Red Blood Cell Contribution to Thrombosis in Polycythemia Vera and Essential Thrombocythemia

Julien Grenier, Wassim El Nemer, and Maria De Grandis

Polycythemia vera (PV) and essential thrombocythemia (ET) are myeloproliferative neoplasms (MPN) characterized by clonal erythrocytosis and thrombocytosis, respectively. The main goal of therapy in PV and ET is to prevent thrombohemorrhagic complications. Despite a debated notion that red blood cells (RBCs) play a passive and minor role in thrombosis, there has been increasing evidence over the past decades that RBCs may play a biological and clinical role in PV and ET pathophysiology. This review summarizes the main mechanisms that suggest the involvement of PV and ET RBCs in thrombosis, including quantitative and qualitative RBC abnormalities reported in these pathologies. Among these abnormalities, we discuss increased RBC counts and hematocrit, that modulate blood rheology by increasing viscosity, as well as qualitative changes, such as deformability, aggregation, expression of adhesion proteins and phosphatidylserine and release of extracellular microvesicles. While the direct relationship between a high red cell count and thrombosis is well-known, the intrinsic defects of RBCs from PV and ET patients are new contributors that need to be investigated in depth in order to elucidate their role and pave the way for new therapeutical strategies.

SRSF2 Mutation in JAK2V617F-Associated MPNs Reduces Polycythemia, Impairs Hematopoietic Progenitor Activity

SFSR2 mutation reduces polycythemia and impairs the activity of hematopoietic stem/progenitor cells in JAK2V617F-associated myeloproliferative neoplasms (MPNs), according to a study published in Blood Cancer Journal. 

Prior research has shown that JAK2V617F is one of the most common somatic mutations associated with MPNs; in turn, SFSR2 mutations are commonly associated with JAK2V617F, especially in myelofibrosis. Nevertheless, the consequences of SRSF2 mutation in JAK2V617F-associated MPNs have yet to be clearly elucidated in existing medical literature.

Researchers conducted a study on Cre-induced SRSF2P95H/+JAK2V617F/+ knock-in mice. The research team induced Mx1Cre expression by injecting mice models with 3 doses of polyinosine-polycytosine (pl-pC) at a dose of 300 μg at 4 weeks after birth. This allowed the researchers to identify the impact of SRSF2 mutation on blood parameters and the bone marrow 24 weeks after pl-pC administration (or 28 weeks after birth).

Additional mutations or genetic abnormalities are required in association with SRSF2P95H and JAK2V617F mutations in the development of full-blown myelofibrosis.

The research team discovered that concurrent SRSF2P95H and JAK2V617F mutations resulted in a reduction in the polycythemia phenotype; mice with concurrent mutations demonstrated a significant reduction in erythrocytes, leukocytes, platelets, neutrophils, and hematocrit parameters compared to mice that only had the JAK2V617F mutation. In addition, mice with concurrent SRSF2P95H and JAK2V617F mutations had higher mean corpuscular volume (MCV) volumes compared to JAK2V617F/+ mice.

Although Jak2V617F/+ mice demonstrated significant splenomegaly, the investigators found that SRSF2P95H/+JAK2V617F/+ mice exhibited reduced spleen size. In addition, whereas JAK2V617F/+ mice exhibited bone marrow hypercellularity alongside significant increases in erythroid precursors and megakaryocyte clusters, SRSF2P95H/+JAK2V617F/+ mice exhibited normal bone marrow cellularity.

The research team found absent/mild bone marrow fibrosis at 24 weeks in both mice groups. They also reported that SRSF2P95H mutation reduced the competitiveness of hematopoietic stem/progenitor cells; in addition, mice with this mutation had reduced transforming growth factor (TGF)-β levels and increased expressions of S100A8 and S100A9 compared to mice without this mutation; overexpression of S100A8 and S100A9 in turn led to erythroid differentiation defects and myelodysplastic syndrome pathogenesis.

“In conclusion, we demonstrate that SRSF2P95H mutant reduces development of bone marrow fibrosis in JAK2V617F-induced MPNs,” the authors of the study wrote in their report. “Additional mutations or genetic abnormalities are required in association with SRSF2P95H and JAK2V617F mutations in the development of full-blown myelofibrosis.”

Read more

Ruxolitinib has positive effect on polycythemia vera symptoms

December 24, 2023

SAN DIEGO — First-line treatment with ruxolitinib led to “clinically meaningful” positive results versus best available therapy in patients with high-risk polycythemia vera, according to data presented at ASH Annual Meeting.

The therapy had a notable improved impact on symptoms, according to a German team headed by Steffen Koschmieder, MD, head of the clinical hematology laboratory at University of Aachen, Germany.

Ruxolitinib (Jakafi, Incyte) is currently approved in the European Union for patients with hydroxyurea-resistant or intolerant polycythemia vera, but hydroxyurea or ropeginterferon-alpha (Besremi, PharmaEssentia) is currently in practice as the first option.

As ruxolitinib had not been examined against best available therapy in patients with previously untreated polycythemia vera, Koschmieder’s team probed the difference, hypothesizing that ruxolitinib may have higher efficacy.

The randomized phase 2B RuxoBEAT trial — a multicenter, two-arm, open-label trial — had a target population of 190 patients in each arm and a primary endpoint of clinicohematologic complete response rate at 6 months.

By the 6-month mark, the RuxoBEAT data showed that patients in the ruxolitinib arm showed lower hematocrit, pruritus and fatigue, as well as fewer headaches, weight loss and abdominal discomfort.

While the best available therapy and ruxolitinib both displayed reduced platelet counts, white blood cell counts, hematocrit and phlebotomy rates, the best available therapy did not have the same effect on symptoms, and it did not show the impact on spleen size, hemoglobin levels or splenomegaly levels that ruxolitinib had.

Koschmieder noted that the trial is continuing and more patients are being enrolled.

Reference:

Koschmieder S, et al. Firstline treatment with ruxolitinib versus best available therapy in patients with polycythemia vera: Pre-specified interim analysis of the randomized phase 2b Ruxobeat clinical trial of the German study group for myeloproliferative neoplasms. Presented at: ASH Annual Meeting and Exposition; Dec. 9-12, 2023; San Diego.

Read more

Polycythemia Vera: Barriers to and Strategies for Optimal Management

Andrea Duminuco,1,2 Patrick Harrington,1 Claire Harrison,1 Natalia Curto-Garcia1

1Department of Haematology, Guy’s and St Thomas’ NHS Foundation Trust, London, UK; 2Haematology with BMT Unit, A.O.U. Policlinico “G.Rodolico-San Marco”, Catania, Italy

Correspondence: Claire Harrison, Guys’ and St Thomas’ Hospital, London, SE1 9RT, UK, Tel +207 188 2742, Email Claire.harrison@gstt.nhs.uk

Abstract: Polycythemia vera (PV) is a subtype of myeloproliferative neoplasms characterized by impaired quality of life and severe complications. Despite the increasingly in-depth knowledge of this condition, it necessitates a multifaceted management approach to mitigate symptoms and prevent thrombotic and hemorrhagic events, ensuring prolonged survival. The therapeutic landscape has been revolutionized in recent years, where venesection and hydroxycarbamide associated with antiplatelet therapy have a central role and are now accompanied by other drugs, such as interferon and Janus kinase inhibitors. Ongoing research and advancements in targeted therapies hold promise for further enhancing the therapeutic choice for PV management.

Keywords: polycythemia vera, barriers to treatment, current approach, future perspectives

Introduction

Polycythemia vera (PV) is a chronic myeloproliferative neoplasm characterized by the presence of erythrocytosis in peripheral blood due to an acquired mutation on the JAK2 gene (~95% with JAK2V617F in exon 14). Consequently, the JAK-STAT pathway is activated, promoting blood cell proliferation and inhibiting apoptosis. PV incidence is estimated at 22 per 100,000 population,1 and the average age of presentation is 65–74 years, although it has been described in younger patients.2,3

Excess erythrocytosis and panmyelosis characteristic of PV can lead to thrombotic events (TE – both arterial and venous), which may be the first presentation of PV or precede the diagnosis.4 Common symptoms in these patients are aquagenic pruritus, erythromelalgia (burning pain in the extremities), or hyperviscosity symptoms (headaches, blurry vision), and fatigue, among others.

