on aspirin and a reduction in total cancer mortality (HR = 0.63; 95% CI, 0.47-0.86; P = .004) from 5 years onward.80 

Some caveats in interpreting these findings should be mentioned. First, cancer incidence and mortality were not prespecified end points of these cardiovascular trials. Second, the analyses by Rothwell et al78-80  excluded the Physicians’ Health Study81  and the Women’s Health Study82  because in these trials aspirin was given every other day. Although mechanistic considerations may justify a separate analysis based on the 24-hour vs 48-hour dosing interval, this was a post hoc separation that may be subject to bias.

The mechanism of the chemopreventive effects of aspirin and other nonsteroidal anti-inflammatory drugs against colorectal adenoma and cancer has long been related to shared inhibition of COX-2 activity in various cell types of the lower gastrointestinal tract.83  The main product of COX-2 in epithelial and stromal cells, PGE2, can modulate apoptosis and cell proliferation.83  Moreover, COX-2 deletion in mice and COX-2 inhibitors in humans were demonstrated to protect against the development or recurrence of both familial and sporadic colorectal adenomas.83,84  However, several findings suggest the need to reconsider both the cellular and molecular targets of aspirin action. These include (1) the demonstration that COX-1 gene deletion is just as protective as COX-2 knockout in a murine model of intestinal polyposis85 ; (2) the results of 4 placebo-controlled randomized trials86-89  of once-daily aspirin, at doses as low as 81 mg, which showed a largely similar chemopreventive effect against the recurrence of a sporadic colorectal adenoma as demonstrated with selective COX-2 inhibitors90,91 ; and (3) the recent finding of Rothwell et al78,79  of a chemopreventive effect of once-daily aspirin regimens, at doses as low as 75-100 mg, against overall cancer incidence and mortality. Thus, the apparent chemopreventive effect of aspirin in humans appears to recapitulate the unique features of its antithrombotic effect, that is, the adequacy of a 24-hour dosing interval (despite a 15- to 20-minute half-life of the drug in the human circulation) and saturability of the protective effect at low doses.84  This, in turn, could reflect a common mechanism of action of the drug in protecting against atherothrombosis and cancer (ie, permanent inactivation of platelet COX-1).92  This working hypothesis could be reconciled with the established role of COX-2 in colorectal carcinogenesis by postulating that activated platelets may induce COX-2 expression in adjacent nucleated cells (eg, stromal cells) at sites of intestinal mucosal injury.84  This hypothetical sequence would involve platelet signaling through paracrine mediators of both lipid (eg, prostanoids) and protein (eg, growth factors, inflammatory cytokines) nature, in turn inducing COX-2 expression and an eicosanoid amplification loop promoting cell proliferation and angiogenesis.84,92 

Using population-based data from the Danish health care system, Frederiksen et al93  have recently reported that MPN patients are at increased risk of developing both new hematologic and nonhematologic cancers. The standardized incidence ratio for developing a nonhematologic cancer was 1.2 (95% CI, 1.0-1.4) for patients with ET and 1.4 (95% CI, 1.3-1.5) for patients with PV. Interestingly, this study showed no increased incidence of colorectal cancer among (mostly aspirin treated) MPN patients in contrast to other solid tumors.93  Clearly, additional studies are required to test the hypothesis that long-term exposure to low-dose aspirin may exert chemopreventive effects in the setting of MPN.

The need for and feasibility of new antiplatelet trials in MPN

As detailed in the previous sections, studies in ET patients with or without previous thrombosis did not show a clear-cut benefit or harm of low-dose aspirin,29,40  possibly reflecting the observational nature and relatively small sample size of the studies. Furthermore, measurement of a surrogate biomarker of aspirin efficacy, serum TXB2,61,76  showed that platelets from ∼80% of aspirin-treated ET patients were inadequately inhibited by the traditional once-daily low-dose regimen.25,74  Extrapolation to ET patients of the 100-mg once-daily regimen tested in the ECLAP trial43  does not seem to be justified in light of altered aspirin pharmacodynamics in ET.25,74  A twice-daily regimen of 100 mg aspirin successfully improved the inhibition of serum TXB2 in a small, proof-of-concept study.25  Although twice-daily dosing may reduce compliance and inhibit prostacyclin (PGI2) biosynthesis as compared with a once-daily regimen, such an approach has been used successfully for stroke prevention in patients with cerebrovascular disease.94 

