Skip to main content
Erschienen in: Annals of Hematology 4/2023

Open Access 14.02.2023 | Review Article

How I manage anemia related to myelofibrosis and its treatment regimens

verfasst von: Srdan Verstovsek

Erschienen in: Annals of Hematology | Ausgabe 4/2023

Abstract

Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by mutations (most frequently in JAK2, CALR, or MPL), burdensome symptoms, splenomegaly, cytopenia, and shortened life expectancy. In addition to other clinical manifestations, patients with MF often develop anemia, which can either be directly related to MF pathogenesis or a result of MF treatment with Janus kinase (JAK) inhibitors, such as ruxolitinib and fedratinib. Although symptoms and clinical manifestations can be similar between the 2 anemia types, only MF-related anemia is prognostic of reduced survival. In this review, I detail treatment and patient management approaches for both types of anemia presentations and provide recommendations for the treatment of MF in the presence of anemia.
Hinweise

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Myelofibrosis (MF) is a myeloproliferative neoplasm (MPN) characterized by abnormal megakaryocyte proliferation, along with reticulin or collagen fibrosis [1]. Nearly all patients (≈90%) have activating mutations in either JAK2, CALR, or MPL, which cause abnormal signaling that promotes cell proliferation and survival, as well as activation of several inflammation pathways [26]. MF clinical manifestations typically include anemia, thrombocytopenia, splenomegaly, and hepatomegaly that when combined can lead to burdensome symptoms such as fatigue, abdominal discomfort, night sweats, bone pain, and pruritus that impact patients’ quality of life [7, 8]. In addition to these burdensome signs and symptoms, patients with MF have increased risk of thrombosis and increased risk of progression to acute leukemia, which both also contribute to reduced survival compared with healthy controls [9, 10].
Anemia, at times reaching severe levels (< 8 g/dL), can be present at MF diagnosis and worsen over time as disease progresses (MF-related anemia), or it can manifest as a result of MF treatment with Janus kinase (JAK) inhibitors (treatment-related anemia) [1115]. Although symptoms and clinical manifestations can be similar, only MF-related anemia is prognostic of reduced survival [1215]. This review provides guidance for managing patients with either type of anemia presentation.

Sample patient—part 1

A 68-year-old female patient presented with shortness of breath. During a physical examination, she was found to have an enlarged spleen of 7 cm below the costal margin and no other significant findings. She also reported fatigue, significant night sweating, and some weight loss. Laboratory results indicated hemoglobin (Hb) of 9.7 g/dL, a white blood cell (WBC) count of 22 × 109/L with 2% blasts, and a platelet count of 122 × 109/L. Lactate dehydrogenase and erythropoietin (EPO) were both elevated (1780 U/L and 35 mU/mL, respectively). Furthermore, a bone marrow biopsy was compatible with MF.

General treatment of MF

In my practice, we would first determine a prognosis of a patient by risk stratification (Fig. 1). There are several prognostic scoring systems in use, among which the Mutation and Karotype-Enhanced IPSS (MIPSS-70 + VERSION 2.0) is probably the most comprehensive, with additional options including Dynamic International Prognostic Scoring System (DIPSS)-Plus if molecular testing is not available, DIPSS if karyotyping is not available, and Myelofibrosis Secondary to PV and ET-Prognostic Model (MYSEC-PM) for secondary MF. We consider a patient to be at higher risk if their risk score corresponds to high, intermediate-2, or a score in the higher intermediate range, consistent with the National Comprehensive Cancer Network guidelines [16]; such patients are typically referred to a stem cell transplant specialist for consideration of transplant procedure.
For patients with symptomatic disease, either lower- or higher-risk MF, typical treatment choice is the oral selective JAK1/JAK2 inhibitor ruxolitinib [16]. Ruxolitinib was the first JAK inhibitor approved for the treatment of MF by the US Food and Drug Administration in 2011 [17], and treatment prolongs survival, reduces symptom burden, and reduces spleen volume, as demonstrated by multiple clinical trials [1820]. Risk of death, as assessed by the prognostic scoring systems mentioned above, does not guide our decision on prescribing medications to control symptoms. Our alternative choice to ruxolitinib is the JAK2 inhibitor fedratinib, approved in the US for patients with intermediate-2 or high-risk MF, which we typically use in the second-line setting [16, 21]. For cytoreduction of high WBC or platelet counts, which are sometimes seen in patients with lower-risk, early, or prefibrotic MF, peginterferon alfa-2a or hydroxyurea are used [16]. For higher-risk patients with MF with severe thrombocytopenia (platelets < 50 × 109/L), pacritinib, an oral selective JAK2 and interleukin-1 receptor-associated kinase (IRAK1) inhibitor, is my preferred first-line treatment option [16, 22, 23]. Although both fedratinib and pacritinib provide clinical benefits, longer follow-up studies are required to determine if either provides an overall survival benefit as seen with ruxolitinib [23, 24].