Beyond the JAK2V617F mutation, other mutations have been associated with PV, such as those in exon 12 of JAK2, and other mutations involving epigenetic (ie, TET2, ASXL1) or splicing (ie, SRSF2, U2AF1); furthermore, abnormal cytogenetics such as del(20q), +8, and +9 are well described.5,6 The risk of disease progression to myelofibrosis (MF) and acute myeloid leukemia (AML) is estimated at 10 years at 4.9–6% and 2–5%, respectively, and at 20 years at 26% for MF and remains below 10% in the case of AML.7 The Mutation-Enhanced International Prognostic Scoring System for PV (MIPSS-PV) integrates genetic and clinical/demographic information to predict OS and risk of transformation to MF or AML, underlying the negative role of adverse mutations (among which spliceosome), age >67 years, leukocytosis ≥15 × 109/L, and thrombotic history.8

The current management of PV is based on risk stratification; hence, high-risk patients are defined as those aged ≥60–65 years old and/or the presence of PV-related TE. Other factors that have been considered are the presence of cardiovascular risk factors (CVRF – hypertension, hypercholesterolemia, diabetes); excess platelet levels ≥1500×109/L; need for venesection (VS) to keep hematocrit (Hct) <0.45, among others; increasingly genomic factors such as JAK2 VAF and additional mutations are also considered. Cytoreductive therapy with VS and antiplatelets is recommended for this high-risk population, while low-risk patients are managed with just venesection and antiplatelets.9,10

Here, we will focus on reviewing the classical and new therapies used in PV patients and the challenges in delivering care to this patient group, often not allowing optimal management.

Management of PV

At present, limited therapies are available to completely eradicate the neoplastic clone from which the MPN phenotype originates. Though recent studies have shown treatments are able to reduce the mutated allelic fraction, its disappearance is almost unprecedented, and the relevance of this reduction is not fully substantiated. The main goals of managing PV patients are to control hematocrit (Hct) and disease-related symptoms and reduce the risk of TE and disease progression. In this aspect, patients without a history of thrombosis and age ≤60/65 are stratified as low-risk disease, while, in case of a previous PV-related TE or age ≥60/65 years as high-risk.9,11 According to this stratification, the management approach ranges from a watch-and-wait to active treatment. Overall, VS and antiplatelet therapies are recommended for all PV patients to reduce the risk of thrombosis and would constitute watch and wait. Recent guidelines suggest cytoreductive treatment for low-risk patients with CVRF, uncontrolled Hct levels, elevated white blood cells (WBC), and extreme thrombocytosis,9 but these considerations can be challenging to implement, and, for example, guidelines are unclear as to what constitutes “elevation or extreme or uncontrolled”.

Prevention of Thrombotic Risk: General ApproachManagement of Cardiovascular Risk Factors and Lifestyle

It is well known that cardiovascular risk factors such as hypertension, hyperlipidemia, diabetes, or smoking habits can increase the risk of TE in the general population. In the PV patients, Cerquozzi et al described the correlation between the presence of hyperlipidemia (p = 0.03) and hypertension (p = 0.02) and arterial events.12 Equally, the survival of PV patients can be affected by the presence of cardiovascular risk factors.13 Therefore, it is highly recommended to manage these factors and promote a healthy lifestyle with an adequate diet, reduced smoking and alcohol consumption, and regular exercise. Moreover, some medications, such as ruxolitinib, can increase cholesterol levels; hence, a routine check of cholesterol levels is suggested in patients with this JAK inhibitor. Implementing routine cardiovascular risk factor screening for all patients and enhanced screening in some patients taking agents such as ruxolitinib is challenging, and which tools and thresholds for realization are unclear.

Few studies have focused on the role of diet in MPN in recent years. The Mediterranean diet has anti-inflammatory properties and could reduce the incidence of cardiovascular risk factors.14,15 The Nutrient Trial has explored the role of the Mediterranean diet in MPN patients, looking at reduction in inflammatory biomarkers and symptom burden, among others. Although no significant differences in the inflammatory markers were found, the adherence to this diet was good.16 Overall, a healthy and varied diet, together with regular exercise, should be encouraged for MPN patients, but, for the large part, these are not implemented in routine care.

Venesection

Venesection (VS) remains the cornerstone treatment for PV. Removing red blood cells from the peripheral blood is primarily aimed at reducing blood viscosity, thus preventing TE with associated complications and helping to alleviate symptoms common to PV, such as pruritus, headache, and fatigue.

In 1986, the PVSG-01 randomized trial (N = 431) prospectively evaluated the use of VS compared to other treatments. It demonstrated remarkable overall survival (OS) in the arm treated with VS (median 13.9 years) compared to patients treated with chlorambucil (8.9 years) or phosphorus-32 (11.8 years). Moreover, the study confirmed the hypothesis of a higher risk of transformation to AML in patients treated with alkylating or radioactive agents.17 Later, the randomized trial CYTO-PV defined the indications for VS and the Hct target to be achieved. The primary endpoint was the incidence of cerebral TE (stroke, transient ischemic attack), myocardial infarction, peripheral arterial occlusions, and venous thromboembolism were studied in two groups (target Hct <45% in one arm and between 45% and 50% in the other). The results reported a lower incidence of events (2.7%) in the group with a Hct target <45% (compared to 9.8% of the other group, p = 0.007). Moreover, considering the occurrence of superficial-vein thrombosis, the same differences were maintained (4.4% versus 10.9%, p = 0.02). No significant distinctions between the groups were reported for adverse bleeding events and/or myelofibrosis (MF)/AML/myelodysplasia progression.18

Thus, beyond the risk stratification, VS is indicated in PV patients with elevated Hct levels (>45%). In case of persistent symptomatic burden (erythromelalgia, amaurosis, persistent headache) or particular circumstances (pregnancy due to hemostatic changes, the greater thrombotic risk), the therapeutic target could also be lower (30–39%).19 Approximately 350/450 mL of blood is generally removed from the circulation during a procedure. The frequency of VS is individualized to maintain the Htc on target.

VS is generally well tolerated, but certain precautions and contraindications should be considered, and patients should be carefully evaluated before addressing the procedure in view of age and/or medical comorbidities such as hypertension or cardiac diseases.20 VS is also time-consuming and resource-intensive; it also results in a “see-saw” effect for patients, while some newer therapies (see below) aim to eradicate the need for this therapy.

Adverse effects of VS are generally mild and transient, including fatigue, light-headedness, and iron deficiency. Rarely serious events are syncope, nerve injury, vasovagal reaction, and infections.21 Typically, one VS reduces the body’s iron amount by about 250 milligrams, and questions often arise about iron supplementation to restore deposits and minimize iron deficiency-associated side effects. However, iron replacement in PV patients should be carefully and individually assessed, and overall, it is not recommended as it can increase the Hct and hemoglobin levels.22

A different procedure to reduce Hct levels may be erythrocyte-apheresis (ECP), although its usage in PV is limited. This apheresis technique is an extracorporeal blood separation whereby blood is extracted from a patient, and through an external machine, the red blood cells are discarded, and the remaining blood is returned to circulation. The risks associated, in this case, are related to the use of a larger venous catheter (with risk of bleeding and infections) or to the citrate used as an anticoagulant, which can reduce serum ionized calcium levels causing dizziness, paresthesia, twitching, muscle cramps, and tetany.23

In summary, guidelines suggest VS should begin as soon as possible after diagnosis to keep Hct <45%. Fluid replacement with VS is based on local practice and the patient’s medical history. The use of cytoreductive therapy can be considered in case of a high number of VS requirements (>6–7 procedures per year) and/or if patients develop resistance/intolerance to the procedure24 or persistent symptoms related to iron deficiency.

Antiplatelet Therapy

The use of aspirin in PV has historically been debated. Initially, in a study conducted by the Polycythemia Vera Study Group (PVSG), the use of high doses of aspirin (900 mg daily) was associated with an elevated risk of gastrointestinal bleeding.25 In 2004, the European Collaborative Low-dose Aspirin (ECLAP) randomized, double-blind trial evaluated the efficacy of aspirin (100 mg daily) versus placebo in preventing TE in PV patients. The study demonstrated that this dosage was associated with a significant reduction in deaths from cardiovascular events and nonfatal thrombosis (eg, stroke, myocardial infarction, venous thrombosis and/or pulmonary embolisms) [hazard ratio (HR) 0.4; 95% confidence interval (CI) 0.18–0.91; p = 0.02] without a significant increase of major bleeding in the aspirin group [relative risk (RR), 1.62; 95% CI, 0.27 to 9.71].26 In the case of intolerance to aspirin, such as mild indigestion, gastric ulcers or bleeding events, among others, clopidogrel (an ADP-receptor antagonist) should be used, although limited data are available about its role in preventing TE in the PV population. Furthermore, it raises an important question in those patients with acute coronary syndrome where clopidogrel is recommended as prevention in the general population, as well as the combined usage with aspirin in indicated cases.27

