Researchers Identify INCA033989 as a Potential Treatment for Myeloproliferative Neoplasms

By Alexandra Gerlach, Associate Editor

Data from a study published in Blood demonstrates the therapeutic potential of INCA033989 as the first targeted therapy for myeloproliferative neoplasms (MPNs) that does not interfere with normal blood cell production. Existing therapeutic options for MPNs are effective at symptom management but have high discontinuation rates due to resistance and inadequate drug tolerability. The development of INCA033989 opens pathways to more effective, targeted options with disease-modifying potential without any negative impact on surrounding blood cells.1

The development of INCA033989 has positive implications for the evolving treatment landscape of patients with MPNs. Image Credit: © Anna – stock.adobe.com

MPNs are a group of malignancies characterized by the overproduction of red and white blood cells and is an umbrella for 6 different disease types: myelofibrosis (MF), essential thrombocythemia, chronic myelogenous leukemia, polycythemia vera, chronic neutrophilic leukemia, and chronic eosinophilic leukemia. Calreticulin (CALR) mutations are responsible for disease development in 20% to 30% of patients with MPNs, which can be either insertions or deletions in exon 9 of CALR. The mutated CALRprotein (mutCALR) is responsible for the stable interaction with thrombopoietin receptors (TPO-R), which are crucial for controlling blood cell production.2,3

Janus kinase (JAK) inhibitors, such as ruxolitinib (Jakafi; Incyte Corp), are the recommended treatment options for patients with MF or other MPNs; however, they are associated with adverse effects (AEs), namely grade 3 or 4 anemia. INCA033989 is a high affinity, fully human immunoglobulin G1 selective monoclonal antibody targeting mutCALR-driven oncogenesis to suppress TPO-R signaling, thereby preventing the proliferation and progression of disease. According to data from the original study announcing the development of this agent, there was an observed synergism between INCA033989 and ruxolitinib which resulted in the inhibition of cell proliferation and indicated the ability of INCA033989 to enhance the efficacy of ruxolitinib.3,4

Read more

Korean Study Finds DOAC Use “Seems Effective” in Patients With MPNs

September 25, 2024

Author(s): Mary Caffrey

A study based on a decade’s worth of Korean insurance data found that use of direct oral anticoagulants (DOACs) to address atrial fibrillation and venous thromboembolism in patients with myeloproliferative neoplasms (MPNs) is effective, with acceptable bleeding risk.

Patients with Philadelphia chromosome–negative myeloproliferative neoplasms (MPNs) face an elevated risk of arterial and venous thrombosis, due to the increased production of mature myeloid blood cells caused by their condition.1 The increased morbidity and mortality caused by atrial fibrillation (AF) and venous thromboembolism (VTE) among patients with MPNs has led the American College of Cardiology and the American Heart Association, among others, to recommend direct oral anticoagulants (DOACs) to prevent blood clots and reduce the risk of major cardiovascular events in patients with MPNs.2

However, a group of authors from Korea, writing in Cancer Research and Treatment, note that the actual amount of evidence regarding the use of DOACs in patients with MPNs is limited. This week, they published a study based on a decade’s worth of Korean insurance data. Based on an analysis of records from 368 patients with MPNs, they concluded that use of DOACs in this population “seems effective with an acceptable bleeding risk.”3

The authors write that a prior study, with very limited data, found the 1-year cumulative incidence of thrombosis was 5.5% and bleeding was 12.3% among patients with MPNs taking DOACs.3 They note their study population involved patients who were somewhat older (average age, 74 years) and had a higher CHA2DS2-VASc score, which evaluates a patient’s risk based on the presence of congestive heart failure, hypertension, age, diabetes status, history of stroke or transient ischemic attack, and vascular disease; risk is doubled if the patient is 75 years or older.

The Korean study was based on data from the Health Insurance Review and Assessment Service, which has information on inpatient and outpatient care for 50 million Koreans. Investigators pulled patient data from the period of January 1, 2011, to January 1, 2021. The cohort of 368 patients had the following characteristics:3

Read more

Can Vaccines Be Developed for MPNs? Study Examines the Challenges

September 18, 2024

Author(s): Mary Caffrey

Researchers from Bulgaria conduct an analysis of the potential for therapeutic vaccines in by comparing testing results for patients from their country with an international data set.

Despite their status as myeloid malignancies, myeloproliferative neoplasms (MPNs) have drawn interest from researchers as candidates for therapeutic vaccines. Giroux et al drew attention in Science in 2022 by investigating MPNs with calreticulin (CALR) mutations, which lack T cells to target this antigen.1 Specifically, Giroux’s team pursued the major histocompatibility complex (MHC-1) allele frequences they observed and developed a heteroclitic peptide vaccine to activate T cells against tumors.

