Basophilia at Diagnosis Associated With Worse Outcomes in Several MPNs

January 20, 2025

The significance of basophilia across essential thrombocythemia (ET), polycythemia vera (PV), and primary myelofibrosis (PMF) has not been previously characterized. This led Lisa Yuen, MD, and colleagues to conduct a retrospective study examining a broad cohort of myeloproliferative neoplasms (MPNs) to determine basophilia’s associations with clinical and molecular features and patient outcomes. The researchers found that a more aggressive disease phenotype and poorer clinical outcomes were associated with higher levels among patients with MPNs. The findings were reported in the American Journal of Hematology.

The investigators analyzed data from 195 patients who were diagnosed with an MPN between 2008 and 2019. All cases were classified according to the revised 4th edition World Health Organization classification.

The researchers defined basophilia as a relative or absolute increase in peripheral blood or bone marrow aspirate within 6 months of the patient’s first diagnostic biopsy.

Basophilia was present in 22% of patients. The investigators noted a lower incidence of basophilia in patients with ET and PV compared with patients with pre-fibrotic PMF, fibrotic PMF, post-ET MF, post-PV MF, or MPN-unclassifiable (8% vs 35%; P<0.001). Among the patients without basophilia at baseline, the researchers found that 12% subsequently developed basophilia within a median of 19.6 months after the initial MPN diagnosis. Patients with basophilia were also significantly older (P<0.001) and had higher white blood cell count (P<0.001) and reticulin grade (P<0.001), lower hemoglobin levels (P=0.01), and lower platelet counts (P<0.001) than patients without basophilia.

Basophilia was also associated with more frequent abnormal cytogenetics (P<0.001), higher mean total of mutations detected by next-generation sequencing (P<0.001), and more frequent JAK2 (P<0.001), SF3B1 (P=0.0083), and SRSF2 (P=0.0159) mutations, but less frequent calreticulin mutations (P=0.0018). At a median follow-up of 63 months, overall survival (OS) and leukemia-free survival (LFS) were significantly shorter in the basophilia group (54 months and 46 months, respectively; P<0.001). The median OS and LFS remained shorter in the basophilia group when patients with ET and PV were excluded (P=0.003 and P=0.002, respectively).

“These results suggest that basophilia could represent an additional prognostic marker in patients with MPNs, by highlighting patients who may have shorter survival and higher risk of progression to blast phase than would be predicted by current risk modeling,” the researchers concluded.

Read more

Impact of Recent Translational and Therapeutic Developments on Clinical Course of BCR::ABL1-Positive and -Negative Myeloproliferative Neoplasms

January 2025

Tariq I MughalJohn MascarenhasRaajit K RampalPrithviraj BoseThomas LionHelen AjufoAbdulraheem YacoubSoheil MeshinchiLucia MasarovaRuben MesaCatriona JamiesonTiziano BarbuiGiuseppe SaglioRichard A Van Etten

Abstract

Despite the study of BCR::ABL1-positive and -negative myeloproliferative neoplasms (MPNs) providing seminal insights into cancer biology, tumor evolution and precision oncology over the past half century, significant challenges remain. MPNs are clonal hematopoietic stem cell-derived neoplasms with heterogenous clinical phenotypes and a clonal architecture which impacts the often-complex underlying genetics and microenvironment. The major driving molecular abnormalities have been well characterized, but debate on their role as disease-initiating molecular lesions continues. The introduction of the ABL1 tyrosine kinase inhibitors have been extremely successful in the treatment of chronic myeloid leukemia with most patients having a near-normal life expectancy. Similar success has, however, not been achieved for BCR::ABL1-negative MPNs in terms of disease course modification and most patients remain incurable. In both disease categories, genomic instability seems to increase the risk of disease progression to accelerated/blast phase, which is resistant/refractory to conventional treatment and associated with a poor prognosis. To address some of these issues, the late John Goldman and Tariq Mughal founded a scientific and clinical platform in 2006, the Post-American Society of Hematology (ASH) MPN workshop, to appraise novel cancer biology, candidate therapeutic targets, treatments and other clinical challenges and pay tribute to all the many scientists and clinicians around the world instrumental to the progress made and continuing advances being made. This paper summarizes some of the recent data discussed at the 18th edition of the workshop and includes reference to some data presented or published after the workshop, including the 26th John Goldman CML conference.

