Metformin for MPN: teaching an old drug new tricks

August 27, 2024

In this issue of Blood Advances, Kristensen et al1 identified an association between metformin use and decreased risk of myeloproliferative neoplasms (MPNs). In this Danish population–based case-control study, 7% of patients with MPN (268 out 3816) had taken metformin compared with 8.2% of the matched general population (1573 out 19 080) without MPNs. Metformin use was associated with lower odds of developing MPNs, with a marked dose-response relationship by cumulative duration in years. Among individuals with long-term metformin use between 5 and 10 years, the adjusted odds ratio was 0.42 (95% confidence interval [CI], 0.29-0.61). This protective effect was observed across all age groups, sex, driver mutations (JAK2-V617F and CALR), and subtypes of classical Philadelphia-chromosome negative MPNs, though most pronounced with polycythemia vera (PV) and essential thrombocythemia (ET). To our knowledge, this study is among the first to examine and report the potential leukemia preventive-impact of metformin.

Philadelphia-negative MPNs comprise a group of chronic leukemias that stem from aberrant clonal expansion of mature myeloid cells. Clinical presentation varies widely across the spectrum of these diseases, but major causes of morbidity and mortality include arterial and venous thromboses, along with transformation to myelofibrosis and acute myeloid leukemia. The majority of MPNs harbor recurrent somatic mutations in JAK2CALR, or MPL genes, all of which result in the dysregulated activation of the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway. The subsequent derangement in immune homeostasis plays a key role in MPN pathogenesis. The mutant hematopoietic clones of MPNs not only thrive in, but also propagate a hyperinflammatory environment through the production of proinflammatory cytokines such as interleukin 6 and tumor necrosis factor alpha among others.2

Metformin is a synthetic derivative of galegine, a natural product of the plant Galega officinalis (goat’s rue or French lilac), with blood glucose-lowering activity that was first reported in 1957 by the French physician Jean Sterne.3 It is now the most commonly prescribed medication for type 2 diabetes mellitus (T2DM) worldwide. Several epidemiologic studies have revealed decreased solid cancer risk and related mortality among patients taking metformin, but this study augments the findings of a previous retrospective investigation, which reported significantly lower risk for developing hematologic malignancies among veterans taking metformin vs those taking sulfonylureas.4 Although the means by which metformin prevents MPNs require further examination to complement the data presented by Kristensen et al, metformin may attenuate leukemogenesis through downregulation of JAK/STAT signaling and subsequent reduction of the inflammatory cytokines that drive MPN. Notable anti-inflammatory mechanisms of metformin on JAK2 V617F-positive MPN cell lines include intracellular reactive oxygen species production and inhibition of downstream mTOR signaling via adenosine monophosphate-activated kinase (AMPK)-dependent pathways, and inhibition of mitochondrial activity and activation of a subfamily of protein tyrosine phosphatase PP2A via AMPK-independent pathways.5

Read more

A randomized, double-blind, placebo-controlled phase 3 study to assess efficacy and safety of ropeginterferon alfa-2b in patients with early/lower-risk primary myelofibrosis

Ghaith Abu-Zeinah, Albert Qin, Harinder Gill, Norio Komatsu, John Mascarenhas, Weichung Joe Shih, Oleh Zagrijtschuk, Toshiaki Sato, Kazuya Shimoda, Richard T. Silver & Ruben Mesa