Anticoagulation Therapy

Direct oral anticoagulation (DOAC) therapy usage has been raised in the last year to prevent and treat TE in cardiovascular diseases.28 DOACs are studied and evaluated in solid cancers, while data in hematological neoplasms are less explored.29 The SELECT-D study, including lymphoma (n = 23) and myeloma (n = 5) patients with deep vein thrombosis (DVT) and pulmonary embolism (PE) compared the efficacy of rivaroxaban versus LMWH (dalteparin). The cumulative incidence of recurrent TE rate at 6 months was lower (4%, [95% CI 2–9]) in rivaroxaban arm versus dalteparin arm (11%, [95% CI 7–16]), with HR 0.43, 95% CI 0.19–0.99, associated with a lower rate of major bleedings events in DOAC arm compared to LMWH [4%, 95% CI; 2–8; vs 6%, 95% CI 3–11; respectively, HR 1.83, 95% CI 0.68–4.96].30 Despite the small number of hematological patients included in these studies, the use of DOAC has been extending in recent years. Ianotto et al presented a study with 25 patients (8 PV and 17 ET) treated with DOAC for atrial fibrillation (AF) and TE, with only one case of TE with treatment and 3 major bleeding (1 due to traumatic injury and 2 post-surgery).31 In a recent small review with MPN patient and disease-associated TE (n = 102), DOAC were given in 32 with only 1 case of mesenteric ischemia without other cases of recurrent TE after 84.7 patients’ years cumulatively. Equally, no major bleeding events were described and only 3 cases of minor bleeding.32 Recently, two retrospective studies led by How et al and Barbui et al have demonstrated the efficacy and safety of DOAC. How et al studied 133 MPN patients who received DOAC for TE and AF, finding a recurrent thrombosis of 5.5% (1.5–9.5%) and bleeding of 12.3% (6.4–18.2%) after 1 year. Moreover, the multivariate analysis described a high-risk of thrombosis in patients with dabigatran and edoxaban, younger age and history of thrombosis, while high WBC was associated with an increased risk of bleeding.33 Barbui et al revised 442 MPN patients with DOAC indication for AF or VTE and described rates of recurrent thrombosis in 10 cases (2.1% patients’ year) and 22 cases (9.2% patients’ year), respectively.34 Based on these, a multicenter Phase 3 prospective, randomized, and open-label trial (AVAJAK, NCT05198960) has been designed to compare the efficacy of apixaban (2.5mg bd)/rivaroxaban (10mg od) versus aspirin 100mg daily in preventing the occurrence of TE. Within the second endpoints are the bleeding events, therapeutic adherence, overall survival, and/or quality of life, among others. This study’s results would help define the role of DOAC in the MPN population.

A brief summary of the described studies is reported in Table 1.

Despite the current limited knowledge, overall, the use of DOAC has been extended and appears safe and efficacious for preventing and treating TE in PV.

Cytoreductive Treatment

Hydroxycarbamide (HC) has been the gold standard treatment in PV patients for years. It is a DNA synthesis inhibitor due to the activity of the hydroxylamine group (-NHOH) that interferes with essential enzymes for DNA synthesis, such as ribonucleotide-diphosphate reductase.35

The PVSG and the French Polycythemia Study Group reported the first evidence of HC’s role in PV in the 1970s. PSVG compared 51 patients treated with HC and 194 only with VS, evidencing an inferior thrombotic risk in the HC arm (9.8% vs 32.8% in the VS group; p = 0.009).36 Conversely, in a randomized trial, the French Polycythemia Study Group compared HC treatment to pipobroman in 292 patients <65 years. During a median follow-up of 9 years, no differences in terms of TE were reported; however, an increased risk of leukemic transformation was observed.37 Furthermore, the extended follow-up (16 years) confirmed a higher risk of AML/myelodysplastic syndrome transformation during pipobroman therapy (cumulative incidence of 52% at 20 years vs 24% with HC)38 that was suggested in previous studies.

A subgroup analysis of the ECLAP cohort compared 1042 patients with PV who received only VS or HC to maintain the Hct level <45%. The occurrence of TE was statistically higher in the VS group than in the HC arm (5.8 vs 3.0 per 100 person-years, p = 0.002) during a comparable observation period (29.9 months for VS and 34.7 for HC). MF progression was reported only in patients not treated with HC (n = 8, 2.3%) and AML transformation in 3 cases (n = 2 in the VS arm and n = 1 in the HC arm). Furthermore, the study demonstrated that the rate of TE and excess mortality was significantly higher in VS-cohort patients with high-risk disease and those who did not achieve target Hct <45% (p = 0.000).39

Recent trials have also reviewed the efficacy of HC compared to interferon-α as described by the randomized Myeloproliferative Disorders Research Consortium (MPD-RC 112) study.40 This trial demonstrated no difference between the overall response rates at 12 months between arms (69.8% for HC and 78% for IFN, p = 0.22). The PROUD-PV phase 3 clinical trial has recently published the results of monopegylated-IFN-α-2b (ropeg-IFN) use in both naïve and previously HC-treated high-risk PV patients. PROUD-PV was designed as a non-inferiority study with HC, and the study reached its primary objective of achieving complete hematological and spleen volume response by 12 months. The CONTI-PV study compared ropeg-IFN with the best available therapy as a continuation of the PROUD-PV study. Surprisingly, in the PROUD-PV study, at 12 months, the complete hematological response (CHR) favored the HC-treated cohort [75% vs 62.1%, p = 0.12],41 although in the CONTI-PV study, as ailed below, these results reverted in favor of IFN arm.42

Despite the good efficacy of HC, various side effects have been reported, as well as the onset of resistance/intolerance leading to treatment suspension. Hematological/oncological toxicities are manifested by myelosuppression, with anemia, leukopenia (the more common side effect), and thrombocytopenia, which leads to increasing infection and bleeding risk. Due to drug interactions, several gastrointestinal side effects (ie, gastritis, mucositis, and oral mucosa ulcers), such as liver toxicity and fatal and nonfatal pancreatitis, have been described. As a chemotherapy drug, it has toxicity on the reproductive system by reducing the count and motility of spermatozoa, with related effects responsible for malformations or fetal-neonatal toxicity, so it is contraindicated in pregnancy and while breastfeeding.43

On the other hand, HC has been associated with increased skin toxicities such as skin ulcers, which is a common cause of treatment interruption, as demonstrated by Antonioli et al among others. The authors showed that between 5% and 10% of MPN patients on HC stopped treatment due to these side effects.44 The development of non-melanoma skin cancers in HC-treated patients has been described in several different studies,45 even if the study MPN-K showed that patients exposed to HC had a risk of skin cancer similar to unexposed patients during a median period of 3 years.46 Patient education and skin monitoring for this toxicity are recommended.

Thus, the central challenge in managing PV patients with HC is the development of intolerance and resistance to the medication, which could lead to the interruption of therapy.47

The 2017 European Leukaemia Network (ELN) guidelines defined resistance as those who required VS to attain the established target of Hct <0.45, with platelets >400 × 109/L, and with failure to reduce splenomegaly by 50%, or to relieve symptoms, despite the use of HC dose of 2 g/day for 3 consecutive months (maximum dosage). Larran et al evaluated in a retrospective study 261 PV patients who received HC for a median of 4.4 years and found that 11.5% (30 patients) were/became refractory, as defined by ELN criteria, while complete response was achieved by 24% of patients. HC resistance was associated with a higher risk of death (HR 5.6, p < 0.001) and progression to AML or MF (HR 6.8, p < 0.001), with a median survival of 1.2 years after resistance was identified.48

Aiming to provide clear and standardized indications, the 2021 ELN guidelines respecified the refractory/intolerance to HC and subsequent treatment change recommendations (detailed in Box 1). While these criteria exist, operationalizing them in practice is challenging and delayed, or lack of recognition of resistance/intolerance has been well described. A further problem in current practice is a lack of definitive data regarding the long-term toxicity of HC, even if the studies available in sickle cell disease confirm its safety in patients treated for many years.49

Interferon

In younger patients, in whom HC use could lead to critical long-term toxicities, or in those refractories to previous therapy, interferon-α (IFN) represents the treatment of choice and, more recently, is considered perhaps the optimum treatment. Several trials aimed to establish the role of IFN and have demonstrated high rates of both hematological and molecular responses through the stimulation of an immune response directed towards neoplastic cells and the antiproliferative effect on hematopoietic precursor cells.19,50 Recently, the new formulation ropeg-IFN has been tested in the PROUD-PV trial (1:1 randomized phase 3 open-label), as introduced above. The study assessed the efficacy and safety of ropeg-IFN versus HC in 254 PV patients, stratified by prior HC exposure, age at enrolment (≤60 or >60 years), and the occurrence of previous thromboembolic events. The CHR (defined as Hct <45% at least 3 months after the last VS, accompanied by platelets <400 × 109/L and white blood cells <10 × 109/L at the 12-month treatment timepoint) was achieved by 43.1% (53 of 123 patients) in the ropeg-IFN arm, confirming thus a non-inferiority versus HC. Normalization of spleen size was not demonstrated. On the other side, the ongoing CONTINUATION-PV study (extension of PROUD-PV trial) evaluated the CHR, normalization of spleen size and improvement in disease burden (such as splenomegaly, microvascular disturbances, pruritus, and headache) in subsets of patients continuing from PROUD-PV. An interim analysis at 36 months reported CHR associated with improved disease burden (including spleen reduction) in 50 (53%) of 95 patients in the ropeg-IFN group, higher than 28 (38%) of 74 patients in the HC group [1.42 (1.01 to 2.00), p = 0.044], thus demonstrating a treatment alternative for patients diagnosed with PV showing advantages compared with HC therapy beyond the second year of treatment.42

