Community Helped a Patient When Their MPN Treatment Was Delayed

Brielle Benyon

Thomas Silver discusses how a community of support helped a patient when his bone marrow transplant was delayed.

Undergoing treatment for a myeloproliferative neoplasm (MPN) can be extremely lonely, so it is beneficial to have a support system throughout the process, explained Thomas Silver, board president of the Cancer Research and Treatment Fund in New York.

Silver, who was also recognized at CURE®’s 11th annual MPN Heroes® recognition event, told the story of his friend, Nick, an MPN survivor who was preparing to undergo a bone marrow transplant — which is currently the only curative therapy for MPNs — before he caught a common cold and had to have the procedure pushed back.

“Well, a month in his life is a long time,” Silver said.

However, Nick, Silver and a group of others are a part of a community that was able to offer support and stay in touch with Nick, who eventually went on to receive the transplant and is doing well, according to Silver.

Transcript

Well, it can be fairly lonely. It’s a really long, hard task. I remember talking to Nick when he was going through his through his trials and his journey. One of the things that happened was he was all ready to go with his transplant. And just a week or two beforehand, he developed a common cold and imagine being ready to go. And then you develop a cold. And they say, “Well, now because you have cold, you’ve got to wait another month.”

Well, a month in his life is a long time. So, the fact that we were around, we were available to talk to him, Nick and I are part of a large fraternity house and we have a number of brothers that we continue to stay in touch with. You have a great time with all the time. Just help him keep his spirits up when he was waiting that extra month to be able to actually begin the process of getting better.

Read more

Management of Ruxolitinib in MF Allows for Continued Survival Benefit

Targeted Oncology Staff

In the second article of a 2-part series, Pankit Vachhani, MD, highlights the impact ruxolitinib has had, and continues, to have in treatment for patients with myelofibrosis and how physicians should manage this treatment for their patients.

CASE

  • A 68-year-old woman presented to her physician with symptoms of mild fatigue.
  • Her spleen was palpable 6-7 cm below the left costal margin​.
  • Medical history: No known comorbidities
  • Next-generation sequence testing: JAK2 V617F mutation​
  • Karyotype: 46XX​
  • Bone marrow biopsy: megakaryocyte proliferation and atypia with evidence of reticulin fibrosis​
  • Blood smear: leukoerythroblastosis​
  • Diagnosis: Primary myelofibrosis​ (MF)
  • Dynamic International Prognostic Scoring System: intermediate-1​
  • Mutation-enhanced International Prognostic Score System 70: intermediate risk
  • The patient was not interested in transplant​.
  • A decision was made to initiate ruxolitinib (Jakafi).

Targeted Oncology: For patients with MF, what were their symptom responses while on ruxolitinib (Jakafi)?

PANKIT VACHHANI, MD: The patient’s symptom responses were recorded using different Quality of Life questionnaires from COMFORT-II study [NCT00934544].Patients on ruxolitinib did better with overall quality of life or functioning, as well as in some individual categories like pain, fatigue, and dyspnea. We also had data from COMFORT-I [NCT00952289], which used a symptom assessment form, [that showed similar results].2

How did the survival data compare between these 2 trials?

COMFORT-I had an inherent crossover designed into it. After 6 months, or 24 weeks, patients who got placebo could cross over to ruxolitinib, and the majority did so.2 Despite that, [we saw] the overall survival [OS] favored the patients who were originally randomized to ruxolitinib vs those who were originally randomized to placebo. That HR of 0.69 [95% CI, 0.50-0.95; P = .025] tells the story, in this case, and the OS data from COMFORT-II had an HR of 0.48 [95% CI, 0.28-0.85; P = .009]….3 These data are all pointing towards a survival advantage for patients who go on ruxolitinib. It’s important to note that there are survival data, but it was not the primary end point; it was the secondary endpoint, and it has been studied elsewhere as well.

How did the duration of treatment with ruxolitinib impact results for these patients?