Based on the high cardiovascular risk associated with previous vascular events in ET, despite standard antiplatelet therapy, a phase 3 study testing the efficacy and safety of an improved dosing schedule vs the traditional aspirin regimen in secondary prophylaxis seems warranted. The most appropriate aspirin dose and dosing interval to be tested against the traditional dosing regimen should be identified through a properly sized, dose-finding study. This should measure mechanism-based, surrogate biomarkers of efficacy and safety, that is, serum TXB261,76  and urinary 2,3-dinor-6-keto-PGF,95  respectively, to test the adequacy of platelet COX-1 inactivation and sparing of endothelial COX-2, throughout a variable dosing interval of aspirin in the low-dose range. The most effective experimental regimen should then be tested vs the standard aspirin regimen on clinical outcomes (eg, the combination of major arterial and venous events). Major issues in designing a phase 3 study would be feasibility and estimating the expected event rate and hypothesized effect size. Concerning feasibility, the previous experience of the ECLAP trial43  has identified key issues in recruiting patients across several European countries with relatively limited funding. A transatlantic network of collaborating centers may help address these issues through a pilot feasibility study of 12-month duration, incorporating biochemical assessment of compliance as a key secondary objective.

A conservative estimate of the annual incidence of major vascular events would be ∼5% in ET patients with previous thrombosis. The benefit of the experimental aspirin regimen would be reasonably anticipated to be a 25% to 30% relative risk reduction in major vascular events.63  Depending on sample size and duration of follow-up, this randomized trial could also evaluate overall cancer incidence and mortality as key secondary end points. Such a study might provide a unique opportunity to assess whether optimizing antithrombotic treatment can reduce bone marrow fibrosis.96  Given the size of the unmet therapeutic need in high-risk ET patients, a 2 × 2 factorial design could also test the hypothesis that dual antiplatelet therapy with a novel P2Y12 blocker (prasugrel or ticagrelor)97  may further reduce the atherothrombotic burden in this setting.

Persistently enhanced platelet activation has been convincingly demonstrated in PV and ET, and this abnormality contributes to a higher risk of both arterial and venous thrombotic complications. The incidence of major bleeding complications (both intra- and extracranial) is somewhat higher in PV and ET patients than in the general population. However, it should be emphasized that other risk factors, such as age and prior ulcer history, may dramatically enhance the risk of upper gastrointestinal bleeding complications (Figure 2) to an extent that is likely to dwarf the enhanced risk due to the MPN state. In the absence of statistically robust estimates of the aspirin effect size in protecting against thrombosis and amplifying bleeding risk, it seems reasonable to suggest that the more reliable estimates of these effects in the non-MPN population44  should be applied to PV and ET patients when assessing the balance of benefits and risks of antiplatelet therapy.

Although for most PV and ET patients with a thrombosis history the benefit/risk profile of antiplatelet therapy with aspirin is likely to be favorable, in patients with no such history this balance will depend importantly on the level of thrombotic and hemorrhagic risks of the individual patient.

Recent evidence for a chemopreventive effect of low-dose aspirin may tilt the balance of benefits and harm in favor of using aspirin more broadly, but the potential for additional benefits needs regulatory scrutiny and novel treatment guidelines.

A clear pharmacodynamic rationale and analytical tools are available for a personalized approach to antiplatelet therapy in ET, and an improved regimen of low-dose aspirin therapy should be tested in a properly sized randomized trial.

The authors thank Ms Daniela Basilico and Ms Patrizia Barbi for expert editorial assistance.

This work was supported by grants from the European Commission (FP6 EICOSANOX Consortium, Innovative Medicines Initiative Joint Undertaking under grant agreement no. 115006, the SUMMIT consortium, the resources of which are composed of financial contribution from the European Union’s Seventh Framework Programme, FP7/2007-2013, and EFPIA companies’ in-kind contribution).

Contribution: C.P., B.R., and V.D.S. designed and wrote the paper.

Conflict-of-interest disclosure: C.P. received consultant and speaker fees from AstraZeneca, Bayer, Eli Lilly, Merck, NicOx, and Novartis and received grant support for investigator-initiated research from the European Commission, FP6 and FP7 Programmes, and Bayer. B.R. received speaker fees from Shire and Astra Zeneca. V.D.S. received honoraria as a consultant from Amgen-Dompè, Celgene, GlaxoSmithKline, Janssen-Cilag, Novartis, and Shire and received research grants from Janssen-Cilag and Shire.

Correspondence: Carlo Patrono, Department of Pharmacology, Catholic University School of Medicine, Largo F. Vito 1, 00168 Rome, Italy; e-mail: carlo.patrono@rm.unicatt.it.