Abstract

Primary myelofibrosis (PMF) is the most aggressive of the myeloproliferative neoplasms and patients require greater attention and likely require earlier therapeutic intervention. Currently approved treatment options are limited in their selective suppression of clonal proliferation resulting from driver- and coexisting gene mutations. Janus kinase inhibitors are approved for symptomatic patients with higher-risk PMF. Additionally, most ongoing clinical studies focus on patients with higher-risk disease and/or high rates of transfusion dependency. Optimal treatment of early/lower-risk PMF remains to be identified and needs randomized clinical trial evaluations. Pegylated interferon alfa is recommended for symptomatic lower-risk PMF patients based on phase 2 non-randomized studies and expert opinion. Ropeginterferon alfa-2b (ropeg) is a new-generation pegylated interferon-based therapy with favorable pharmacokinetics and safety profiles, requiring less frequent injections than prior formulations. This randomized, double-blind, placebo-controlled phase 3 trial will assess its efficacy and safety in patients with “early/lower-risk PMF”, defined as pre-fibrotic PMF or PMF at low or intermediate-1 risk according to Dynamic International Prognostic Scoring System-plus. Co-primary endpoints include clinically relevant complete hematologic response and symptom endpoint. Secondary endpoints include progression- or event-free survival, molecular response in driver or relevant coexisting gene mutations, bone marrow response, and safety. Disease progression and events are defined based on the International Working Group criteria and well-published reports. 150 eligible patients will be randomized in a 2:1 ratio to receive either ropeg or placebo. Blinded sample size re-estimation is designed. Ropeg will be administered subcutaneously with a tolerable, higher starting-dose regimen. The study will provide important data for the treatment of early/lower-risk PMF for which an anti-clonal, disease-modifying agent is highly needed.

Read more

Platelet proteomic profiling reveals potential mediators of immunothrombosis and proteostasis in myeloproliferative neoplasms

August 14, 2024

Myeloproliferative neoplasms (MPNs) are chronic bone marrow malignancies characterized by clonal proliferation of hematopoietic precursors and elevated cell counts in peripheral blood.1 Patients with MPN are at risk of progression to myelofibrosis or acute leukemia and experience a substantial burden of microvascular symptoms.2,3 However, thrombosis (both arterial and venous) represents the leading cause of morbidity and mortality for patients with polycythemia vera (PV) and essential thrombocythemia (ET).4-6

Translational studies have indicated that the platelet proteome influences pathways relating to immune response, inflammation, and malignancy.7,8 Thrombocytosis and platelet hyperactivity are hallmarks of MPN;9 however, platelet count in isolation is not predictive of clinical outcome, and conventional antiplatelet therapy does not fully mitigate thrombotic risk.10 A comprehensive picture of the MPN platelet molecular profile is lacking, and to date, no studies have evaluated the unbiased platelet proteome in a sizable clinical cohort of affected patients. Here, we performed untargeted quantitative profiling of the platelet proteome in a large (n = 140) cohort of patients with PV and ET.

Using standardized platelet isolation protocols (supplemental Methods), we prepared purified platelets from peripheral blood samples of patients with an established diagnosis of MPN (World Health Organization defined, n = 59 ET, n = 41 PV) and a cohort of healthy controls (n = 40) recruited across 2 sites: Hospital Papa Giovanni XXIII, Bergamo, Italy and Mater Misericordiae University Hospital, Dublin, Ireland. Pertinent clinical features are shown in Figure 1 (and listed in supplemental Table 1). Interpatient variability, including age, sex, and treatment, as well as experimental batch effects, were adjusted as confounding factors in downstream expression analyses (supplemental Methods). Focusing on the most prothrombotic subtypes of MPNs, we hypothesized that the platelet proteome differs in MPN, and its characterization would offer insights into the underlying pathobiology and possible mechanisms underlying the associated clinical complications.

Read more

Development of a natural language processing pipeline for assessment of cardiovascular risk in myeloproliferative neoplasms

August 8, 2024

Andrea DuminucoJoshua Au YeungRaj VaghelaSukhraj VirdeeClaire WoodleySusan AsirvathamNatalia Curto-GarciaPriya SriskandarajahJennifer O’SullivanHugues de Lavallade, et al.

A central feature of myeloproliferative neoplasms (MPN) is an increased risk of cardiovascular thrombotic complications, and this is the primary determinant for the introduction of cytoreductive therapy.1 The landmark ECLAP study in polycythemia vera (PV) patients, showed cardiovascular mortality accounted for 45% of all deaths, with a thrombosis incidence rate of 1.7/100 person/year and a cumulative incidence of 4.5% over a median follow-up of 2.8 years.2