[With ruxolitinib, we all ask] when should we begin treatment. Well, this was studied indirectly. The patients who went on the COMFORT studies were pooled together, so all the patients who were on ruxolitinib between these 2 studies were pulled together and [patients given] placebo and best available therapy were pooled on the other side.4 In terms of OS, if ruxolitinib was begun within 12 months of their MF diagnosis, those patients did the best in terms of survival, compared with patients who began ruxolitinib a little bit later, [at least] more than a year after diagnose [odds ratio (OR), 2.08; 95% CI, 1.12-3.90]. That’s an important point, which is [suggesting that] maybe beginning ruxolitinib treatment early might be associated with a better survival outcome. Similarly, the spleen volume responses…were also better for those who begin ruxolitinib earlier rather than later [at 47.6% vs 32.9%, respectively (P = .06)], which is also important to keep in mind [when treating these patients].4

What are the major hematologic adverse events (AEs) physicians should be aware of when using ruxolitinib?

In both COMFORT-I and COMFORT-II, cytopenias were some of the more common AEs. Grade 3/4 anemia, for example, with ruxolitinib was seen in 45% and 42% [of patients] and thrombocytopenia in 13% and 8%, [respectively].1,2 These can be managed through either transfusion, a drug hold, or maybe dose reductions, but the point is that [these AEs are] ruxolitinib related, [possibly due to the] inhibition of the JAK-STAT pathway.

The concern that I’ve heard from many is if [the patient] starts off with anemia, are they not responding as well [to ruxolitinib] compared with those who don’t start off with anemia? Or just the fact that ruxolitinib can cause anemia, does it make the overall outcomes worse? The answer is that ruxolitinib will lead to comparable outcomes, whether [the patient] has anemia or not.2 The spleen responses will be comparable, similarly and the symptom responses will be comparable. The underlying point is that the efficacy is maintained in patients with new onset anemia. We must separate drug-induced anemia of ruxolitinib, with the bad effects of anemia from MF, as these are 2 different types.

References:

1. Harrison C, Kiladjian JJ, Al-Ali HK, et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med. 2012;366(9):787-98. doi:10.1056/NEJMoa1110556

2. Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366(9):799-807. doi:10.1056/NEJMoa1110557

3. Cervantes F, Vannucchi AM, Kiladjian JJ, et al; COMFORT-II investigators. Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood. 2013;122(25):4047-53. doi:10.1182/blood-2013-02-485888

4. Verstovsek S, Kiladjian JJ, Vannucchi A, et al. Early intervention in myelofibrosis and impact on outcomes: A pooled analysis of the COMFORT‐I and COMFORT‐II studies. Cancer. 2023;129:1681-1690. doi:10.1002/cncr.34707

Read more

AI Tool Accurately Differentiates MPNs Using Bone Marrow Biopsies

Jordyn Sava

Andrew Srisuwananukorn, MD, assistant professor at The Ohio State University Comprehensive Cancer Center, discussed the use of a novel artificial intelligence model that aids in the differentiating between prefibrotic primary myelofibrosis and essential thrombocythemia.

An artificial intelligence (AI) model is being investigated that will help clinicians distinguish between prefibrotic primary myelofibrosis (pre-PMF) and essential thrombocythemia (ET) using bone marrow biopsy images from 200 patients.

The AI tool had previously been trained with 32,000 pan-cancer biopsy images. It was also familiar with general pathologic features. From this, investigators tested if the AI could differentiate between the 2 types of myeloproliferative neoplasms (MPNs) in patients.

According to findings presented by Andrew Srisuwananukorn, MD, at the American Society of Hematology 2023 Meeting, the tool demonstrated a 92.3% rate of agreement with human experts, and the sensitivity and specificity for pre-PMF diagnosis was 66.6% and 100%, respectively.

Based on these promising findings, experts will continue to update the AI tool and test it in larger data sets.

“I view this type of tool as a companion diagnostic tool, but I do not believe [that] AI tools can replace the judgment of a human physician. I think it is really up to us as pathologists and clinicians to say when an AI algorithm tool is not working appropriately. What I hope is that this can be used for better information for the patient to understand their disease,” said Srisuwananukorn, assistant professor at The Ohio State University Comprehensive Cancer Center, in an interview with Targeted OncologyTM.

In the interview, Srisuwananukorn discussed the use of a novel AI model that aids in the differentiating between prefibrotic primary myelofibrosis and essential thrombocythemia.

Targeted Oncology: What can you tell us about this AI tool?