1
Tefferi
 
A
Thiele
 
J
Vardiman
 
JW
The 2008 World Health Organization classification system for myeloproliferative neoplasms: order out of chaos.
Cancer
2009
, vol. 
115
 
17
(pg. 
3842
-
3847
)
2
Gruppo Italiano Studio Policitemia
Polycythemia vera: the natural history of 1213 patients followed for 20 years.
Ann Intern Med
1995
, vol. 
123
 
9
(pg. 
656
-
664
)
3
Passamonti
 
F
Rumi
 
E
Pungolino
 
E
, et al. 
Life expectancy and prognostic factors for survival in patients with polycythemia vera and essential thrombocythemia.
Am J Med
2004
, vol. 
117
 
10
(pg. 
755
-
761
)
4
Marchioli
 
R
Finazzi
 
G
Landolfi
 
R
, et al. 
Vascular and neoplastic risk in a large cohort of patients with polycythemia vera.
J Clin Oncol
2005
, vol. 
23
 
10
(pg. 
2224
-
2232
)
5
Vannucchi
 
AM
Insights into the pathogenesis and management of thrombosis in polycythemia vera and essential thrombocythemia.
Intern Emerg Med
2010
, vol. 
5
 
3
(pg. 
177
-
184
)
6
Tefferi
 
A
Polycythemia vera and essential thrombocythemia: 2012 update on diagnosis, risk stratification, and management.
Am J Hematol
2012
, vol. 
87
 
3
(pg. 
284
-
293
)
7
Rocca
 
B
Ciabattoni
 
G
Tartaglione
 
R
, et al. 
Increased thromboxane biosynthesis in essential thrombocythemia.
Thromb Haemost
1995
, vol. 
74
 
5
(pg. 
1225
-
1230
)
8
Landolfi
 
R
Ciabattoni
 
G
Patrignani
 
P
, et al. 
Increased thromboxane biosynthesis in patients with polycythemia vera: evidence for aspirin-suppressible platelet activation in vivo.
Blood
1992
, vol. 
80
 
8
(pg. 
1965
-
1971
)
9
Viallard
 
JF
Solanilla
 
A
Gauthier
 
B
, et al. 
Increased soluble and platelet-associated CD40 ligand in essential thrombocythemia and reactive thrombocytosis.
Blood
2002
, vol. 
99
 
7
(pg. 
2612
-
2614
)
10
Arellano-Rodrigo
 
E
Alvarez-Larrán
 
A
Reverter
 
JC
, et al. 
Platelet turnover, coagulation factors, and soluble markers of platelet and endothelial activation in essential thrombocythemia: relationship with thrombosis occurrence and JAK2 V617F allele burden.
Am J Hematol
2009
, vol. 
84
 
2
(pg. 
102
-
108
)
11
Davì
 
G
Patrono
 
C
Platelet activation and atherothrombosis.
N Engl J Med
2007
, vol. 
357
 
24
(pg. 
2482
-
2494
)
12
Vejar
 
M
Fragasso
 
G
Hackett
 
D
, et al. 
Dissociation of platelet activation and spontaneous myocardial ischemia in unstable angina.
Thromb Haemost
1990
, vol. 
63
 
2
(pg. 
163
-
168
)
13
Ciabattoni
 
G
Ujang
 
S
Sritara
 
P
, et al. 
Aspirin, but not heparin, suppresses the transient increase in thromboxane biosynthesis associated with cardiac catheterization or coronary angioplasty.
J Am Coll Cardiol
1993
, vol. 
21
 
6
(pg. 
1377
-
1381
)
14
van Kooten
 
F
Ciabattoni
 
G
Koudstaal
 
PJ
, et al. 
Increased platelet activation in the chronic phase after cerebral ischemia and intracerebral hemorrhage.
Stroke
1999
, vol. 
30
 
3
(pg. 
546
-
549
)
15
Santilli
 
F
Davì
 
G
Consoli
 
A
, et al. 
Thromboxane-dependent CD40 ligand release in type 2 diabetes mellitus.
J Am Coll Cardiol
2006
, vol. 
47
 
2
(pg. 
391
-
397
)
16
Davì
 
G
Guagnano
 
MT
Ciabattoni
 
G
, et al. 
Platelet activation in obese women: role of inflammation and oxidant stress.
JAMA
2002
, vol. 
288
 
16
(pg. 
2008
-
2014
)
17
Davì
 
G
Alessandrini
 
P
Mezzetti
 
A
, et al. 
In vivo formation of 8-Epi-prostaglandin F2 α is increased in hypercholesterolemia.
Arterioscler Thromb Vasc Biol
1997
, vol. 
17
 
11
(pg. 
3230
-
3235
)
18
Di Minno
 
G
Davì
 
G
Margaglione
 
M
, et al. 
Abnormally high thromboxane biosynthesis in homozygous homocystinuria. Evidence for platelet involvement and probucol-sensitive mechanism.
J Clin Invest
1993
, vol. 
92
 