In a new recent re-evaluation after overall 5 years of treatment, 53/95 patients (55.8%) in the ropeg-IFN arm and 33/75 (44.0%) in the HC arm confirmed CHR. The occurrence of PV progression among ropeg-IFN-treated patients was 0.2%-patient-years (MF = 1), while this was 1%-patient-years in the control treatment arm (n = 4 cases, MF = 2 and AML = 2). Regarding TE, no clear differences were reported with 5 events (1%-patient-year, n = 2 in the same patient) in the ropeg-IFN arm and 5 events (1.2%-patient-year).51 The ongoing ECLIPSE PV, phase 3b, open-label, multicenter study, aims to evaluate the efficacy (in terms of hematologic response at 24 weeks), safety, and tolerability of ropeg-IFN utilizing higher dose (from 250 to 500mcg) compared to the currently labeled dosing (Q2W starting at 100 up to 500, with 50mcg increases) (NCT05481151). Finally, peginterferon importantly reduced the JAK2 V617F allelic burden or VAF significantly by 35% (starting from a median of 41%) by week 18 of treatment (p = 0.479), and to 25% by week 50 (p < 0.001).52

Concerning side effects, the use of interferon-α (IFN) is limited by the wide range of side effects. Commonly, there are flu-like symptoms, fatigue and neuropsychiatric manifestations, such as depression and anxiety, that generally lead to treatment discontinuation in approximately 24–40% of patients within 1–2 years. Neutropenia is a frequent hematological toxicity, noted in 20% of patients in some studies, as well as autoimmune disease (mainly thyroid disorders), hepatotoxicity and retinopathies.53,54 Hence, we recommend completing a baseline liver, thyroid and autoimmune profile and retinal screening before starting IFN. During the treatment period, it is essential to closely monitor for these toxicities and adjust the dose of pegylated interferon (peg-IFN) to reduce the possible side effects. Problematically, no standardized criteria exist for IFN intolerance or resistance, and the role of JAK2 VAF monitoring requires guidance, as discussed later.

Janus Kinase Inhibitor

The Janus kinase inhibitors (JAKi) widely impacted the treatment of MF, above all in the patients with worse prognosis according to the standard prognostic models,55 where over the years, several JAKi were tested and are currently used with significant results in terms of symptoms control, improvement in quality of life, and aiming to modify the disease outcome, alone or combined to other molecules with a different mechanism of action.56,57 In PV, ruxolitinib is the only JAKi licensed and widely used in intolerance or resistance to HC cases. A multicenter phase 3 (RESPONSE) trial compared ruxolitinib to other physician choices in a second-line therapy setting following HC in PV patients with splenomegaly, stratifying patients according to inadequate response or unacceptable side effects. The primary endpoints were Hct control (less than 45% in the absence of VS) and a 35% reduction of spleen size, evaluating as secondary aims the duration of response, the symptoms control and the safety. After 81 weeks in the ruxolitinib and 34 in the control arms (the median exposure to therapy assumed as data cut-off), 84.5% and 3.6% continued treatment, respectively, in the two groups, and the composite primary efficacy endpoints (Hct and spleen reduction taken together) were significantly more achieved by patients treated with JAKi (20.9% vs 0.9% in control arm, p < 0.001), without any difference according to whether patients were refractory or intolerant to HC. Moreover, at week 32, 36 of 74 patients in the ruxolitinib group (49%) reported at least a 50% reduction in the MPN-SAF total symptom score, higher than the standard-therapy group (4 of 81 patients, 5%) and a CHR in 24%.58 The RESPONSE-2 trial (enrolling HC-resistant or intolerant PV patients without splenomegaly) confirmed the efficacy of ruxolitinib compared to the best available treatment (HC, IFN, pipobroman, lenalidomide, or no therapy). Hct control (as primary endpoint) was achieved in 46 (62%) of ruxolitinib-treated patients, significantly higher than 14 (19%) of those who received the best available therapy (OR 7.28 [95% CI 3.43–15.45]; p < 0.0001).59 These results have allowed authorization of the use of ruxolitinib by different regulatory agencies such as FSA and EMA. Recently, the results of the Phase 2 trial MAJIC-PV confirmed the extended safety of ruxolitinib as a second-line treatment versus BAT in a UK cohort, assessing the complete response within 1 year, duration of response, event-free survival (EFS), control of symptoms, and the molecular response. CHR was achieved in 43% of patients on ruxolitinib compared to 26% on BAT, associated with improved thromboembolic-event-free survival (HR 0.56, 95% CI 0.32, 1.00, p = 0.05). The molecular response, evaluated as >50% reduction in JAK2 VAF, was more frequent with ruxolitinib and was correlated with reduced progression-free-survival, EFS and overall survival (all p < 0.01).60

Despite the well-assessed efficacy of ruxolitinib in controlling Hct levels, reducing spleen size, and improving symptom burden, several occurrences and/or adverse effects may limit ruxolitinib use, and its efficiency in reducing thrombosis and later events was unclear. Hematological toxicity is mainly represented by anemia and thrombocytopenia.61 Furthermore, not being a specific inhibitor of mutated JAK2, it may be related to impaired immune function with an increased risk of new infections or reactivation of latent infections,62,63 or impaired response to immunological stimulation (eg, in cases of vaccines both in MF and PV-treated patients).64,65

A further important and severe effect is the appearance of non-melanoma skin cancers, for which patients should be routinely screened for suspicious skin lesions.66 Also, in this case, MPN-K study, demonstrated how ruxolitinib shows a high risk (OR = 3.87, 95% CI 1.18–12.75), without excess risk of carcinoma and hematological second cancer compared with unexposed patients.46 Focusing on the cohort of MF patients in the COMFORT-1 trial, ruxolitinib-treated patients reported different metabolic side effects, such as weight gain and increased total cholesterol.67

Regardless of the potential adverse effects widely reported, the long-term data from the MAJIC-PV study demonstrated an acceptable safety with significative benefits, without any death related to infections, and not confirming metabolic side effects.60

The routine use of ruxolitinib as a second-line therapy is limited by cost, recognition of resistance or intolerance to HC and a lack of guidance for IFN failure. In addition, identifying early predictive parameters for an acceptable response to ruxolitinib represents a research target, such as the RR6 score for myelofibrosis.68,69 Currently, ruxolitinib is tested as a first-line PV therapy in the MITHRIDATE trial, compared to HC or IFN (any formulation) to evaluate the event-free survival (in terms of first major thrombosis/hemorrhage, death, progression/transformation), and, as second endpoints, the impact in quality-of-life, JAK2 V617 VAF reduction, and the adverse events (NCT04116502) during a follow-up of 3 years.

Pipobroman, Busulfan and Phosphorus-32

As mentioned above, pipobroman, busulfan and radioactive phosphorus were in the past widely used in the management of these conditions. They are currently recommended by the ELN only in subsequent lines of treatment after intolerance/loss of response to therapies such as either HC or IFN.70 In a Swedish cohort study, two or more lines of cytoreductive treatment (including an alkylating agent) were correlated with a 2.9-fold increase in transformation in AML/myelodysplastic syndrome rate.71 Because of the potential leukemogenicity, these agents should be reserved for elderly patients (>80 years) with a low risk of progression or those with a disease where the risk of thrombosis is superior to the risk of transformation.

A brief explanation of the available strategies to control PV clinical manifestations and reduce thrombotic risk, with their common side effects, is reported in Table 2.

Management of Symptoms

Symptomatic management of PV can be problematic. As already described, among the symptoms commonly present in patients with PV, pruritus is the most frequent (up to 70–85% of cases), with a spontaneous occurrence or evoked by water or changes in temperature (pruritus aquagenic) and causes a significant negative impact on quality of life.72 It is usually treated simultaneously with cytoreductive therapy/Hct control but can persist even in patients with well-controlled blood counts.73 Other purely symptomatic approaches are antihistamines (both H1 and H2 receptor antagonists),74,75 selective serotonin reuptake inhibitors (SSRIs),76 or anticonvulsant drugs. The referral to a dermatologist could be considered for refractory patients trying to treat with narrow-band-ultraviolet (UVB) or ultraviolet A (UVA), associated or not with oral psoralen to UV light (PUVA).77 These options are preferred in patients with only the presence of these symptoms but not with a thrombotic risk that requires cytoreductive therapy. In any case, persistent pruritus resistant to the above options can be an indication to start an active treatment just to control the symptom burden.

Regarding fatigue, no specific medicine can effectively treat this condition secondary to MPN or to side effects of treatment. Anxiety and depression related to cancer can increase it. Lifestyle modifications are central in the management, with adequate sleep with scheduled breaks, abstention from smoking and alcohol, hydration, and a balanced diet, associated with a referral to a psychologist to discuss one’s condition and receive support. The management of fatigue is a significant challenge in the modern management of PV.