Read more

N-acetylcysteine inhibits thrombosis in a murine model of myeloproliferative neoplasm

Brianna M Craver 1Gajalakshmi Ramanathan 1Summer Hoang 1Xinyue Chang 1Laura F Mendez Luque 1Stefan Brooks 1Hew Yeng Lai 1Angela G Fleischman

January 2020

Abstract

Thrombosis is a major cause of mortality in patients with myeloproliferative neoplasms (MPNs), though there is currently little to offer patients with MPN beyond aspirin and cytoreductive therapies such as hydroxyurea for primary prevention. Thrombogenesis in MPN involves multiple cellular mechanisms, including platelet activation and neutrophil-extracellular trap formation; therefore, an antithrombotic agent that targets one or more of these processes would be of therapeutic benefit in MPN. Here, we treated the JAK2V617F knockin mouse model of polycythemia vera with N-acetylcysteine (NAC), a sulfhydryl-containing compound with broad effects on glutathione replenishment, free radical scavenging, and reducing disulfide bonds, to investigate its antithrombotic effects in the context of MPN. Strikingly, NAC treatment extended the lifespan of JAK2V617F mice without impacting blood counts or splenomegaly. Using an acute pulmonary thrombosis model in vivo, we found that NAC reduced thrombus formation to a similar extent as the irreversible platelet inhibitor aspirin. In vitro analysis of platelet activation revealed that NAC reduced thrombin-induced platelet-leukocyte aggregate formation in JAK2V617F mice. Furthermore, NAC reduced neutrophil extracellular trap formation in primary human neutrophils from patients with MPN as well as healthy controls. These results provide evidence that N-acetylcysteine inhibits thrombosis in JAK2V617F mice and provide a pre-clinical rationale for investigating NAC as a therapeutic to reduce thrombotic risk in MPN.

Read more

 

 

In Charleston, West Virginia, Tracking MPN Symptoms Becomes a Priority

January 17, 2025

Author(s): Mary Caffrey

When patients with myeloproliferative neoplasms (MPNs) must travel 2 hours for appointments with a cancer care team, capturing all their symptoms to ensure proper treatment is vital.

So, when Charleston Area Medical Center (AMC) Vandalia Health, located in Charleston, West Virginia, signed on to be part of the Association of Cancer Care Centers (ACCC) MPN Quality Improvement Program, systematic and accurate recording of patients’ symptoms was a priority. In a 12-month period, the cancer program serves 78 patients with polycythemia vera, 111 patients with essential thrombocythemia, and 48 patients with primary myelofibrosis.

Charleston AMC serves a large area in southern West Virginia, including some heavily rural areas. Its team includes general medical oncologists and hematologists who treat all types of cancers, 5 patient navigators, and 2 financial navigators who deal with insurance coverage, transportation issues, and other barriers to access. There is 1 social worker as well as nurses and other team members. Complementing the team are outside partners who help Charleston AMC meet multiple social needs.

As with ACCC Quality Improvement Program participants from Perlmutter Cancer Center at NYU Langone Health and Kent Hospital in Rhode Island, the multidisciplinary team in Charleston West Virginia, first took part in a webinar on MPN patient management.1

The ACCC Quality Improvement program was supported by Incyte.

The Charleston AMC team learned about the MPN Symptom Assessment Form Total Symptom Score (MPN-SAF TSS),2 a validated MPN patient-reported outcome tool that is recommended in the National Comprehensive Cancer Network Guidelines. Patients complete the assessment when they arrive at their appointments, giving providers an overview of symptoms that can be tracked over time.

Read more

RSK1 is an exploitable dependency in myeloproliferative neoplasms and secondary acute myeloid leukemia

Published January 16, 2025

Tim Kong, Angelo B. A. Laranjeira, Christopher T. Letson, LaYow Yu, Shuyang Lin, Jared S. Fowles, Daniel A. C. Fisher, Sherwin Ng, Wei Yang, Fan He, Minyoung Youn, et al.

Abstract

Myeloid malignancies are heterogenous disorders characterized by distinct molecular drivers but share convergence of oncogenic signaling pathways and propagation by ripe pro-inflammatory niches. Here, we establish a comprehensive transcriptional atlas across the spectrum of myeloproliferative neoplasms (MPN) and secondary acute myeloid leukemia (sAML) through RNA-sequencing of 158 primary samples encompassing CD34+ hematopoietic stem/progenitor cells and CD14+ monocytes. Supported by mass cytometry (CyTOF) profiling, we reveal aberrant networks of PI3K/AKT/mTOR signalling and NFκB-mediated hyper-inflammation. Combining ATAC-Seq, CUT&Tag, RNA-seq, and CyTOF, we demonstrate that targeting of ribosomal protein S6 kinase A1 (RSK1) suppresses NFκB activation and diminishes pro-inflammatory mediators including tumor necrosis factor (TNF) associated with MPN disease severity and transformation. We further evaluate a therapeutic approach utilizing a first-in-class RSK inhibitor, PMD-026, currently in Phase 2 development for breast cancer, for use in myeloid malignancies. Treatment with PMD-026 suppressed disease burden across seven syngeneic and patient-derived xenograft leukemia mouse models spanning the spectrum of driver and disease-modifying mutations. These findings uncover a therapeutic avenue for a conserved dependency across MPN and sAML.