3
(pg. 
1400
-
1406
)
19
Ciabattoni
 
G
Davì
 
G
Collura
 
M
, et al. 
In vivo lipid peroxidation and platelet activation in cystic fibrosis.
Am J Respir Crit Care Med
2000
, vol. 
162
 
4 pt 1
(pg. 
1195
-
1201
)
20
Guthikonda
 
S
Lev
 
EI
Patel
 
R
, et al. 
Reticulated platelets and uninhibited COX-1 and COX-2 decrease the antiplatelet effects of aspirin.
J Thromb Haemost
2007
, vol. 
5
 
3
(pg. 
490
-
496
)
21
Grove
 
EL
Hvas
 
AM
Kristensen
 
SD
Immature platelets in patients with acute coronary syndromes.
Thromb Haemost
2009
, vol. 
101
 
1
(pg. 
151
-
156
)
22
Watanabe
 
K
Takeuchi
 
K
Kawai
 
Y
, et al. 
Automated measurement of reticulated platelets in estimating thrombopoiesis.
Eur J Haematol
1995
, vol. 
54
 
3
(pg. 
163
-
171
)
23
Moles-Moreau
 
MP
Ternisien
 
C
Tanguy-Schmidt
 
A
, et al. 
Flow cytometry-evaluated platelet CD36 expression, reticulated platelets and platelet microparticles in essential thrombocythaemia and secondary thrombocytosis.
Thromb Res
2010
, vol. 
126
 
5
(pg. 
e394
-
e396
)
24
Panova-Noeva
 
M
Marchetti
 
M
Buoro
 
S
, et al. 
JAK2V617F mutation and hydroxyurea treatment as determinants of immature platelet parameters in essential thrombocythemia and polycythemia vera patients.
Blood
2011
, vol. 
118
 
9
(pg. 
2599
-
2601
)
25
Pascale
 
S
Petrucci
 
G
Dragani
 
A
, et al. 
Aspirin-insensitive thromboxane biosynthesis in essential thrombocythemia is explained by accelerated renewal of the drug target.
Blood
2012
, vol. 
119
 
15
(pg. 
3595
-
3603
)
26
Michiels
 
JJ
Berneman
 
Z
Schroyens
 
W
, et al. 
Platelet-mediated erythromelalgic, cerebral, ocular and coronary microvascular ischemic and thrombotic manifestations in patients with essential thrombocythemia and polycythemia vera: a distinct aspirin-responsive and coumadin-resistant arterial thrombophilia.
Platelets
2006
, vol. 
17
 
8
(pg. 
528
-
544
)
27
Tefferi
 
A
Vardiman
 
JW
Classification and diagnosis of myeloproliferative neoplasms: the 2008 World Health Organization criteria and point-of-care diagnostic algorithms.
Leukemia
2008
, vol. 
22
 
1
(pg. 
14
-
22
)
28
Tefferi
 
A
Skoda
 
R
Vardiman
 
JW
Myeloproliferative neoplasms: contemporary diagnosis using histology and genetics.
Nat Rev Clin Oncol
2009
, vol. 
6
 
11
(pg. 
627
-
637
)
29
De Stefano
 
V
Za
 
T
Rossi
 
E
, et al. 
GIMEMA CMD-Working Party
Recurrent thrombosis in patients with polycythemia vera and essential thrombocythemia: incidence, risk factors, and effect of treatments.
Haematologica
2008
, vol. 
93
 
3
(pg. 
372
-
380
)
30
Cortelazzo
 
S
Viero
 
P
Finazzi
 
G
, et al. 
Incidence and risk factors for thrombotic complications in a historical cohort of 100 patients with essential thrombocythemia.
J Clin Oncol
1990
, vol. 
8
 
3
(pg. 
556
-
562
)
31
Colombi
 
M
Radaelli
 
F
Zocchi
 
L
, et al. 
Thrombotic and hemorrhagic complications in essential thrombocythemia. A retrospective study of 103 patients.
Cancer
1991
, vol. 
67
 
11
(pg. 
2926
-
2930
)
32
Harrison
 
CN
Campbell
 
PJ
Buck
 
G
, et al. 
United Kingdom Medical Research Council Primary Thrombocythemia 1 Study
Hydroxyurea compared with anagrelide in high-risk essential thrombocythemia.
N Engl J Med
2005
, vol. 
353
 
1
(pg. 
33
-
45
)
33
Passamonti
 
F
Rumi
 
E
Arcaini
 
L
, et al. 
Prognostic factors for thrombosis, myelofibrosis, and leukemia in essential thrombocythemia: a study of 605 patients.
Haematologica
2008
, vol. 
93
 