Srisuwananukorn: Our research is in developing an artificial intelligence tool to differentiate between rare myeloid malignancies, including prefibrotic myelofibrosis and essential thrombocythemia. As a brief overview, these diagnoses are challenging to differentiate because there’s similar criteria, including clinical and laboratory abnormalities, mutational profiling, and assessment of the bone marrow, which can be very subjective, particularly when looking at the megakaryocyte morphologies in the fibrosis. Our hope is to create a more objective or at least consistent tool using artificial intelligence to differentiate between the 2.

What is the motivation behind the development of this tool? What specific challenges or clinical needs there are in distinguishing between different disease types?

Our motivation is that this is a diagnostic dilemma for our patients. Essential thrombocythemia and prefibrotic primary myelofibrosis behave quite differently. The prefibrotic myelofibrosis cases have more symptoms and are at higher risk for progression to acute myeloid leukemia. To me, it behooves the physician to know exactly which disease they have. It’ll help guide their therapies in the future. Our hope is that a tool such as this can help guide that management and potentially to help enroll in clinical trials for more appropriate diagnosis and therapy creations.

In your study, how did AI demonstrate its efficacy when differentiating between the myelofibrosis and ET?

Our model had very high performance with [an] area under the receiver operator curve of 0.9, a sensitivity of 66.6% specificity of 100%, and an accuracy of 92.3% in diagnosing prefibrotic myelofibrosis. In addition, we did a qualitative analysis to try to understand what is being used in these AI algorithms to make those predictions. With this qualitative interpretation of quote unquote, opening the black box of our AI algorithms, we were able to see that preferentially, areas of bone marrow cellularity were chosen for the prediction of 1 vs the other disease. Reassuringly, the algorithm was not using nonsensical portions of the image, such as fat or cortical bone or even background artifacts. We believe this AI algorithm is using biological reasons.

What distinguishes this tool from others and how can it be interpreted in these settings moving forward?

I view this type of tool as a companion diagnostic tool, but I do not believe [that] AI tools can replace the judgment of a human physician. I think it is really up to us as pathologists and clinicians to say when an AI algorithm tool is not working appropriately. What I hope is that this can be used for better information for the patient to understand their disease.

Are there any particular patient subgroups that the AI tools showed notable effectiveness?

For right now, we’re only looking at 2 particular diseases: prefibrotic myelofibrosis and essential thrombocythemia. However, that does not mean that this is our only goal. These algorithms are agnostic of disease and outcome. Potentially, the same types of algorithms or workflows can be used for any type of disease that other clinicians might be able to implement in their clinical practice. Our particular goal is to do better for patients [with MPNs], and we have multiple ideas of how we can do that.

Moving forward, what are some of the next steps for this research?

Our next steps are in 2 domains. One, for this particular algorithm in differentiating pre-PMF and ET, we hope to validate it in further, larger retrospective cohorts at other academic centers and potentially within clinical trials that enroll patients [with ET]. Our thought is potentially, the ET trials did not have great performance because they were accidentally enrolling these pre-PMF cases. That’s our next step into rigorously seeing if this algorithm can be effective. Outside of that, we’re hoping that we can develop similar algorithms in other outcomes of interest, including risk stratification and therapy response.

What should oncologists know about the growing use of AI?

For physicians at large, what I want to reiterate is that it is important that we understand how it’s being used and when to use it. I do not believe that algorithms can supplant physician judgment, but I do think that they will be used in clinical practice and it’s up for us to know when it’s not working. I hope that it’s a support tool and will help guide your decision management but it’s always up to the physician.

REFERENCE
Srisuwananukorn A, Loscocco GG, Kuykendall AT, et al. Interpretable Artificial Intelligence (AI) Differentiates Prefibrotic Primary Myelofibrosis (prePMF) from Essential Thrombocythemia (ET): A Multi-Center Study of a New Clinical Decision Support Tool. Blood. 2023;142(suppl 1): 901.doi:10.1182/blood-2023-173877

Read more

MPN Outcomes Have Improved in the Last 20 Years

Brielle Benyon

Years ago, when Dr. Shamim Salman started treating patients with myeloproliferative neoplasms (MPNs), she, unfortunately saw many patients die from the disease because there was limited knowledge and treatment options.

However, outcomes are much better now with the advent of new therapies such as JAK inhibitors, Salman, hematologist-oncologist at Richmond University Medical Center in Staten Island, New York, said in an interview with CURE®.