Management of Thrombotic Events

The occurrence of a thrombotic event (TE) represents, as previously described, the factor that fixes patients as being at high risk of further thrombosis. Acute events should be managed according to current guidelines. TEs are divided into arterial (such as stroke or transient ischemic attack, acute myocardial infarction, and peripheral arterial thrombosis) and venous (deep vein thrombosis, superficial thrombophlebitis, and pulmonary embolism). Splanchnic venous thrombosis deserves special mention as, when necessary, it presents behind an MPN in a variable number of cases between 10% and 50%,78 where the specific cytoreductive therapy does not show effectiveness in reducing the risk of recurrence.79 For acute events involving the venous system, the first choice is low molecular weight heparin (LMWH), followed by indefinite secondary prophylaxis with vitamin K antagonists (VKA), as per guidelines.80 Novel direct oral anticoagulants (DOACs) are increasingly used as secondary prophylaxis with efficacy, although data on MPNs are still limited,31 as discussed earlier. In any case, every patient should be screened for CVRF, such as hypertension, hyperlipidemia, diabetes mellitus, and smoking history, as well as for prothrombotic-associated conditions (ie, antiphospholipid antibody syndrome, coagulation factor V and II mutations).

Conversely, in the case of an arterial event, the acute aim is the reperfusion of the affected extremity through thrombolysis or recanalization of the occluded vessel. It could be via catheter-directed thrombolysis (CDT, as interventional radiology treatment), surgical treatment (open bypass surgery, or the more common endovascular therapy, such as stenting, balloon angioplasty, or atherectomy) or use of thrombolytic drugs (serine proteases that cleave plasminogen into active plasmin such as streptokinase, alteplase, reteplase).81

Special conditions may require a particular approach. In the case of cerebral venous thrombosis, patients are stratified based on risk factors for poor outcome (malignancy, coma, deep venous thrombosis, mental status disturbance, male sex, intracranial hemorrhage) and treated with LMWH in case of lower risk, or endovascular procedures (endovascular thrombolysis or thrombectomy) or neurosurgery (decompressive craniotomy) in case of risk of high risk, followed by oral anticoagulation lifelong in case of the simultaneous severe prothrombotic condition, such as PV.82

Management of Hemorrhagic Events

Hemorrhagic events have been documented in individuals with PV, primarily affecting the integumentary system, mucous membranes, and gastrointestinal tract (particularly notable in cases with existing oesophageal varices). These occurrences are commonly attributed to persistent anticoagulation usage or platelet counts exceeding 1500 × 109/L, which increase the risk for acquired von Willebrand syndrome.83 The bleeding episodes are generally less frequent and severe events than the thrombotic counterpart and require management with tranexamic acid and/or platelet transfusion, associated with a subsequent cease/reduce aspirin shifting to DOACs (as mentioned above), and optimizing of cytoreductive treatment.80

Future Directions

In addition to the classic and established therapies currently available, there are several molecules (newly or used for different conditions) that are being tested in phase 2/3 trials and which, in the years to come, could represent one more possibility in the management of patients with PV in need of further treatment.

Concerning already available drugs, the efficacy and safety of peg-IFNα-2b in combination with ruxolitinib versus peg-IFNα-2b alone for treating HC-resistant/intolerant PV are currently being tested in a multicenter phase 2 study. The results, expected in 2028, could open a new window in the combination therapy between two already widely used molecules (NCT05870475).

Among the new molecules, rusfertide is a hepcidin-mimetic that binds ferroportin, causing a reduced availability of iron in the bone marrow, a fundamental element for erythrocytosis.84 It was tested in two different phase 2 trials. REVIVE trial (NCT04057040) enrolled patients with uncontrolled Hct despite VS (≥3 in the 6 months before study entry) associated with a cytoreductive therapy. This trial was divided into a 28-week open-label dose-finding phase (starting from rusfertide 20 mg subcutaneously, with subsequent dose titration, and associated with any prior cytoreductive treatment), followed by a 3-month double-blind randomization (compared to placebo), and a 3-year open-label extension, with rusfertide for all the patients enrolled. The results showed that the average clinically effective dose is approximately 40 mg weekly, without an increase in toxicity, whether rusfertide was administered with concurrent VS or cytoreductive therapy.85 The second trial (PACIFIC) enrolled patients with high Hct (>48%) for an induction period with 40mg twice weekly until the control of Hct value (<45%) and maintenance, with a schedule that may be adjusted every 2 to 4 weeks to maintain Hct, with a target of <43% (NCT04767802).

Based on these results, the phase 3 VERIFY trial is ongoing, recruiting PV patients who required ≥3 VS in the previous 6 months or ≥5 in the last 12 months due to inadequate Hct control. Patients with a stable cytoreductive regimen must be in a well-controlled scheme, with no need for dose modification, while patients managed only with VS must have interrupted cytoreductive therapy 2–6 months before screening. Patients are double-blinding randomized to 32 weeks of rusfertide or placebo added to each subject’s ongoing treatment.86 The results will definitively indicate the role of rusfertide in the PV setting.

Another class of targeted agents is histone deacetylase (HDAC) inhibitors. Givinostat, an HDAC inhibitor specific for JAK2 V617F-mutated cells, was tested in a phase 2 trial. Givinostat was administered in MPNs patients (including 12 with PV) at a starting dose of 50 mg twice daily. Apart from 2 patients who discontinued treatment, 7 out of 10 PV patients (70%) reached VS independence, and splenomegaly and pruritus were resolved in 70% and 90% of patients, respectively, with a reported reduction of JAK2 V617F allele burden. It was well tolerated, with no grade 4 toxicities.87

Subsequently, in a multicenter clinical trial, givinostat long-term efficacy and safety were tested. Of 45 patients enrolled and receiving treatment for a median of 4 years, complete/partial hematological remissions were described in 11% and 89%, respectively, with a Hct control (<45%) without VS and standard spleen size both observed in 56% of patients. The incidence of thrombosis was 2.3% of patients per year. Only three grade 3 and no grade 4 toxicities were observed.88 During an extended follow-up, the excellent safety profile was confirmed, with an overall response rate always more significant than 80%,89 laying the foundations for future phase 3 trials.

Vorinostat, another HDAC inhibitor, was tested in a phase 2 trial. Most patients who completed 24 weeks of treatment (24/33; 72%) reported a response, although 44% of patients discontinued the therapy due to adverse events, of which 25% of cases were severe (deep vein thrombosis, diarrhea, headache, progression to AML, palpitations, neuropathy, fatigue, and renal impairment).90 Using lower dosages of vorinostat combined with other drugs (eg, JAKi) will probably represent the therapy capable of conferring good responses without excessive toxicities.

In addition, several other molecules are currently being studied in phase 2 studies, which may represent potential mechanisms in the future. Summaries of these trials are shown in Table 3.

Conclusion

In the last decade, considerable advancements have transpired within the therapeutic landscape of PV, predominantly inspired by the revelation of the disease’s fundamental molecular driver mutations. However, in order to deliver effective care for patients, attention to details such as vascular risk, optimum control, defining how to utilize molecular assays, and recognizing when patients fail to continue to be a challenge. Notably, optimal management of debilitating symptoms, particularly fatigue, and cases with an associated complex clinical background necessitate the provision of an enhanced approach.

Read more

SRSF2 Mutations Lead to Lessened Frequency of Polycythemia in Preclinical MPN Models

Kyle Doherty

The presence of mutated SRSF2 in knock-in mouse models of JAK2 V617F–driven myeloproliferative neoplasms (MPNs) reduced the rate of polycythemia and hampered hematopoietic progenitor functions, according to findings from a preclinical study published in Blood Cancer Journal.

Findings from the study demonstrated that coexpression of mutant SRSF2 P95H decreased red blood cell (RBC), neutrophil, and platelet counts, as well as attenuated splenomegaly in JAK2 V617F-positive mice. Notably, bone marrow fibrosis was not induced in JAK2 V617F-positive mice. Coexpression of SRSF2 P95H was also found to reduce the competitiveness of JAK2 V617F–mutated hematopoietic stem/progenitor cells.

Additionally, RBC, hemoglobin, and hematocrit levels were significantly reduced in the bone marrow of JAK2 V617F–positive mice that displayed enforced expression of S100A9. Mutated SRSF2 P95H decreased TGF-β levels and increased S100A8 and S100A9 expression in JAK2 V617F–positive mice.

“We demonstrated that SRSF2 P95H mutant reduces polycythemia and impairs competitiveness of JAK2 V617F–mutant hematopoietic stem/progenitor cells but does not promote the development of bone marrow fibrosis inJAK2 V617F-induced MPN,” lead study author Yue Yang, MD, of the Department of Biochemistry and Molecular Genetics at the University of Virginia School of Medicine in Charlottesville, and coinvestigators wrote.

To conduct their study, investigators created JAK2 V617F knock-in, SRSF2 P95H knock-in, and Mx1Cre transgenic mouse models, all on a C57BL/6 background. Intraperitoneal injection of 3 doses of polyinosine-polycytosine 300 μg were given at 4 weeks after birth in order to induce Mx1Cre expression. Wild-type C57BL/6 and UBC-GFP mice were acquired from an outside laboratory.