Now, a team from Bulgaria follows Giroux with a statistical approach, with results appearing in Frontiers in Immunology.2 The group first made comparisons between patients with MPNs and healthy controls within the homogenous population of Bulgaria before completing a meta-analysis involving patients and healthy controls from the 1000 Genomes Project, an international effort to collect human genome samples.3

To start, the team established that human leukocyte antigen class I (HLA-I) and class II (HLA-II) alleles alter how JAK2 V617F and CALR mutations create cancer cells in MPNs, but that the role of immune response in MPNs is not well known. Thus, the team sought to explore the role of HLA genes in MPNs with CALR mutations. They conducted analyses involving 42 patients with CALR mutations and 158 with JAK2 V6127F mutations, as well as 1083 healthy controls.2

As the authors explained, mutations in 3 genes drive all MPNs; they are JAK2, CALR, and MPL. “These mutations originate at the level of hematopoietic stem cells, but, depending on the intrinsic and extrinsic factors, can lead to differential skewing of hematopoiesis predominantly into one of the myeloid lineages presenting clinically with 1 of the 3 phenotypes,” which they noted are essential thrombocythemia, polycythemia vera, and primary myelofibrosis.2

Mutations may appear just as cancer cells form but also before symptoms appear, in a status called clonal hematopoiesis of indeterminate potential, or CHIP; it may take a long time for CHIP to convert to malignancy, and different mutations follow different paths.

Read more

Novel approach may eliminate survival disparity in HSCT, greatly expand access

September 17, 2024

Key takeaways:

  • Post-transplant cyclophosphamide prophylaxis reduced the OS disparity in matched vs. mismatched unrelated donor hematopoietic stem cell transplant.
  • The approach could expand access to HSCT.

Use of post-transplant cyclophosphamide prophylaxis to prevent graft-versus-host disease could greatly expand access to hematopoietic stem cell transplantation, according to results of a retrospective study.

An analysis of patients who received post-transplant cyclophosphamide (PTCy) showed no statistically significant difference in OS or GVHD-free RFS (GRFS) between patients with matched (8/8) or mismatched (7/8) unrelated donors.

The ability to find a suitable unrelated donor with a 7/8 HLA match is “much greater” than finding one with 8/8 HLA match, according to researcher Steven M. Devine, MD, chief medical officer at NMDP and senior scientific director at Center for International Blood and Marrow Transplant Research (CIBMTR), told Healio.

“For an African American patient, [chances] go from 30% to over 80%,” Devine said. “It’s even higher for Hispanic or Asian individuals — into the 90% range.

“If you can go even lower [to a 6/8 match or 5/8 match], you can pretty much find a volunteer unrelated donor for almost 100% of patients,” Devine added. “We are enabling a transplant for everyone, regardless of their ancestry.”

Access disparities

Allogeneic HSCT — used to treat multiple blood cancers and blood disorders — produces the best results when stem cells of a related or unrelated donor matches at 8/8 HLA markers at the HLA-A, -B, -C and -DRB1 genes, according to study background.

Only 30% of patients have siblings, who are HLA-identical matches and therefore could donate.

Non-Hispanic white individuals have a 79% likelihood of finding an unrelated matched donor in the NMDP registry. The rate is between 29% and 58% for people of other races and ethnicities.

“Historically, there’s been roughly a 10% lower chance of survival with each level of mismatch,” Devine said. “That’s why for years the focus has been on trying to find full matches for all patients.”

Cyclophosphamide, a chemotherapy drug used to treat a variety of solid tumors and hematologic cancers, has been repurposed for about 20 years to prevent GVHD after HSCT.

“It’s really revolutionized [stem cell transplant] because its use is associated with a much lower risk for both the acute and more chronic forms of GVHD,” Devine said. “It’s improved outcomes overall, and it’s allowed us to perform mismatched transplants both from related and unrelated donors. So, [for this study], we [wondered whether] those historical differences in outcomes between matched and mismatched transplant [are] as great as they were years ago now that we’re using PTCy.”