“But now I’ve seen so many new things, so much research, so many new drugs and I’m so happy about it,” Salman, who was also honored at the 11th annual MPN Heroes® recognition event. “My patients are living longer. I feel so happy.”

Transcription

I have seen with my own eyes so many patients are dying with myeloproliferative neoplasms, especially I will mention myelofibrosis. I had so many patients (die because) at that time (when I started), there was no (knowledge of) JAK2, no JAK inhibitors, nothing. And patients would ask me, or their daughters or their sons (would ask), “Doctor, you will do something, right? You will make my mom better?” And I would say, “We will try.”

But now I’ve seen so many new things, so much research, so many new drugs and I’m so happy about it. My patients are living longer. I feel so happy. My patients 20 years ago, they died much sooner. They couldn’t see their granddaughter’s wedding or whatever their dreams were. Sometimes, I cried for them because I knew there’s nothing.

Not everybody was eligible for bone marrow transplant because some of them were elderly. Bone marrow transplant was there. I have sent my patient with myelofibrosis for bone marrow transplant. But unfortunately, if they were elderly, they died of the complications. So I tell my fellows, “You guys are so lucky now, so many drugs, so many research, so many things going on.”

And usually, my style is if I see a case, I tell them, go read about it today. And by the end of the day, again, we will talk and do look for clinical trials, look for the new research. I’m in Staten Island, but I’m connected with all the big institutions. And from the day one, we try to do the right thing for them.

Read more

How I Treat: Myeloproliferative Neoplasms in Pregnancy

Whilst MPN are traditionally considered diseases of adults in their sixth or seventh decade these conditions do occur in young patients for example for essential thrombocythaemia in particular there is a second peak in women of reproductive age. Pregnancy therefore is an uncommon but not rare occurrence and clinical challenge in some scenarios. Here we discuss in detail our local approach to the management of pregnancy in MPN patients taking a case-based approach. We take time to include relevant updates in the field and point to a future research strategy which should be internationally focused in order to obtain as much information in as short a time as possible.

What is Cytoreduction in MPNs?

Alex Biese

Dr. Douglas Tremblay of the Ichan School of Medicine explains the necessity of cytoreductive therapy for patients with MPNs such as essential thrombocythemia and polycythemia vera.

Cytoreduction is crucial for preventing serious complications for patients with essential thrombocythemia (ET) and polycythemia vera (PV) — both types of blood cancers that fall under the category of myeloproliferative neoplasms (MPNs), as Dr. Douglas Tremblay, explained in an interview during the 65th American Society of Hematology (ASH) Annual Meeting.

“Thrombosis is the leading cause of morbidity and mortality in patients with polycythemia vera and essential thrombocythemia, and these (cytoreductive therapy) medications are given in order to mitigate that risk,” said Tremblay, an assistant professor of medicine at the Icahn School of Medicine at Mount Sinai in New York City.

Tremblay, writing in Hematology, ASH Education Program, explained that approved treatments for cytoreduction among patients with ET and PV include hydroxyurea and long-acting interferons, plus anagrelide for patients with ET and Jakafi (ruxolitinib) for those with PV.

Tremblay discussed cytoreductive therapy, when it is best to start treatment and support services that are available for patients.

Q: Can you discuss what cytoreduction is and how it works for ET and PV?

A: Cytoreductive therapy refers to multiple medications which are given with the purpose of reducing blood counts in both essential thrombocythemia and polycythemia vera but are also given for the purpose of reducing thrombotic risk, which, thrombosis is the leading cause of morbidity and mortality in patients with polycythemia vera and essential thrombocythemia, and these medications are given to mitigate that risk.

Q: What factors influence the decision to recommend cytoreduction for these patients?

A: The discussion around when to start cytoreductive therapy around chronic MPN patients (with) PV and ET really has to do with risk stratification and the risk stratification scheme is available (and) largely used through the ELN risk classification, which dictates (that) high-risk patients older than an age of 60 or those who have a prior thrombosis. And, in ET, there are additional risk stratification schemas, including the IPSET-thrombosis (model) and the revised subset thrombosis score.

These can dictate which patients are at high risk for thrombosis and (who) then would benefit from cytoreductive therapy. But many of these different factors exist on a continuum such as age in particular, and what I highlighted in my talk (as ASH) is that there’s no difference in your risk stratification (on) the day that you turn 61 years old. It’s very important to understand someone’s biological age, including their cardiovascular risk factors, that can inform their overall risk of having a blood clot in these diseases.