To create non-competitive bone marrow transplantation assays, 1 x 106 bone marrow cells were taken from the mice in each of the 4 groups (control of wild-type or Mx1Cre; SRSF2 P95H-positive; JAK2 V617F-positive; and SRSF2 P95H/JAK2 V617F-positive) and transplanted into lethally irradiated C57BL/6 mice. Polyinosine-polycytosine was administered to the recipient mice at a dose of 300 μg 3 times at 4 weeks following transplantation.

Competitive transplantation assays were created by mixing bone marrow cells from uninduced JAK2 V617F-positive/GFP-positive or SRSF2 P95H/JAK2 V617F-positive/GFP-positive were mixed with wild-type competitor bone marrow cells at a 1:1 ratio and transplanted into wild-type C57BL/6. Recipient mice received 3 doses of polyinosine-polycytosine 300 μg at 4 weeks post transplantation.

To create colony-forming assays, investigators plated 2 X 104 mouse bone marrow cells in cytokine-containing complete methylcellulose medium. After 1 week, burst forming units-erythroid and granulocyte-macrophage colony-forming units were tallied. Spleen cells at a quantity of 1 x 105 were plated in MethoCult M3234 medium without cytokine to detect epo-independent colony-forming units-erythroid.

Epo-independent colony-forming units-erythroid were stained with benzidine solution and counted 2 days afterwards. Colony-forming units-megakaryocytes were determined by plating 1 x 105 bone marrow cells in collagen-based MegaCult medium with Tpo, IL-3, IL-6, and IL-11. Colony-forming units-megakaryocytes were scored at day 8.

S100A8 or S100A9 overexpression’s effect on granulocyte-macrophage colony-forming units and burst forming units-erythroid formation of JAK2 V617F-positive bone marrow, cells lineage-negative cells were isolated from the bone marrow. Puromycin 2.5 μg/mL administered for 48 hours was used to select infected cells and 2.5 × 103 lineage-negative cells were plated in duplicates in cytokine-supplemented complete methylcellulose medium.

Study authors analyzed the mice models using flow cytometry and real-time quantitative PCR. Additionally, the TGF-β1 ELISA kit was used to determine TGF-β1 serum levels.

Further findings revealed that mice with heterozygous JAK2 V617F displayed polycythemia vera with increased white blood cell, neutrophil, platelet, RBC, hemoglobin, and hematocrit counts in peripheral blood compared with control mice. Those with heterozygous SRSF2 P95H experienced decreased hemoglobin with increased mean corpuscular volume vs the control group. SRSF2 P95H/JAK2 V617F-positive mice had significantly decreased white blood cell, neutrophil, platelet, RBC, hemoglobin, and hematocrit levels vs JAK2 V617F–positive mice. Concurrent expression of JAK2 V617F and SRSF2 P95H mutations resulted in higher mean corpuscular volume values and reduced spleen size and weight vs JAK2 V617F–positive mice, which displayed splenomegaly.

JAK2 V617F–positve mice bone marrow sections had hypercellularity with significant increase in erythroid precursors and megakaryocyte clusters compared with JAK2 V617F/SRSF2 P95H–positive mice, which had normal bone marrow cellularity and a reduction of erythroid precursors and megakaryocyte clusters. At 24 weeks, reticulin staining of bone marrow of SRSF2 P95H/JAK2 V617F–positive mice did not reveal fibrosis; bone marrow fibrosis was also not observed at 1 year following induction.

Together, JAK2 V617F and SRSF2 P95H mutations significantly reduced LSK, short- and long-term hematopoietic stem cell, and multipotent progenitor counts in the bone marrow of mice with both alterations. In comparison, mice with only JAK2 V617F mutations had increased frequencies and totals in terms of LSK, short- and long-term hematopoietic stem cells, and multipotent progenitors. The presence of both mutations also resulted in decreased frequency and total numbers of myeloid progenitors, common myeloid progenitors, granulocyte-macrophage progenitors, and megakaryocyte-erythroid progenitors in the bone marrow compared with mice with JAK2 V617F mutations alone.

Expression of an SRSF2 P95H mutation was also found to reduce the competitiveness of JAK2 V617F hematopoietic stem/progenitor cells. Mice that received of JAK2 V617F–positive bone marrow displayed significantly higher percentages of GFP-positive granulocyte, erythroid, megakaryocyte, B-lymphocyte, and T-lymphocyte cells in the peripheral blood compared with those that received SRSF2 P95H/ JAK2 V617F–positive bone marrow. Reduced percentages of the same hematopoietic stem/progenitor cells were observed in the bone marrow of mice that received SRSF2 P95H/ JAK2 V617F–positive bone marrow vs JAK2 V617F–positive bone marrow.

“Similar observations [to ours] have been made in a recent study by Willekens et al. Additional mutations or genetic abnormalities are required in association with SRSF2 P95H and JAK2 V617F mutations in the development of full-blown myelofibrosis,” the study investigators concluded.

Reference

Yang Y, Abbas S, Sayem MA, et al. SRSF2 mutation reduces polycythemia and impairs hematopoietic progenitor functions in JAK2V617F-driven myeloproliferative neoplasm. Blood Cancer J. 2023;13(1):171. doi:10.1038/s41408-023-00947-y

Read more

Managing blood cancer: Claire Harrison on ruxolitinib for polycythemia vera

Ruxolitinib is a Janus kinase 2 (JAK2) inhibitor with diverse indications covering conditions such as eczema, psoriasis, vitiligo and myelofibrosis.

Most recently, in the UK at least, it has been added to the treatment arsenal for the rare blood cancer polycythaemia vera (PV) after its recommendation in October 2023 by the National Institute for Health and Care Excellence (NICE).

Professor Claire Harrison, consultant haematologist, professor of myeloproliferative neoplasms at Guy’s and St. Thomas’ NHS Foundation Trust in London, UK, has dedicated much of the last decade to researching ruxolitinib for use in PV and was instrumental in securing NICE’s endorsement.

‘It’s [a good] example of following a scientific story from discovery of the mutation, development of the drug, testing it in a more severe but related condition (myelofibrosis) then putting it into second line for PV,’ she says.

Despite the approval, there’s still work to be done to achieve the next ambition of ruxolitinib being approved for first-line use in PV. And Professor Harrison is on hand to get the ball rolling.

Diagnosing and managing polycythaemia vera

Today, diagnosing PV is much easier than when Professor Harrison first started as a consultant in 2001.

Previously, the diagnosis was arrived at following a series of tests to exclude all other possible causes of a patient’s symptoms and abnormal blood count. But after the description of a JAK2 mutation which is present in 97–98% of patients with PV, the diagnosis can be arrived at much more quickly.

‘This particular mutation for which it’s very easy to test for. It’s a cheap test and, for the most part, if it’s present, the patient has either got PV or one of the family of conditions, or it has very low levels of mutations but is likely to change into that,’ Professor Harrison says.

In general, the lay perception of a cancer diagnosis is that it represents a death sentence, and it becomes difficult to assuage patients of this fear.

While PV is incurable and lowers life-expectancy, it is not usually life-threatening, although Professor Harrison says some patients do present with life-threatening blood clots.

‘It’s a cancer but some low-risk patients we just treat with aspirin and phlebotomy, so removing blood,’ she says. ‘So that’s quite tricky saying “you’ve got cancer, but all we’re going to give you is an aspirin and take a pint of blood off you”, and other patients we do give treatments to, but we have limited options.’

Moreover, while these treatments do provide a clinical benefit in some patients, they frequently fail to alleviate symptoms.

This has become abundantly clear from the findings of the ongoing prospective REVEAL study. This showed that patients with PV experience symptoms that affect their quality of life and lead to work productivity impairments with an overall negative impact on their lives.

‘So, 80% of patients will complain of fatigue,’ Professor Harrison says. ‘It’s PV, it’s not a nothing condition: 20% of patients have to give up work or reduce their working time, others do die of the condition and the average life-expectancy is probably 15 to 20 years.’

Professor Harrison also highlights poor awareness of PV. ‘What patients would say, probably, is that people don’t understand the condition, GPs don’t understand the condition and their employers don’t understand the condition. It takes up a lot of their time and it has a big burden on their quality of life.’

She hopes that greater awareness of the condition will make it easier for people to support and make adaptations for this patient group.

Addressing unmet needs in PV

There haven’t been any new treatments for PV in around 20 years, with hydroxyurea and interferon alpha having long been the two options.

But that all changed with the description of the mutation. Professor Harrison says that after first helping with diagnosis, these learnings aided the development of drugs that could target the downstream effects of that mutation, principally JAK inhibitors.

‘These were first tested and used in more aggressive conditions in the family such as myelofibrosis. But with the advent of these drugs and their use in PV, we have been able to show that we can address some of the other unmet needs for patients,’ she says.