Natural language processing (NLP) is a branch of machine learning involving computational interpretation and analysis of human language. CogStack (https://github.com/CogStack), is an open-source software ecosystem, that retrieves structured and unstructured components of electronic health records (EHR). The Medical Concept Annotation Toolkit (MedCAT), the NLP component of CogStack, structures clinical free text by disambiguating and capturing synonyms, acronyms, and contextual details, such as negation, subject, and grammatical tense, and mapping text to medical Systematized Nomenclature of Medicine–Clinical Terms (SNOMED-CT) concepts. This technique is known as “named entity recognition and linkage” (NER+L). MedCAT has previously been used and validated in many studies to structure EHR data across a range of medical specialties for auditing, observational studies, de-identifying patient records, operational insights, disease modeling, and prediction.38

We employed our NLP pipeline, Cogstack, and MedCAT, to determine the prevalence and impact of cardiovascular risk factors upon thrombotic events during follow-up. We used Cogstack to retrieve outpatient hematology clinic letters and hematology discharge letters. MedCAT was then used for NER+L of relevant clinical free-text to respective SNOMED-CT codes that were determined by two hematology specialists. The base MedCAT model was trained unsupervised on >18 million EHR documents, and this was further fine-tuned using a 80:20 train:test split with 600 clinician-annotated MPN-specific documents. Total SNOMED-CT code counts were aggregated and grouped by individual patient, a unique threshold count was then applied to “infer” presence of the respective SNOMED code. In this process, hematology specialists read through clinical documents and manually highlight correct words or phrases detected by MedCAT that correspond to the SNOMED concept of interest.

Read more

Efficacy of a JAK2/mTOR Inhibitor Combination in Controlling Acute Graft-vs-Host Disease

By The ASCO Post Staff
Posted: 8/8/2024

Adding a Janus kinase 2 (JAK2) inhibitor to standard immunosuppressive drugs may not improve prevention of acute graft-vs-host disease in patients with hematologic malignancies undergoing treatment with allogeneic hematopoietic cell transplantation, according to a recent study published by Pidala et al in Blood.

Background

Hematopoietic cell transplantation may offer a potential cure in patients with hematologic malignancies; however, between 10% and 20% of patients who receive stem cells from a donor through allogeneic hematopoietic cell transplantation develop acute graft-vs-host disease within the first 100 days following transplant. This condition occurs when a donor’s immune cells identify the patient’s cells as foreign and attack them. Apart from disease recurrence, graft-vs-host disease can be life threatening and greatly impact a patient’s quality of life posttransplant.

While JAK inhibition is often effective in treating graft-vs-host disease—the JAK1/2 inhibitor ruxolitinib is indicated for the treatment of refractory graft-vs-host disease—the researchers conducting the recent trial addressed whether JAK inhibitors could have a role in graft-vs-host disease prophylaxis. JAK2 inhibitors are capable of turning off the JAK2 gene—which promotes inflammation and contributes to the development of graft-vs-host disease.

“JAK inhibitors are active in treating [graft-vs-host disease] that does not respond to steroids,” explained senior study author Brian Betts, MD, Vice Chair of Strategic Initiatives for Transplant & Cellular Therapy at the Roswell Park Comprehensive Cancer Center. “But the question over the past 10 years has been whether JAK inhibition could prevent [graft-vs-host disease],” he added.

Read more

Cardiovascular Risk in Philadelphia-Negative Myeloproliferative Neoplasms: Mechanisms and Implications—A Narrative Review

by Samuel Bogdan TodorCristian IchimAdrian Boicean, and Romeo Gabriel Mihaila

Abstract

Myeloproliferative neoplasms (MPNs), encompassing disorders like polycythemia vera (PV), essential thrombocythemia (ET), and primary myelofibrosis (PMF), are characterized by clonal hematopoiesis without the Philadelphia chromosome. The JAK2 V617F mutation is prevalent in PV, ET, and PMF, while mutations in MPL and CALR also play significant roles. These conditions predispose patients to thrombotic events, with PMF exhibiting the lowest survival among MPNs. Chronic inflammation, driven by cytokine release from aberrant leukocytes and platelets, amplifies cardiovascular risk through various mechanisms, including atherosclerosis and vascular remodeling. Additionally, MPN-related complications like pulmonary hypertension and cardiac fibrosis contribute to cardiovascular morbidity and mortality. This review consolidates recent research on MPNs’ cardiovascular implications, emphasizing thrombotic risk, chronic inflammation, and vascular stiffness. Understanding these associations is crucial for developing targeted therapies and improving outcomes in MPN patients.