Q: What support services can be made available for patients during and after cytoreductive treatment?

A: I think a lot of support services for patients with PV and ET during cytoreductive therapy really revolve around mitigating toxicities and trying to understand what some of these toxicities are. And I think it’s really helpful to partner with your physician to try to understand what sort of additional resources are available.

Some of these medications, (they) can also have a high financial cost too. There are support services available to help patients who are eligible to try to enjoy the benefits of some of these newer medications to improve their outcomes.

A lot of treatment of PV and ET really involves more of a holistic approach and not just focusing on the blood counts, but really focusing on other parameters that each patient has and trying to optimize those in order to ensure the best long-term outcomes.

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

Komrokji Analyzes Strategies to Address Anemia With Ruxolitinib in Myelofibrosis

Targeted Oncology Staff

During a Targeted Oncology™ Case-Based Roundtable™ event, Rami Komrokji, MD, discussed issues related to dosing and management of patients receiving ruxolitinib for myelofibrosis.

CASE SUMMARY

  • A 68-year-old woman presented to her physician with symptoms of mild fatigue, moderate night sweats, and abdominal pain/fullness lasting 4 months; she also reported increased bruising and an unexplained 12-lb weight loss.
  • Her spleen was palpable 8 cm below the left costal margin.
  • Karyotype: 46,XX
  • Bone marrow biopsy results: megakaryocyte proliferation and atypia with evidence of reticulin fibrosis
  • Genetic testing results: JAK2 V617F mutation; CALR mutation negative
  • A blood smear result revealed leukoerythroblastosis.
  • Laboratory values
    • Red blood cell count: 3.40 × 1012/L
    • Hemoglobin level: 13.2 g/dL
    • Hematocrit: 36%
    • Mean corpuscular volume: 94 fL
    • White blood cell count: 23.0 × 109/L
    • Platelet count: 450 × 109/L
    • Peripheral blood blasts: 1%
  • Diagnosis: primary myelofibrosis
  • Risk
    • International Prognostic Scoring System: intermediate-2
    • Mutation and Karyotype-Enhanced International Prognostic Scoring System for Primary Myelofibrosis in adults 70 years or younger: intermediate
  • The patient refused stem cell transplant and received ruxolitinib (Jakafi).

Targeted Oncology: What effect did the titrated dose of ruxolitinib have on spleen volume and total symptom score in the COMFORT-I trial (NCT00952289)?

KOMROKJI: The spleen volume [reduction] is dose dependent [Figure 11]. You will rarely see a meaningful spleen reduction at below 10 mg twice a day, and it takes 3 to 4 months. That becomes the challenge. For the patients who [have] borderline [cytopenia], the more you’re going to push those [patients] to get the spleen response, the more they are going to [develop cytopenia]. The symptom improvement can be seen daily at 5 mg or 10 mg twice a day and [comes] faster. You could achieve that with a lower [dosage] than the spleen response [Figure 21]. Ruxolitinib is typically given twice a day because it has a short half-life. Sometimes in real-life practice, especially if the patients [were prescribed] a certain [dosage] of the pill, until we get the other [dosage of the] pill, we sometimes do it once a day or we tell them to cut the pill and do it twice a day. But ideally, it should be done twice daily.

What hematologic toxicities were observed with ruxolitinib in the COMFORT-I and COMFORT-II (NCT00934544) trials?

This drug, in general, is safe. The major issues with ruxolitinib are anemia, thrombocytopenia, and less often neutropenia, and those are very predictable, particularly the anemia. It’s almost a 2 g/dL hemoglobin [level] drop by week 8. If you started somebody with hemoglobin [level] of 13 g/dL…[by] month 2, it will be 11 g/dL. But for many of those patients, their symptoms are better and their spleen [enlargement] is gone, so they don’t mind the 11-g/dL [hemoglobin level]. If somebody started with hemoglobin [level] of 9 g/dL or 8 g/dL and [it is] dropping to 7 g/dL, then they will probably need transfusions.