‘I could comment on how disappointing it is that the UK is five years behind the rest of Europe in the approval of ruxolitinib for PV, but I would prefer to celebrate that it’s a really important milestone for patients that they have an alternative therapy.’

This is particularly important as resistance or intolerance to treatments can develop in some patients, and there are other side effects and contraindications that mean traditional treatments may not be suitable.

‘An important side effect of hydroxyurea, which is also a side effect of ruxolitinib, is skin cancer,’ says Professor Harrison. ‘That’s something that we need to manage very carefully. If a patient has a skin cancer on hydroxyurea, we will sometimes change the therapy.

‘Interferon does cause quite serious mood disturbance – sometimes suicidal ideation – so it can’t really be used in patients who’ve got a significant history of anxiety or depression.

‘Similarly, [hydroxyurea] can’t be used for the 20% of patients below the age of 40 who might want to conceive a child. But we have the option to alternate between the first-line therapies.’

Emerging benefits of ruxolitinib

One of the key studies that led to the approval of ruxolitinib for PV in the UK was MAJIC-PV. This phase II trial, for which Professor Harrison was the lead author, randomised patients to either ruxolitinib or best available care in those intolerant or resistant to hydroxyurea, which is the current standard care therapy.

What was clear from MAJIC-PV was the superiority of ruxolitinib, with 43% of patients achieving a complete response based on several haematological criteria compared with only 26% of those receiving current best practice care.

While ruxolitinib does not cure PV, the MAJIC-PV trial provided reassurance that over five years no new longer-term safety issues emerged, Professor Harrison notes.

The trial also uncovered several additional biological actions of the drug. During the study, researchers measured the amount of abnormal JAK2 present in patients. This enabled clinicians to determine whether treatments had any effect on the aberrant mutations that were present.

Surprisingly, in those assigned ruxolitinib, there was a reduction in the level of this mutation.

As such, the MAJIC-PV study hinted at a mutation-specific effect of the drug which hadn’t previously been observed. Furthermore, this reduction in the level of abnormal JAK was associated with an increased life expectancy and a reduction in PV-related complications for patients.

‘Interestingly, when we were using the drug to treat patients with myelofibrosis, colleagues in Italy were reporting that their patients who had myelofibrosis but had the autoimmune condition alopecia, the hair was coming back,’ Professor Harrison adds. This hints at the wider benefits of ruxolitinib as an anti-inflammatory drug which is being harnessed in for example the treatment of eczema.

Delving deeper, Professor Harrison also describes how in research by colleague Adam Mead ruxolitinib appeared to modify PV at the stem cell level using research tools enaling the analysis of mutations at a single cell level.

Ruxolitinib as a first-line option?

Despite MAJIC-PV showing that ruxolitinib reduced levels of the abnormal JAK mutations, the current NICE approval recommends that the drug is used second-line for patients who either become resistant to or intolerant of hydroxyurea.

Notwithstanding this restriction, Professor Harrison still feels that it is important for patients to have access to ruxolitinib as another treatment option, either because of contra-indications or adverse effects from the currently available drugs.

Another consideration is the issue of drug resistance. ‘All of the available drugs are generally effective for the majority of patients, but over time, around 20–25% of patients will become resistant to that drug,’ she explains.

As a result, relying on a single drug isn’t the most effective way of controlling a patient’s blood count over time.

Encouraged by the findings from MAJIC-PV, a further phase III open-label trial, MITHRIDATE, for which Professor Harrison is the chief investigator, is starting to enrol patients.

It is designed to compare ruxolitinib with either hydroxyurea or interferon alpha as first-line therapy for high-risk PV patients.

Ruxolitinib also has a powerful effect on disease related symptoms for example patients with PV experience pruritus (itching) which can be extremely disabling, and the drug has a big impact on this troublesome symptom.

Although it is too early to draw any conclusions, Professor Harrison is hopeful that the MITHRIDATE trial will demonstrate the advantages of using ruxolitinib as a first-line treatment option and perhaps offer further insight into the drug’s disease-modifying properties.

Future treatment developments

While the introduction of JAK inhibitors such as ruxolitinib are a welcome addition to a clinician’s arsenal in the treatment of PV, Professor Harrison believes that future treatments need to focus on the off-target effects of these drugs such as immune suppression.

Although the development of JAK mutation-specific therapies in PV would be an advantage, Professor Harrison is of the opinion that it is just as important to improve understanding of how and when to use ruxolitinib in patients with PV.

Alongside the potential development of mutation-specific drugs, there is increasing interest in immune-mediated therapy.

‘I think we’ve had this massive step forward with description of molecular markers and therapies targeting JAK. We’ll probably go to treating PV earlier, and treating with a disease-modifying therapy,’ she concludes.

But focusing on the latest developments, Professor Harrison believes the introduction of innovations such as ruxolitinib would not have been possible without the support of various charities such as MPN Voice and Blood Cancer UK, as well as various academic centres and companies such as Novartis.

Working collaboratively, it has been possible to clearly demonstrate that ruxolitinib can go a long way towards helping to relieve the symptom burden of patients living with polycythaemia vera and improve quality of life for this under-represented patient population.

Read more

Blood Cancer: AOP Health Announces New Findings in Patients With Polycythemia Vera to Be Presented at the American Society of Hematology (ASH) 65th Annual Meeting

December 7, 2023

SAN DIEGO–(BUSINESS WIRE)–AOP Orphan Pharmaceuticals GmbH (AOP Health), Vienna, Austria, announced the results of an analysis assessing the impact of an individually optimized dosing regimen of ropeginterferon alfa-2b on treatment response in patients with low-risk polycythaemia vera (PV)1 These new data show that some low-risk PV patients require and can tolerate high ropeginterferon alfa-2b doses, and that the optimal dose varies substantially between patients.

“The results of this analysis expand the depth of data and add the clinically relevant and important evidence which can support health care professionals in their treatment decisions”

The first author of the abstract, Professor Heinz Gisslinger from the Medical University of Vienna/Austria, and his research team conducted the present analysis in the cohort of low-risk PV patients from the large trial PROUD-PV and its extension CONTINUATION-PV. The goal was to examine the impact of various baseline characteristics such as body mass index as well as individually optimized dose levels of ropeginterferon alfa-2b on complete hematologic response (CHR), the state when blood cell counts have returned to normal, at 12, 24, and 72 months.1

“The results of this analysis expand the depth of data and add the clinically relevant and important evidence which can support health care professionals in their treatment decisions”, Gisslinger concludes.

Gisslinger H et al. Individualized dosing of ropeginterferon alfa-2b ensures optimal response in patients with low-risk polycythemia vera (PV). ASH 2023, Abstract #4563 (https://ash.confex.com/ash/2023/webprogram/Paper173499.html)

About Polycythaemia Vera
Polycythaemia Vera (PV) is a rare cancer of the blood-building stem cells in the bone marrow resulting in a chronic increase of red blood cells, white blood cells and platelets. This condition increases the risk for circulatory disorders such as thrombosis and embolism, its symptoms lead to a reduced quality of life and on the long run may progress to myelofibrosis or transform to leukemia. While the molecular mechanism underlying PV is still subject of intense research, current results point to blood-building stem cells in the bone marrow with a set of acquired mutations, the most important being a mutant form of JAK2 that make up the malignant clone.

Important PV treatment goals are to achieve healthy blood counts (hematocrit below 45%), improve quality of life and to slow or delay the progression of disease.

Read more

Diagnosis and Management of Cardiovascular Risk in Patients with Polycythemia Vera

November 22, 2023

Giulia Benevolo,1 Monia Marchetti,2 Remo Melchio,3 Eloise Beggiato,1 Chiara Sartori,4 Carlo Alberto Biolé,5 Davide Rapezzi,6 Benedetto Bruno,1,7 Alberto Milan8

1University Hematology Division, Città della Salute e della Scienza di Torino, Turin, Italy; 2Hematology and Transplant Unit, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy; 3Division of Internal Medicine, A.O. S. Croce E Carle, Cuneo, Italy; 4Cardiology, Azienda Ospedaliera SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy; 5SCDO Cardiology, AOU San Luigi Gonzaga Orbassano, Turin, Italy; 6Hematology Division A.O. S. Croce e Carle, Cuneo, Italy; 7Department of Molecular Biotechnolgies and Medical Sciences, University of Turin, Turin, Italy; 8Department of Medical Sciences, University of Turin, Città della Salute e della Scienza di Torino, Turin, Italy

Correspondence: Giulia Benevolo, University Hematology Division, Città della Salute e della Scienza di Torino, via Genova 3, Turin, 10126, Italy, Tel +39 011 633 4301, Fax +39 011 633 4187, Email gbenevolo@cittadellasalute.to.it

Abstract: Polycythemia vera (PV) is a myeloproliferative neoplasm characterized by aberrant myeloid lineage hematopoiesis with excessive red blood cell and pro-inflammatory cytokine production. Patients with PV present with a range of thrombotic and hemorrhagic symptoms that affect quality of life and reduce overall survival expectancy. Thrombotic events, transformation into acute myeloid leukemia, and myelofibrosis are largely responsible for the observed mortality. Treatment of PV is thus primarily focused on symptom control and survival extension through the prevention of thrombosis and leukemic transformation. Patients with PV frequently experience thrombotic events and have elevated cardiovascular risk, including hypertension, dyslipidemias, obesity, and smoking, all of which negatively affect survival. To reduce the risk of thrombotic complications, PV therapy should aim to normalize hemoglobin, hematocrit, and leukocytosis and, in addition, identify and modify cardiovascular risk factors. Herein, we review what is currently known about the associated cardiovascular risk and propose strategies for diagnosing and managing patients with PV.