Read more

Baseline Basophilia Associated With Aggressive MPN and Poor Outcomes

A more aggressive disease phenotype and poorer clinical outcomes were associated with higher baseline basophil levels among patients with myeloproliferative neoplasms (MPNs), according to the results of a retrospective study published in the American Journal of Hematology.

In the study, researchers analyzed data from 195 patients who were diagnosed with an MPN between 2008 and 2019 at a single center. Cases of chronic myeloid leukemia, chronic eosinophilic leukemia, and chronic neutrophilic leukemia were excluded. Basophilia was defined as a relative or absolute increase in peripheral blood or bone marrow aspirate within 6 months of the patient’s first diagnostic biopsy.

Of the 195 patients, 40.5% had essential thrombocythemia (ET), 23.1% had overt fibrotic phase primary myelofibrosis (PMF), 10.8% had post-ET myelofibrosis (MF), 8.2% had pre-fibrotic PMF, 8.2% had MPN-unclassifiable (MPN-U), 7.2% had polycythemia vera (PV), and 2.1% had post-PV MF.

Basophilia were present among 22% of patients. The lowest level of basophilia was present among patients with ET and PV at 8% compared with 35% among patients with pre-PMF, F-PMF, post-ET MF, post-PV MF, or MPN-U (P <.0001). There were 9% of patients who demonstrated basophilia in the bone marrow, but not the blood.

Of the patients without basophilia at baseline, researchers found that 12% developed basophilia within a median of 19.6 months after diagnosis. Older age (P <.001), higher white blood cell count (P <.001), higher reticulin grade (P =.0007), lower hemoglobin levels (P =.01), and lower platelet counts (P <.001) were significantly associated with basophilia.

Read more

HMGA2 overexpression with specific chromosomal abnormalities predominate in CALR and ASXL1 mutated myelofibrosis

Shivani Handa, Christoph Schaniel, Joseph Tripodi, Daiva Ahire, Md. Babu Mia, Sophie Klingborg, Douglas Tremblay, Bridget K. Marcellino, Ronald Hoffman & Vesna Najfeld

December 23, 2024

Abstract

Although multiple genetic events are thought to play a role in promoting progression of the myeloproliferative neoplasms (MPN), the individual events that are associated with the development of more aggressive disease phenotypes remain poorly defined. Here, we report that novel genomic deletions at chromosome 12q14.3, as detected by a high-resolution array comparative genomic hybridization plus single nucleotide polymorphisms platform, occur in 11% of MPN patients with myelofibrosis (MF) and MPN-accelerated/blast phase (AP/BP) but was not detected in patients with polycythemia vera or essential thrombocythemia. These 12q14.3 deletions resulted in the loss of most of the non-coding region of exon 5 and MIRLET7 binding sites in the 3’UTR of the high mobility group AT hook 2 (HMGA2), which negatively regulate HMGA2 expression. These acquired 12q14.3 deletions were predominately detected in MF patients with CALR and ASXL1 co-mutations and led to a greater degree of HMGA2 transcript overexpression, independent of the presence of an ASXL1 mutation. Patients with 12q structural abnormalities involving HMGA2 exhibited a more aggressive clinical course, with a higher frequency of MPN-AP/BP evolution. These findings indicate that HMGA2 overexpression associated with genomic deletion of its 3’UTR region is a newly recognized genetic event that contributes to MPN progression.

Read more

 

Thrombosis Linked With Second Cancer Risk in MPNs

Among patients with myeloproliferative neoplasms (MPNs), arterial thrombosis incidence appears to raise the risk of second cancers (SCs) and consequently, mortality, according to an analysis published in Blood Cancer Journal. Inflammatory biomarkers in these diseases suggest a more aggressive disease etiology, the authors added.

In the case of polycythemia vera (PV) or essential thrombocythemia (ET), previous research suggested that thrombosis may heighten the risk of progression to secondary myelofibrosis, which has a high mortality rate. For this retrospective analysis of MPN-patient data, researchers aimed to determine the elements of thrombosis that promote this risk.

Overall, data were evaluated from 1545 patients with PV, 891 patients with ET, 180 patients who were pre-primary myelofibrosis (PMF), and 707 patients with PMF. The median follow-up periods in the PV, ET, pre-PMF, and PMF groups were 5.6 months, 5.6 months, 6.1 months, and 2.92 months, respectively; 19%, 12%, 15%, and 7% of patients had a thrombosis event.