The only way to mitigate the cytopenia most of the time is dose reductions. Sometimes for anemia, we add ESAs [erythropoiesis-stimulating agents]. The COMFORT-I trial did not allow adding any ESAs. The COMFORT-II trial from Europe had a small subset who had ESA addition.2 Technically, ruxolitinib inhibits the JAK/STAT pathway where erythropoietin signals through, but because it has a short half-life, it’s a reversible inhibition. There is a window of time where the ESA can work. Sometimes we do that in practice to mitigate anemia.

There is no difference in the response based on [grade 3 or 4] anemia, so whether the patients [had anemia] or not, in terms of their spleen and symptoms, they improved.3 Sometimes we used to push the dose before having the newer JAK2 [Janus kinase 2] inhibitors, and I used to struggle with this. The University of Texas MD Anderson Cancer Center [in Houston] had a philosophy of starting the patients on the higher dose even if they [have] transfusion [dependence]; then you back off. We escalated the dosing a little more, but at least the spleen and symptom improvement, even in the setting of anemia, can be achievable. I struggle with rendering somebody [with] transfusion [dependence] especially now that we have other choices.

What else should physicians keep in mind when using ruxolitinib?

If someone gets admitted for pneumonia or surgery, especially if they are on higher dosing and [are] responding, those drugs cannot be stopped cold turkey, particularly ruxolitinib and probably fedratinib [Inrebic] as well, because patients will have rebound cytokines. And if they are sick with pneumonia, they will [risk experiencing] SIRS [systemic inflammatory response syndrome] or septic shock. Even if they are going to surgery, we try not to interrupt if they can take [medication] orally. If your patients are not able to take oral medications, sometimes we bridge them with steroids as they are admitted.

If the case patient’s baseline hemoglobin level of 13 g/dL reduced to below 8 g/dL, is that a sign of ruxolitinib failure or progression?

That’s not typical, but I’ve seen some patients who will get profound cytopenia [later], so whether it’s a red flag or just sensitivity, I don’t know. Sometimes it depends on the timing. If somebody was on ruxolitinib for…a couple of years and I see this anemia, that’s…a red flag [ for] if there is anything changing the disease. If it was the first few months of ruxolitinib, I would assume it’s more likely to be ruxolitinib related. I would try to back off the dosing, [do] the classical things in medicine, make sure that the patient does not bleed [in case] something else was going on. But it will probably be somebody who is more sensitive. With that degree of drop, if I cannot get a spleen response or constitution symptoms relieved with a lower dose than nowadays, sometimes we would consider shifting to other JAK2 inhibitors if it was ruxolitinib related and I could not manage the symptoms with a lower dose.

How do you address the increased risk of skin cancer with ruxolitinib?

There are not very conclusive data, but there are data all over the place that any patients [with myelofibrosis] are at higher risk of skin cancer. For both hydroxyurea [Hydrea] and ruxolitinib, [it’s] a little higher than in patients who were not on treatment.4,5 Practicing in Florida adds another risk factor [for our patients developing skin cancer]. We tell them to get a baseline [dermatology exam] when we are starting and then get surveillance every 6 months. We don’t have many alternatives for treatment because both hydroxyurea and ruxolitinib increase the risk. There is no literature to say it’s dose dependent. I’ve rarely stopped the treatment because most of the time, if you’re doing surveillance, you can detect those early, but I’ve had patients who had bad…squamous cell carcinoma that [required] resection…. I held the treatment and tapered down, not knowing what else to do, because it…was extensive. There is literature, but there is no guidance.

What is the lowest dose of ruxolitinib you would use before making a switch?

The dosing of ruxolitinib is dependent on what I’m trying to achieve. If I’m going for a spleen response, I want them at least to be on 10-mg dosing because I rarely see a spleen response below 10 mg twice daily. [With] symptom improvement and [if] they’re feeling better [with] 5 mg to 10 mg, I settle and may not push the dosing more.

How do you safely switch from ruxolitinib to pacritinib (Vonjo)?