Read more

Managing Ruxolitinib-Associated Liver Toxicity in Patients With Myelofibrosis Requires a Nuanced Approach

William J. Hogan, MBBCh, MRCPI

Key Points:

  • Ruxolitinib plays an important role in the treatment of symptomatic myelofibrosis, but its associated toxicities and adverse withdrawal effects can pose a challenge.
  • Myelofibrosis is linked with hepatic dysfunction, and ruxolitinib—despite its associated risks of hepatic transaminitis—may limit inflammation and progressive fibrosis, with liver injury resolution reported after treatment.
  • Overall, management of liver injury related to ruxolitinib depends on the cause and severity.

 

Question: In patients with myelofibrosis, how do you manage liver toxicity from ruxolitinib use beside dose de-escalation?

Answer: Ruxolitinib is a small-molecule JAK1/2 inhibitor that has an established role in the treatment of patients with symptomatic myelofibrosis, hydroxyurea-resistant polycythemia vera, and acute and chronic graft-versus-host disease. It has a potent anti-inflammatory effect that can provide very useful palliation of constitutional symptoms and splenomegaly in patients with myelofibrosis via inhibition of the JAK 1 and 2 pathways, which are involved in the production of inflammatory cytokines and hematopoietic growth factors.1-3 Common toxicities include myelosuppression, hepatic transaminitis, diarrhea, fatigue, headache, and peripheral edema. In patients with advanced myelofibrosis or florid inflammatory states, sudden withdrawal can lead to a systemic inflammatory response syndrome that can precipitate cardiopulmonary decompensation in frail patients, especially those with tenuous cardiopulmonary function, and corticosteroid prophylaxis or treatment may be required.4

In a mouse model of liver injury using carbon tetrachloride, JAK1/2 expression was implicated in progression of liver fibrosis. Inhibition of JAK1/2 downregulates downstream signaling, reduces progression to fibrosis, and even accelerates fibrosis reversal by inhibiting proliferation, migration, and activation of hepatic stellate cells in vitro.5 Myelofibrosis is associated with hepatic dysfunction by several mechanisms, including infiltration by hematopoietic stem cells (ie, extramedullary hematopoiesis [EMH]), portal vein thrombosis, and obliterative portal venopathy. EMH may respond favorably to ruxolitinib, and liver injury resolution after ruxolitinib treatment has been reported in patients with severe liver compromise related to myelofibrosis.6 These findings suggest that the drug may be able to reduce the consequences of inflammation—limiting progressive fibrosis in some circumstances—and may be of durable benefit in selected patients. A retrospective review of patients with liver injury and underlying myeloproliferative disorders treated with ruxolitinib and evaluated by biopsy demonstrated a variety of etiologies7; however, it is not always easy to determine how much is related to the underlying disease versus drug effects based on the biopsy findings, and this distinction is predominantly a clinical decision.

Liver toxicity typically manifests as mild hepatic transaminitis and can be exacerbated by drug–drug interactions. The incidence of hepatic transaminitis has been reported to be between 25% and 50%. Typically, these elevations are mild and self-limited, with < 1.5% of patients having values > 5 times the upper limit of normal. The drug is metabolized in the liver predominantly via the CYP3A4 pathway, and liver injury may be the result of the production of a toxic intermediate. Drug–drug interactions, such as with azole antifungals, can increase the effective exposure and enhance the potential for toxicity. Because suppression of intracellular signaling impairs immune response, suppression of viral replication may be impaired, increasing the potential for reactivation of quiescent viruses. This has been reported with reactivation of hepatitis B, resulting in clinically meaningful liver injury in patients who are at risk. It appears that the risk of viral reactivation with transaminitis and hyperbilirubinemia is greater in patients with HBsAg positivity, but reactivation has also been reported in those with anti-HBc. Efficacy with entecavir has been reported in treating viral reactivation and prophylaxis, so a nucleoside analog such as entecavir or tenofovir should be considered in patients who are at high risk.8-10

Management of liver injury related to ruxolitinib depends on the cause and severity. Identifying contributing causes, such as drug–drug interactions and prior viral hepatitis exposure and excluding other hepatotoxins are the initial imperative. Prior viral exposure with reactivation should be treated or prophylaxed when applicable. For mild transaminase elevations without hyperbilirubinemia, monitoring or temporary dose reduction may be appropriate. In more significant liver injury, dose interruption may be necessary. Consideration of an alternative JAK inhibitor may occasionally be warranted, as there does not appear to be significant evidence to suggest cross-reactivity between ruxolitinib and other JAK inhibitors. Of note, a retrospective review of liver injury occurring in patients with myeloproliferative neoplasms receiving ruxolitinib suggested a variety of potential etiologies, including those unrelated to the drug itself (EMH), in addition to obstructive portal vein apathy and drug-induced liver injury.

Read more

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