11
(pg. 
1645
-
1651
)
34
Carobbio
 
A
Finazzi
 
G
Antonioli
 
E
, et al. 
Thrombocytosis and leukocytosis interaction in vascular complications of essential thrombocythemia.
Blood
2008
, vol. 
112
 
8
(pg. 
3135
-
3137
)
35
Carobbio
 
A
Thiele
 
J
Passamonti
 
F
, et al. 
Risk factors for arterial and venous thrombosis in WHO-defined essential thrombocythemia: an international study of 891 patients.
Blood
2011
, vol. 
117
 
22
(pg. 
5857
-
5859
)
36
Finazzi
 
G
Carobbio
 
A
Thiele
 
J
, et al. 
Incidence and risk factors for bleeding in 1104 patients with essential thrombocythemia or prefibrotic myelofibrosis diagnosed according to the 2008 WHO criteria.
Leukemia
2012
, vol. 
26
 
4
(pg. 
716
-
719
)
37
Palandri
 
F
Polverelli
 
N
Catani
 
L
, et al. 
Bleeding in essential thrombocythaemia: a retrospective analysis on 565 patients.
Br J Haematol
2012
, vol. 
156
 
2
(pg. 
281
-
284
)
38
McIntyre
 
KJ
Hoagland
 
HC
Silverstein
 
MN
, et al. 
Essential thrombocythemia in young adults.
Mayo Clin Proc
1991
, vol. 
66
 
2
(pg. 
149
-
154
)
39
Ruggeri
 
M
Finazzi
 
G
Tosetto
 
A
, et al. 
No treatment for low-risk thrombocythaemia: results from a prospective study.
Br J Haematol
1998
, vol. 
103
 
3
(pg. 
772
-
777
)
40
Alvarez-Larrán
 
A
Cervantes
 
F
Pereira
 
A
, et al. 
Observation versus antiplatelet therapy as primary prophylaxis for thrombosis in low-risk essential thrombocythemia.
Blood
2010
, vol. 
116
 
8
(pg. 
1205
-
1210, quiz 1387
)
41
Barbui
 
T
Thiele
 
J
Carobbio
 
A
, et al. 
Disease characteristics and clinical outcome in young adults with essential thrombocythemia versus early/prefibrotic primary myelofibrosis.
Blood
2012
, vol. 
120
 
3
(pg. 
569
-
571
)
42
Goldhaber
 
SZ
Venous thromboembolism: epidemiology and magnitude of the problem.
Best Pract Res Clin Haematol
2012
, vol. 
25
 
3
(pg. 
235
-
242
)
43
Landolfi
 
R
Marchioli
 
R
Kutti
 
J
, et al. 
European Collaboration on Low-Dose Aspirin in Polycythemia Vera Investigators
Efficacy and safety of low-dose aspirin in polycythemia vera.
N Engl J Med
2004
, vol. 
350
 
2
(pg. 
114
-
124
)
44
Baigent
 
C
Blackwell
 
L
Collins
 
R
, et al. 
Antithrombotic Trialists’ (ATT) Collaboration
Aspirin in the primary and secondary prevention of vascular disease: collaborative meta-analysis of individual participant data from randomised trials.
Lancet
2009
, vol. 
373
 
9678
(pg. 
1849
-
1860
)
45
Kearney
 
PM
Baigent
 
C
Godwin
 
J
, et al. 
Do selective cyclo-oxygenase-2 inhibitors and traditional non-steroidal anti-inflammatory drugs increase the risk of atherothrombosis? Meta-analysis of randomised trials.
BMJ
2006
, vol. 
332
 
7553
(pg. 
1302
-
1308
)
46
Becattini
 
C
Agnelli
 
G
Schenone
 
A
, et al. 
WARFASA Investigators
Aspirin for preventing the recurrence of venous thromboembolism.
N Engl J Med
2012
, vol. 
366
 
21
(pg. 
1959
-
1967
)
47
Patrono
 
C
García Rodríguez
 
LA
Landolfi
 
R
, et al. 
Low-dose aspirin for the prevention of atherothrombosis.
N Engl J Med
2005
, vol. 
353
 
22
(pg. 
2373
-
2383
)
48
Barbui
 
T
Barosi
 
G
Birgegard
 
G
, et al. 
European LeukemiaNet
Philadelphia-negative classical myeloproliferative neoplasms: critical concepts and management recommendations from European LeukemiaNet.
J Clin Oncol
2011
, vol. 
29
 
6
(pg. 
761
-
770
)
49
Michiels
 
JJ
Abels
 
J
Steketee
 
J
, et al. 
Erythromelalgia caused by platelet-mediated arteriolar inflammation and thrombosis in thrombocythemia.
Ann Intern Med
1985
, vol. 
102
 