Plain Language Summary: Patients with the myeloproliferative neoplasm (MPN) polycythemia vera (PV) are at increased risk of cardiovascular (CV) events, including stroke, heart attacks, and peripheral arterial disease. High blood pressure, smoking, and dyslipidemia are common in MPN and contribute to the increased cardiovascular risk. Effectively controlling cardiovascular risk factors in PV, along with appropriate hematological therapy such as direct-acting oral anticoagulants alone or in combination with aspirin, may improve the outcomes of patients with PV, but further research is needed.

Read more

Presentation at MPN Congress and ASH Annual Meeting Reinforce Clinical Role of ropeginterferon alfa-2b-njft

November 2, 2023

BURLINGTON, Mass., November 02, 2023–(BUSINESS WIRE)–PharmaEssentia USA Corporation, a subsidiary of PharmaEssentia Corporation (TPEx:6446), a global biopharmaceutical innovator based in Taiwan leveraging deep expertise and proven scientific principles to deliver new biologics in hematology and oncology, today announced that new abstracts on ropeginterferon alfa-2b-njft will be presented during the 15th International Congress on Myeloproliferative Neoplasms (MPN Congress) in Brooklyn, NY on November 2-3, 2023, and during the 65th American Society of Hematology (ASH) Annual Meeting in San Diego, CA on December 9-12, 2023.

Key highlights from the accepted abstracts include:

  • AI-based Discovery: Application of AI technology to identify a potentially important association between myeloproliferative neoplasms (MPNs) and neurodegenerative diseases that may reflect common disease mechanisms and shared targets, including inhibitory immunoreceptors. The analysis suggests that dysregulation of specific immune checkpoints may promote chronic inflammation and thrombosis in MPNs and targeting these pathways may represent a novel approach to restoring immune and vascular homeostasis in these diseases.
  • Patient Survey: A qualitative analysis of responses to a survey distributed to MPN patients in partnership with two MPN advocacy organizations was conducted to help understand the patient experience on ropeginterferon alfa-2b-njft. In the interim analysis, themes that emerged from MPN patient responses ranged from satisfaction of observed outcomes with ropeginterferon alfa-2b-njft, management of safety concerns and comments on the ease of the injection.
  • Clinical Trial in Progress: Study design details of the Phase 2b clinical study EXCEED-ET evaluating ropeginterferon alfa-2b-njft for the investigational treatment of adults with essential thrombocythemia (ET) in the U.S. and Canada will be shared.
  • Medical Chart Review: A description of the study details for a quantitative, retrospective review of medical charts to assess the longitudinal clinical and economic burden of illness in patients with polycythemia vera (PV).
  • Investigator-led Korean study: Interim results from an independent, single-arm, open-label, multicenter study showed that with ropeginterferon alfa-2b-njft therapy and an accelerated dose titration at 12 months, 63% of participants achieved a complete hematological response, 61% achieved molecular response, as well as an overall reduction in JAK2 allele burden. The treatment was well tolerated in evaluated patients with PV.

“PharmaEssentia strives to be an essential partner to the MPN community, and these findings are a testament to the breadth and depth of the current and planned clinical and real-world evidence supporting the safety and efficacy of ropeginterferon alfa-2b-njft as a therapeutic option,” said Raymond Urbanski, M.D., Ph.D., Senior Vice President and U.S. Head of Clinical Development and Medical Affairs at PharmaEssentia. “We believe these encouraging data will help healthcare providers advance important discussions around improving care and outcomes for people living with MPNs who continue to face challenges managing their rare blood cancers.”

MPN Congress Abstract Details

  • Causal AI dissection of RNAseq datasets pinpoints connections between MPNs and neurodegenerative diseases
    • Abstract 127 – Thursday, November 2, 2023, 5:15 – 7 PM ET
  • Sharing the treatment experience of ropeginterferon alfa-2b-njft: A qualitative analysis of patient responses
    • Abstract 141 – Thursday, November 2, 2023, 5:15 – 7 PM ET
  • The clinical and economic burden of illness in patients with polycythemia vera: A retrospective medical chart audit study
    • Abstract 133 – Thursday, November 2, 2023, 5:15 – 7 PM ET
  • EXCEED-ET: A single-arm multicenter study to assess the efficacy, safety, and tolerability of ropeginterferon alfa-2b-njft (P1101) in North American adults with essential thrombocythemia
    • Abstract 137 – Thursday, November 2, 2023, 5:15 – 7 PM ET
  • A single-arm, open-label, multicenter study to assess molecular response of P1101 therapy in patients with polycythemia vera and elevated hematocrit
    • Abstract 116 – Thursday, November 2, 2023, 5:15 – 7 PM ET

ASH Abstract Details

  • A single-arm, open-label, multicenter study to assess molecular response of P1101 therapy in patients with polycythemia vera and elevated hematocrit: results from 12-month core study (New Data)
    • Abstract 4575 – Monday, December 11, 2023, 6 – 8 PM PT

Follow PharmaEssentia USA on Twitter and LinkedIn for news and updates at the meetings.

About Polycythemia Vera (PV)

Polycythemia vera (PV) is a cancer originating from a disease-initiating stem cell in the bone marrow resulting in a chronic increase of red blood cells, white blood cells, and platelets. PV may result in cardiovascular complications such as thrombosis and embolism, and often transforms to secondary myelofibrosis or leukemia. While the molecular mechanism underlying PV is still subject of intense research, current results point to a set of acquired mutations, the most important being a mutant form of JAK2.1

About Essential Thrombocythemia (ET)

Essential thrombocythemia (ET) is a myeloproliferative neoplasm (MPN) characterized by an overproduction of platelets in the blood that results from a genetic mutation; data indicates a JAK2 gene mutation is present in approximately half of diagnosed patients. ET is estimated to affect up to 57 per 100,000 people in the U.S. The disease is most commonly diagnosed through routine blood work and is most common in people over the age of 50, with women 1.5 times more likely to be diagnosed than men. As a chronic, progressive disease, ET requires regular monitoring and appropriate treatment. Over time, the disease may progress into more deadly conditions such as myelofibrosis or acute leukemia.2,3

Read more

Statin Use and Outcomes With Polycythemia Vera or Essential Thrombocythemia

October 19, 2023

Vicki Moore, PhD

In patients with polycythemia vera (PV) or essential thrombocythemia (ET), researchers found that statin therapy was associated with possible benefits related to survival and thrombosis in a new study. The researchers reported their findings in the journal Cancer Medicine.

In this cohort analysis, the researchers evaluated data on statin use and outcomes for 4010 adults with PV or ET who were of age 66 through 99 years at diagnosis and who were identified through the Surveillance, Epidemiology, and End Results-Medicare database.

The researchers analyzed patients in 2 cohorts, based on using either propensity score matching (PSM) or inverse probability of treatment weighting (IPTW), to evaluate possible relationships between statin use and outcomes. Cox proportional hazards analyses were performed to evaluate outcomes related to survival and first incident thrombotic events. The median follow-up time was 3.92 years.

The study included 1809 patients with PV and 2201 patients with ET. Patients had a median age at diagnosis of 77 years in both the PV and ET subgroups. In the first year after being diagnosed with PV or ET, over half (55.8%) of the patients overall had used statins.

For patients with PV, with a median follow-up of 4.00 years, 35.0% of those who used statins had died whereas 43.0% of patients not using statins had died. Among patients with ET, at a median follow-up of 3.84 years, deaths were reported among 35.7% of those who used statins and in 40.9% of those who did not use statins. A sensitivity analysis suggested that survival differences with statin use were significant for patients who had not been receiving statin therapy prior to their PV or ET diagnosis.

Statin use also was associated with a lower risk of thrombosis across the overall study population. In the PSM cohort, the HR was 0.63 (95% CI, 0.51-0.78; P <.01) for this association, and in the IPTW cohort, the HR was 0.57 (95% CI, 0.49-0.66; P <.01). A lower risk of thrombosis with statin use was also observed in PV and ET subgroups.

“Overall, our study demonstrated that statins improved survival and decreased the incidence of thrombotic events in older patients with PV and ET,” the researchers wrote in their report.

Reference

Podoltsev NA, Wang R, Shallis RM, et al. Statin use, survival and incidence of thrombosis among older patients with polycythemia vera and essential thrombocythemia. Cancer Med. Published online September 13, 2023. doi:10.1002/cam4.6528

Read more