In terms of shifting, there is not much guidance, but we always taper down the ruxolitinib. We don’t stop it. If we’re shifting to pacritinib, [physicians] do different things. We taper down the ruxolitinib and sometimes put them on a bridge of steroids. I’ve had some colleagues who overlapped them in the first couple of weeks. Pacritinib has a much longer half-life, so it takes longer to kick in.6 With ruxolitinib, you see symptom control in a week or 2; with pacritinib, it takes 3 to 4 weeks. The approach is not to stop immediately…. Tapering ruxolitinib is 100% agreed on, particularly if you are stopping because of cytopenias, [but if] the patients were responding, that rebound will be even worse than [for] somebody who’s [experiencing progression]. The alternative strategies are to put them on a steroid bridge or overlap them…for a week or two and then stop the ruxolitinib completely. There is not much guidance if somebody was on pacritinib if you’re bridging them to something else, [and the same applies] with fedratinib. Pacritinib has less or no JAK1 effect, so in my experience, stopping it immediately is more tolerable. You don’t have to taper it. But we do with fedratinib. There are not many data yet with momelotinib [Ojjaara].

For patients who do not respond to management of anemia and require transfusions every 2 weeks, would you be concerned about iron overload?

There is not much literature about iron overloading in myelofibrosis, but it’s [a] very similar story to MDS [myelodysplastic syndrome]. Myelofibrosis is an inflammatory condition, so patients could have a high baseline [level] of ferritin. To [identify] somebody [having] iron overload, you have to get to the threshold of 15 to 20 units of blood transfusions.

The responses to ESA are [fewer] than in MDS because those patients have a higher endogenous serum erythropoietin level; you rarely see responses. There have been studies looking at…combination ruxolitinib/danazol and ruxolitinib/thalidomide…with modest activity.7,8 There is an ongoing phase 3 trial [INDEPENDENCE; NCT04717414] looking at ruxolitinib vs ruxolitinib/luspatercept-aamt [Reblozyl] because in the phase 2 trial [ACE-536-MF-001; NCT03194542], it seemed…that luspatercept can work in myelofibrosis to help with ruxolitinib-induced anemia.9 If patients have lower-risk myelofibrosis or I estimate their survival to be in years and they [have] high transfusion burden, I may consider some iron chelation. The problem [is] that the decision has to be individualized because even in MDS, we do the same because those iron chelations come with their own adverse events and toxicities. The subcutaneous pump is not easy for patients, and the oral ones have renal insufficiency, etc. But in selected cases, you could consider it.

If a patient with baseline hemoglobin level of 8 g/dL receives ruxolitinib and develops transfusion dependence, at what point do you consider it to be ruxolitinib failure?

[With] momelotinib approved, it will probably start taking that area of patients who [have anemia] at baseline because momelotinib does have anemia response in almost 25% of patients.10 I would say [to wait] 3 to 4 months for the main thing you are [managing]. If you’re [managing] the spleen and constitutional symptoms [and] you don’t see the spleen response by 3 to 4 months, then that’s failure. The [time to consider it] intolerance to treatment if they develop profound cytopenias is 2 to 3 months as well. I look at…am I achieving the primary end point? Did I at least reduce the spleen [size] by 25%? Is the patient feeling better at 4 months? If yes, I would continue. The other part I look at is if I make the patient [have] transfusion [dependence], are they having severe cytopenias? Sometimes you can start making the call on that at 2 months.

What approaches to controlling anemia related to myelofibrosis are most promising?

Whatever you use for anemia, [approximately] 30% response is the best [we’ve achieved]. In my experience of patients who [have cytopenia] without any spleen or constitutional symptoms, the most success had been with thalidomide plus prednisone, [where] 1 of 4 patients would respond. But if there was a response, some of the responses were very durable. You do the steroids the first 3 months, you take them off, [and] then you continue to thalidomide. I’ve had patients on [JAK inhibition] for 7 or 8 years. With the newer JAK2 inhibitors, pacritinib and momelotinib, the story is changing a little bit, particularly if they have spleen and constitutional symptoms. Then you are [managing] both. We still have to see data on somebody who [has cytopenia] without any spleen or constitutional symptoms [and] how much they will benefit from something like momelotinib or pacritinib for anemia.