4
(pg. 
466
-
471
)
50
van Genderen
 
PJ
Mulder
 
PG
Waleboer
 
M
, et al. 
Prevention and treatment of thrombotic complications in essential thrombocythaemia: efficacy and safety of aspirin.
Br J Haematol
1997
, vol. 
97
 
1
(pg. 
179
-
184
)
51
Cortelazzo
 
S
Finazzi
 
G
Ruggeri
 
M
, et al. 
Hydroxyurea for patients with essential thrombocythemia and a high risk of thrombosis.
N Engl J Med
1995
, vol. 
332
 
17
(pg. 
1132
-
1136
)
52
Tartaglia
 
AP
Goldberg
 
JD
Berk
 
PD
, et al. 
Adverse effects of antiaggregating platelet therapy in the treatment of polycythemia vera.
Semin Hematol
1986
, vol. 
23
 
3
(pg. 
172
-
176
)
53
Campbell
 
PJ
MacLean
 
C
Beer
 
PA
, et al. 
Correlation of blood counts with vascular complications in essential thrombocythemia: analysis of the prospective PT1 cohort.
Blood
2012
, vol. 
120
 
7
(pg. 
1409
-
1411
)
54
Castaman
 
G
Lattuada
 
A
Ruggeri
 
M
, et al. 
Platelet von Willebrand factor abnormalities in myeloproliferative syndromes.
Am J Hematol
1995
, vol. 
49
 
4
(pg. 
289
-
293
)
55
Budde
 
U
Dent
 
JA
Berkowitz
 
SD
, et al. 
Subunit composition of plasma von Willebrand factor in patients with the myeloproliferative syndrome.
Blood
1986
, vol. 
68
 
6
(pg. 
1213
-
1217
)
56
Tefferi
 
A
Gangat
 
N
Wolanskyj
 
AP
Management of extreme thrombocytosis in otherwise low-risk essential thrombocythemia; does number matter?
Blood
2006
, vol. 
108
 
7
(pg. 
2493
-
2494
)
57
Rocca
 
B
Secchiero
 
P
Ciabattoni
 
G
, et al. 
Cyclooxygenase-2 expression is induced during human megakaryopoiesis and characterizes newly formed platelets.
Proc Natl Acad Sci USA
2002
, vol. 
99
 
11
(pg. 
7634
-
7639
)
58
Burch
 
JW
Stanford
 
N
Majerus
 
PW
Inhibition of platelet prostaglandin synthetase by oral aspirin.
J Clin Invest
1978
, vol. 
61
 
2
(pg. 
314
-
319
)
59
Patrono
 
C
Ciabattoni
 
G
Patrignani
 
P
, et al. 
Clinical pharmacology of platelet cyclooxygenase inhibition.
Circulation
1985
, vol. 
72
 
6
(pg. 
1177
-
1184
)
60
Santilli
 
F
Rocca
 
B
De Cristofaro
 
R
, et al. 
Platelet cyclooxygenase inhibition by low-dose aspirin is not reflected consistently by platelet function assays: implications for aspirin “resistance.”
J Am Coll Cardiol
2009
, vol. 
53
 
8
(pg. 
667
-
677
)
61
Patrono
 
C
Ciabattoni
 
G
Pinca
 
E
, et al. 
Low dose aspirin and inhibition of thromboxane B2 production in healthy subjects.
Thromb Res
1980
, vol. 
17
 
3-4
(pg. 
317
-
327
)
62
Patrignani
 
P
Filabozzi
 
P
Patrono
 
C
Selective cumulative inhibition of platelet thromboxane production by low-dose aspirin in healthy subjects.
J Clin Invest
1982
, vol. 
69
 
6
(pg. 
1366
-
1372
)
63
Antithrombotic Trialists’ Collaboration
Collaborative meta-analysis of randomised trials of antiplatelet therapy for prevention of death, myocardial infarction, and stroke in high risk patients.
BMJ
2002
, vol. 
324
 
7329
(pg. 
71
-
86
)
64
Frelinger
 
AL
Li
 
Y
Linden
 
MD
, et al. 
Association of cyclooxygenase-1-dependent and -independent platelet function assays with adverse clinical outcomes in aspirin-treated patients presenting for cardiac catheterization.
Circulation
2009
, vol. 
120
 
25
(pg. 
2586
-
2596
)
65
Reilly
 
IA
FitzGerald
 
GA
Inhibition of thromboxane formation in vivo and ex vivo: implications for therapy with platelet inhibitory drugs.
Blood
1987
, vol. 
69
 