Future therapies should focus on targeting the complex mechanisms involved in both atherogenesis and thrombogenesis…and anti-inflammatory drugs for primary and secondary prevention of thrombosis.

Analysis of the patient data showed that arterial, but not venous or splanchnic, thrombosis was linked with a greater risk of SCs (odds ratio [OR], 2.53; 95% CI, 2.4-5.17). A white blood cell count of at least 11 x 109/L appeared to trend toward a greater risk of SCs, but this link was not significant (OR, 1.27; 95% CI, 0.96-1.67); this was also true of a PMF vs ET diagnosis (OR, 2.54; 95% CI, 0.97-6.61).

Read more

In Myeloid Neoplasms, Next-Generation Sequencing Can Be Inappropriate—and Money-Wasting

December 12, 2024

Author(s): Gayle Turim Dickstein

Using more careful consideration when deciding whether or not to order next-generation sequencing (NGS) for a patient with an actual or suspected myeloid neoplasm (MN) could save institutions thousands of dollars annually without compromising care, according to a Yale School of Medicine team. They have created a set of criteria to determine the appropriateness of NGS testing for MN (MN-NGS), with the goal of maximizing actionable results. Writing in eJHaem, the team also noted that the results of NGS, when not clinically indicated but performed anyway, can foster fruitless investigative paths and amplify patient anxiety.1

Close-up illustration of a MN | image credit: sawaratch – stock.adobe.com

They noted that, indeed, MNs often harbor pathogenic mutations that go undetected by karyotyping and fluorescence in situ hybridization, and NGS is truly necessary for diagnosis, risk stratification, and therapy.2 Among the 6 situations that would, if present, warrant NGS (ie, approval criteria) would be clinical suspicion of new, relapsed, or worsening disease, and end-of-induction chemotherapy.

The 6 “cancellation criteria”—situations in which these investigators say NGS clearly should not be performed—include, first, having a suspicion of only nonmyeloid disease (ie, the diagnosis is a nonmyeloid disease, or there is no suspicion for acute myeloid leukemia [AML], myelodysplasia, myeloproliferative neoplasm [MPN], or another MN). Other situations are having no suspicion of progression of a known MN; no evidence for recurrence post-transplant; a diagnosis of chronic myeloid leukemia (CML) with no concern for AML; and cases using blood when a concurrent bone marrow NGS is being performed. The 6th and final criterion is that none of the above cancellation criteria have been met, but no approval criteria have been met either.

The actionable results that should emerge from NGS done for the proper reason include making a new MN diagnosis, characterizing a MN with baseline mutational status for follow-up purposes, and altering a patient’s treatment plan, noted the investigators.

Read more

Fedratinib Alleviates Symptoms, Reduces Spleen Volume in MDS, MPNs

Fedratinib can reduce symptoms and spleen volume in patients with myelodysplastic syndrome (MDS) or myeloproliferative neoplasms (MPNs), according to research presented at the ASH Annual Meeting 2024.

Researchers are evaluating fedratinib, a JAK2 inhibitor, in a phase 2 trial. The trial (NCT05177211) enrolled 25 patients with atypical chronic myeloid leukemia (n=6), chronic neutrophilic leukemia (n=5), MDS/MPN-unclassifiable (n=8), and MDS/MPN with ring sideroblasts and thrombocytosis (n=6).

At baseline, the median patient age was 68.8 (range, 39.9-84.7) years, and the median time from diagnosis to treatment was 7.1 months. Most patients had splenomegaly (83%), and the median MPN-Symptom Assessment Form Total Symptom Score was 21 (range, 1-73). Prior treatments included hydroxyurea (36%), ruxolitinib (20%), luspatercept (8%), and hypomethylating agents (12%). Patients had a median of 3 pathogenic mutations.

“Most of these patients had multiple mutations, and most had a signaling mutation, an epigenetic mutation, and a splicing mutation,” said study presenter Andrew Kuykendall, MD, of the Moffitt Cancer Center in Tampa, Florida.

The patients received fedratinib at a dose of 400 mg daily in 28-day cycles. They could continue on treatment as long as they had a clinical benefit. At last follow-up, 11 patients were still on study treatment.

The median duration of fedratinib treatment was 10.8 months, and 21 patients were evaluable for efficacy at 24 weeks. Three patients discontinued fedratinib prior to 24 weeks for reasons unrelated to toxicity or lack of efficacy (eg, cost) and were considered non-responders.

Read more