REFERENCES

1. Verstovsek S, Gotlib J, Gupta V, et al. Management of cytopenias in patients with myelofibrosis treated with ruxolitinib and effect of dose modifications on efficacy outcomes. Onco Targets Ther. 2013;7:13-21. doi:10.2147/OTT.S53348

2. McMullin MF, Harrison CN, Niederwieser D, et al. The use of erythropoiesis-stimulating agents with ruxolitinib in patients with myelofibrosis in COMFORT-II: an open-label, phase 3 study assessing efficacy and safety of ruxolitinib versus best available therapy in the treatment of myelofibrosis. Exp Hematol Oncol. 2015;4:26. doi:10.1186/s40164-015-0021-2

3. Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366(9):799-807. doi:10.1056/NEJMoa1110557

4. Lin JQ, Li SQ, Li S, et al. A 10-year retrospective cohort study of ruxolitinib and association with nonmelanoma skin cancer in patients with polycythemia vera and myelofibrosis. J Am Acad Dermatol. 2022;86(2):339-344. doi:10.1016/j.jaad.2021.10.004

5. Hydrea. Prescribing information. Bristol Myers Squibb; 2021. Accessed November 20, 2023. https://tinyurl.com/dt9v7jft

6. Mascarenhas J, Hoffman R, Talpaz M, et al. Pacritinib vs best available therapy, including ruxolitinib, in patients with myelofibrosis: a randomized clinical trial. JAMA Oncol. 2018;4(5):652-659. doi:10.1001/jamaoncol.2017.5818

7. Gowin K, Kosiorek H, Dueck A, et al. Multicenter phase 2 study of combination therapy with ruxolitinib and danazol in patients with myelofibrosis. Leuk Res. 2017;60:31-35. doi:10.1016/j.leukres.2017.06.005

8. Duan M, Zhou D. Improvement of the hematologic toxicities of ruxolitinib in patients with MPN-associated myelofibrosis using a combination of thalidomide, stanozolol and prednisone. Hematology. 2019;24(1):516-520. doi:10.1080/16078454.2019.1631509

9. Gerds AT, Harrison C, Kiladjian JJ, et al. Safety and efficacy of luspatercept for the treatment of anemia in patients with myelofibrosis: results from the ACE-536-MF-001 study. J Clin Oncol. 2023;41(suppl 16):7016. doi:10.1200/JCO.2023.41.16_suppl.7016

10. Gerds AT, Verstovsek S, Vannucchi AM, et al. Momelotinib versus danazol in symptomatic patients with anaemia and myelofibrosis previously treated with a JAK inhibitor (MOMENTUM): an updated analysis of an international, double-blind, randomised phase 3 study. Lancet Haematol. 2023;10(9):e735-e746. doi:10.1016/S2352-3026(23)00174-6

Read more

Preliminary data shows lowered cytokine levels in myelofibrosis with PIM1 kinase inhibitor

December 19, 2023

SAN DIEGO — Treatment with a selective PIM1 kinase inhibitor showed positive clinical reactions in a sample of 23 patients with relapsed/refractory myelofibrosis, according to preliminary data presented at ASH Annual Meeting.

TP-3654, an oral investigational highly selective PIM1 kinase inhibitor, is the subject of the phase 1/phase 2 study evaluating monotherapy in relapsed/refractory myelofibrosis, according to results published by a team led by Lindsay A.M. Rein, MD, a hematologic oncologist at Duke Cancer Center.

Elevated circulating cytokines have previously been noted as having a strong association with myelofibrosis, and preclinical studies evaluating TP-3654 as both a monotherapy and in tandem with ruxolitinib (Jakafi, Incyte) displayed lowered cytokine response and other responses, such as spleen reduction and bone marrow fibrosis reduction.

As such, the phase 1/phase 2 study further examined TP-3654 monotherapy in a cohort of patients with intermediate or high-risk myelofibrosis, and who had been previously treated with or were ineligible for JAK inhibitor treatment.

The 23 patients — who had a median age of 73 years and all but one of whom had previously received JAK inhibitor treatment — were monitored for cytokine changes over 12 weeks of TP-3654 monotherapy. “Broad reductions” in cytokine levels were observed in as little as 24 hours, and over the course of 12 weeks, patients with higher cytokine reduction levels were found to have greater reduction in symptoms.

Twelve of 13 eligible patients saw a reduction in their total symptom score, with seven having their score reduced by 50% or more. Treatment-related adverse events, such as nausea and vomiting, were observed in over 20% of patients.

Rein noted that enrollment is still ongoing.

Reference:

Rein LAM, et al. Phase 1/2 study of TP-3654, a selective PIM1 kinase inhibitor: Preliminary data showed clinical activity and cytokine reductions in relapsed/refractory myelofibrosis patients. Presented at: ASH Annual Meeting and Exposition; Dec. 9-12, 2023; San Diego.

Read more