1
(pg. 
180
-
186
)
66
Smith
 
JP
Haddad
 
EV
Taylor
 
MB
, et al. 
Suboptimal inhibition of platelet cyclooxygenase-1 by aspirin in metabolic syndrome.
Hypertension
2012
, vol. 
59
 
3
(pg. 
719
-
725
)
67
Blais
 
N
Pharand
 
C
Lordkipanidzé
 
M
, et al. 
Response to aspirin in healthy individuals. Cross-comparison of light transmission aggregometry, VerifyNow system, platelet count drop, thromboelastography (TEG) and urinary 11-dehydrothromboxane B(2).
Thromb Haemost
2009
, vol. 
102
 
2
(pg. 
404
-
411
)
68
Lordkipanidzé
 
M
Pharand
 
C
Schampaert
 
E
, et al. 
A comparison of six major platelet function tests to determine the prevalence of aspirin resistance in patients with stable coronary artery disease.
Eur Heart J
2007
, vol. 
28
 
14
(pg. 
1702
-
1708
)
69
Muir
 
AR
McMullin
 
MF
Patterson
 
C
, et al. 
Assessment of aspirin resistance varies on a temporal basis in patients with ischaemic heart disease.
Heart
2009
, vol. 
95
 
15
(pg. 
1225
-
1229
)
70
Krishna
 
V
Diamond
 
GA
Kaul
 
S
Do platelet function testing and genotyping improve outcome in patients treated with antithrombotic agents?: the role of platelet reactivity and genotype testing in the prevention of atherothrombotic cardiovascular events remains unproven.
Circulation
2012
, vol. 
125
 
10
(pg. 
1288
-
1303, discussion 1303
)
71
Michelson
 
AD
Cattaneo
 
M
Eikelboom
 
JW
, et al. 
Platelet Physiology Subcommittee of the Scientific and Standardization Committee of the International Society on Thrombosis and Haemostasis
Working Group on Aspirin Resistance
Aspirin resistance: position paper of the Working Group on Aspirin Resistance.
J Thromb Haemost
2005
, vol. 
3
 
6
(pg. 
1309
-
1311
)
72
Tefferi
 
A
Overcoming “aspirin resistance” in MPN.
Blood
2012
, vol. 
119
 
15
(pg. 
3377
-
3378
)
73
Gruppo Italiano Studio Policitemia Vera (GISP)
Low-dose aspirin in polycythaemia vera: a pilot study.
Br J Haematol
1997
, vol. 
97
 
2
(pg. 
453
-
456
)
74
Dragani
 
A
Pascale
 
S
Recchiuti
 
A
, et al. 
The contribution of cyclooxygenase-1 and -2 to persistent thromboxane biosynthesis in aspirin-treated essential thrombocythemia: implications for antiplatelet therapy.
Blood
2010
, vol. 
115
 
5
(pg. 
1054
-
1061
)
75
Barbui
 
T
Finazzi
 
G
Carobbio
 
A
, et al. 
Development and validation of an International Prognostic Score of thrombosis in World Health Organization-essential thrombocythemia (IPSET-thrombosis).
Blood
2012
, vol. 
120
 
26
(pg. 
5128
-
5133
)
76
The European Agency for the Evaluation of Medicinal Products
 
Position paper on the regulatory requirements for the authorisation of low-dose modified release ASA formulations in the secondary prevention of cardiovascular events. EMEA/CPMP/EWP/282/02/Final. http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003340.pdf. Accessed January 30, 2013.
77
Flossmann
 
E
Rothwell
 
PM
British Doctors Aspirin Trial and the UK-TIA Aspirin Trial
Effect of aspirin on long-term risk of colorectal cancer: consistent evidence from randomised and observational studies.
Lancet
2007
, vol. 
369
 
9573
(pg. 
1603
-
1613
)
78
Rothwell
 
PM
Wilson
 
M
Elwin
 
CE
, et al. 
Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials.
Lancet
2010
, vol. 
376
 
9754
(pg. 
1741
-
1750
)
79
Rothwell
 
PM
Fowkes
 
FGR
Belch
 
JFF
, et al. 
Effect of daily aspirin on long-term risk of death due to cancer: analysis of individual patient data from randomised trials.
Lancet
2011
, vol. 
377
 
9759
(pg. 
31
-
41
)
80
Rothwell
 
PM
Price
 
JF
Fowkes
 
FGR
, et al. 
Short-term effects of daily aspirin on cancer incidence, mortality, and non-vascular death: analysis of the time course of risks and benefits in 51 randomised controlled trials.
Lancet
2012
, vol. 
379
 
9826
(pg. 
1602
-
1612
)
81
Steering Committee of the Physicians’ Health Study Research Group
Final report on the aspirin component of the ongoing Physicians’ Health Study.
N Engl J Med
1989
, vol. 
321
 