Underreporting of Patient-Reported Outcomes Seen in Blood Cancer Trials

The incorporation of patient-reported outcomes (PROs) in clinical trials provides a means for researchers to measure health-related quality of life (HRQOL) and patient-specific outcomes. When evaluating subjective symptoms, for example, patients are usually the most reliable source of feedback. Consequently, PROs are seen as the gold standard for assessing subjective symptoms.1

According to the authors, PROs are not often collected or reported in solid tumor trials, but less is known about RCTs focusing on blood cancers.2

Read more

Prospective Study to Evaluate Fedratinib Plus Nivolumab in Myelofibrosis

A single-arm, phase 2 study of fedratinib, a selective JAK2 inhibitor, plus nivolumab is planned for patients with myelofibrosis (MF) who had a suboptimal or no response to a JAK inhibitor was initiated, according to a report published in the Annals of Hematology.

“This study will provide new findings that may contribute to advancing the treatment landscape for MF patients with suboptimal responses and limited alternatives,” the researchers wrote in their report. Currently, 23 of 30 planned patients are enrolled in the study and recruitment is expected to be completed by December 2024.

The open-label FRACTION trial will treat patients with MF from 9 academic centers in Germany, who will receive 400 mg of fedratinib daily in 28-day cycles, followed by 240 mg of nivolumab every 2 weeks beginning in cycle 2. Treatment will be given until progressive disease, relapse, death, or study discontinuation.

This study will provide new findings that may contribute to advancing the treatment landscape for MF patients with suboptimal responses and limited alternatives.

The primary efficacy endpoints will be response rate within 12 treatment cycles and RCT independency. Secondary endpoints will include safety, incidence of leukemic transformation, clinical benefit, duration of response, progression-free survival, overall survival, and disease burden. Molecular analyses will also serve as exploratory endpoints for the study.

Patients with MF primary or secondary MF are eligible if they had a suboptimal or no response to a JAK inhibitor, which is defined by persistent symptoms, splenomegaly, cytopenia, or hyperproliferation. Patients who have received a prior immune checkpoint inhibitor or history of uncontrolled autoimmune disease are not eligible for the study.

Disclosures: This research was supported in part by Celgene/Bristol Myers Squibb. Please see the original reference for a full list of disclosures.

Read more

High Neutrophil Ratio as a Thrombosis Predictor in Myeloproliferative Neoplasms

July 24, 2024

The following is a summary of “Predictive significance of high neutrophil ratio for thrombosis in myeloproliferative neoplasms: JSH-MPN-R18 subanalysis,” published in the July 2024 issue of Hematology by Nagaharu et al.


A nationwide study was conducted to identify clinical features of thrombosis in patients with myeloproliferative neoplasms to inform risk-based treatment strategies.

Researchers conducted a retrospective study to determine which complete blood count (CBC) parameters predicted thrombosis in patients with myeloproliferative neoplasms.

They investigated patients from JSH-MPN-R18, focusing on essential thrombocythemia (ET; n = 1152) and polycythemia vera (PV; n = 456). Using Welch’s T-test, the CBC parameters were compared with those with or without thrombotic events. Statistical analyses were conducted with R software.

The results showed that 74 patients with ET experienced thrombotic events. In multivariate analysis, patients with ET and thrombosis had a slightly but significantly higher neutrophil ratio compared to those without thrombosis (P<0.05). The absolute neutrophil count (aNeu) emerged as a valuable predictor for thrombosis in patients with low-risk according to the revised International Prognostic Score of Thrombosis for ET. Among patients with PV, those with thrombosis had notably higher hematocrit and aNeu levels than those without thrombosis. Additionally, the neutrophil ratio was slightly but significantly higher in patients with ET with thrombosis, potentially reflecting a greater JAK2 V617F allelic frequency, though was not detailed in JSH-MPN-R18.

Readmore