3
(pg. 
129
-
135
)
82
Ridker
 
PM
Cook
 
NR
Lee
 
IM
, et al. 
A randomized trial of low-dose aspirin in the primary prevention of cardiovascular disease in women.
N Engl J Med
2005
, vol. 
352
 
13
(pg. 
1293
-
1304
)
83
Thun
 
MJ
Henley
 
SJ
Patrono
 
C
Nonsteroidal anti-inflammatory drugs as anticancer agents: mechanistic, pharmacologic, and clinical issues.
J Natl Cancer Inst
2002
, vol. 
94
 
4
(pg. 
252
-
266
)
84
Thun
 
MJ
Jacobs
 
EJ
Patrono
 
C
The role of aspirin in cancer prevention.
Nat Rev Clin Oncol
2012
, vol. 
9
 
5
(pg. 
259
-
267
)
85
Chulada
 
PC
Thompson
 
MB
Mahler
 
JF
, et al. 
Genetic disruption of Ptgs-1, as well as Ptgs-2, reduces intestinal tumorigenesis in Min mice.
Cancer Res
2000
, vol. 
60
 
17
(pg. 
4705
-
4708
)
86
Sandler
 
RS
Halabi
 
S
Baron
 
JA
, et al. 
A randomized trial of aspirin to prevent colorectal adenomas in patients with previous colorectal cancer.
N Engl J Med
2003
, vol. 
348
 
10
(pg. 
883
-
890
)
87
Baron
 
JA
Cole
 
BF
Sandler
 
RS
, et al. 
A randomized trial of aspirin to prevent colorectal adenomas.
N Engl J Med
2003
, vol. 
348
 
10
(pg. 
891
-
899
)
88
Benamouzig
 
R
Deyra
 
J
Martin
 
A
, et al. 
Daily soluble aspirin and prevention of colorectal adenoma recurrence: one-year results of the APACC trial.
Gastroenterology
2003
, vol. 
125
 
2
(pg. 
328
-
336
)
89
Logan
 
RFA
Grainge
 
MJ
Shepherd
 
VC
, et al. 
ukCAP Trial Group
Aspirin and folic acid for the prevention of recurrent colorectal adenomas.
Gastroenterology
2008
, vol. 
134
 
1
(pg. 
29
-
38
)
90
Baron
 
JA
Sandler
 
RS
Bresalier
 
RS
, et al. 
APPROVe Trial Investigators
A randomized trial of rofecoxib for the chemoprevention of colorectal adenomas.
Gastroenterology
2006
, vol. 
131
 
6
(pg. 
1674
-
1682
)
91
Bertagnolli
 
MM
Eagle
 
CJ
Zauber
 
AG
, et al. 
APC Study Investigators
Celecoxib for the prevention of sporadic colorectal adenomas.
N Engl J Med
2006
, vol. 
355
 
9
(pg. 
873
-
884
)
92
Patrono
 
C
Patrignani
 
P
García Rodríguez
 
LA
Cyclooxygenase-selective inhibition of prostanoid formation: transducing biochemical selectivity into clinical read-outs.
J Clin Invest
2001
, vol. 
108
 
1
(pg. 
7
-
13
)
93
Frederiksen
 
H
Farkas
 
DK
Christiansen
 
CF
, et al. 
Chronic myeloproliferative neoplasms and subsequent cancer risk: a Danish population-based cohort study.
Blood
2011
, vol. 
118
 
25
(pg. 
6515
-
6520
)
94
Diener
 
HC
Cunha
 
L
Forbes
 
C
, et al. 
European Stroke Prevention Study. 2. Dipyridamole and acetylsalicylic acid in the secondary prevention of stroke.
J Neurol Sci
1996
, vol. 
143
 
1-2
(pg. 
1
-
13
)
95
FitzGerald
 
GA
Oates
 
JA
Hawiger
 
J
, et al. 
Endogenous biosynthesis of prostacyclin and thromboxane and platelet function during chronic administration of aspirin in man.
J Clin Invest
1983
, vol. 
71
 
3
(pg. 
676
-
688
)
96
Campbell
 
PJ
Bareford
 
D
Erber
 
WN
, et al. 
Reticulin accumulation in essential thrombocythemia: prognostic significance and relationship to therapy.
J Clin Oncol
2009
, vol. 
27
 
18
(pg. 
2991
-
2999
)
97
Patrono
 
C
Andreotti
 
F
Arnesen
 
H
, et al. 
Antiplatelet agents for the treatment and prevention of atherothrombosis.
Eur Heart J
2011
, vol. 
32
 
23
(pg. 
2922
-
2932
)
Sign in via your Institution