Treatment options for anemia resulting from MF

Mild to moderate anemia is often present at MF diagnosis and can worsen with disease progression. Importantly, unlike JAK inhibitor treatment–related anemia discussed later in this review, MF-related anemia is associated with reduced overall survival, so proper management is essential [14]. An overview of treatment recommendations for patients with MF-related anemia in my practice is presented in Fig. 1 [16]. Patients should first be evaluated for contributing factors, including MF-related factors and exacerbating causes not directly related to MF. MF-related causes include reduced erythropoiesis, splenomegaly, and inflammatory cytokines [12, 25]. Additionally, vitamin B6, an essential element of heme synthesis, may be deficient in patients with primary or secondary MF, leading to anemia [26]. Functional iron deficiency due to inflammation is also frequently observed in patients with MF [27, 28], in which pro-inflammatory cytokine signaling upregulates hepcidin that in turn promotes storage of iron and ultimately iron-restricted anemia [29]. Functional iron deficiency is identifiable by low transferrin saturation despite normal ferritin levels [27, 28], and these patients should be treated with intravenous iron [29]. In addition, new targeted therapies are in development to modulate hepcidin signaling, including the JAK1/JAK2 and type 1 kinase activin A receptor or activin receptor-like kinase-2 (ACVR1/ALK2) inhibitor momelotinib (discussed in greater detail in the “Treatment options for anemia resulting from JAK inhibition” section) [30, 31] and the ACVR1/ALK2 inhibitor INCB000928 [32]. Although seemingly very rare, cases of patients developing primary MF and autoimmune hemolytic anemia have been reported [33]. Exacerbating causes not directly related to MF include underlying occult or gastrointestinal bleeding and deficiencies in iron folate and vitamin B12, which can lead to megaloblastic anemia [12, 25, 34, 35]. Deficiencies in iron folate and vitamin B12 are not uncommon in elderly patient populations [36] such as the MF population and are reversible via dietary or vitamin supplementation [35]. For patients with contributing factors not related to MF, the underlying cause should be treated per appropriate guidelines, and patients should be treated normally for MF, regardless of anemia presence [16, 35].
Management of patients with MF-related anemia begins with blood transfusions, with subsequent evaluation for additional anemia treatments [16]. For patients with serum EPO < 500 mU/mL, erythropoiesis-stimulating agents (ESAs) are a viable option that offers clinical benefits [16]. Up to half of the patients in this population may achieve an anemia response with ESAs, and dose escalation should be considered to achieve full benefit [37]. Importantly, ESAs can be safely added to ruxolitinib to effectively improve anemia in some patients with MF [38]. Additional treatment options are available for patients with serum EPO ≥ 500 mU/mL. The erythroid maturation agent, luspatercept, has demonstrated anemia benefits in patients with MF and myelodysplastic syndrome/MPN with ring sideroblasts who carry the SF3B1 mutation [39, 40]. It is important to note that the studies that evaluated luspatercept in MF had small patient populations, and additional investigation is warranted to further evaluate safety and efficacy. Anabolic steroid medication such as danazol can also be used for the treatment of anemia in patients with MF [16, 25]. Danazol treatment has been associated with an anemia response in these patients, including those who are transfusion-dependent [41]. Immunomodulatory imide agents (IMiDs), such as thalidomide and lenalidomide, have also demonstrated an anemia benefit in patients with MF, including those who were transfusion-dependent [42, 43]. However, this benefit was not observed in patients with myeloid metaplasia with MF who received thalidomide [44] or those with MF treated with pomalidomide, another IMiD [45]. Importantly, various treatments can be combined with ongoing ruxolitinib treatment, although the coadministration of IMiDs with steroids is currently a topic of debate. The combination of ruxolitinib with prednisone, thalidomide, and danazol has been associated with an anemia benefit in patients with MF [46]. Similarly, luspatercept combined with ruxolitinib demonstrated transfusion independence in some patients with MF [39]. Details for studies of ruxolitinib in combination with other agents, including ongoing/exploratory trials, are shown in Table 1.
Table 1
Key clinical trials featuring ruxolitinib-based combination therapy in patients with MF and anemia
Agent
(class)
Study patients, N
Phase
(NCT#)
Main inclusion criteria
Anemia results
Safety overview
Danazol [52]
(androgen)
N = 14
2
(NCT01732445)
• Anemia (Hb < 10 g/dL)
• Age ≥ 18 years
• ECOG PS ≤ 2
• ANC ≥ 1 × 109/L
• PLT, ≥ 50 × 109/L
• 4/5 (80%) JAKi-naive patients had stable or increasing Hb
• 5/9 (56%) patients who had received JAKi had stable or increasing Hb
Hematologic grade ≥ 3 AEs:
71% (n = 10)
Nonhematologic grade ≥ 3 AEs:
14% (n = 2)
Luspatercept [39, 53]
(TGFβ superfamily receptor ligand trap)
N = 79
2
(NCT03194542)
• Anemia
• Age ≥ 18 years
• ECOG PS ≤ 2
• ANC ≥ 1 × 109/L
• PLT, ≥ 50 × 109/L
• Mean Hb increase ≥ 1.5 g/dL from BL: NTD + RUX = 8/14 (57%); NTD, no RUX = 3/20 (15%)
• RBC-TI ≥ 12 weeks during study: TD + RUX = 8/22 (36%); TD, no RUX = 4/21(19%)
• ≥ 50% reduction in RBC transfusion burden: TD + RUX = 10/22 (46%); TD, no RUX = 8/21 (38%)
TRAEs in ≥ 5% of patients:
• Hypertension, 13%
• Bone pain, 9%
• Diarrhea, 5%
• 10% discontinued because of drug-related toxicity
Sotatercept [54]
(TGFβ superfamily receptor ligand trap)
Sotatercept monotherapy: n = 24
RUX combination cohort: n = 9
2
(NCT01712308)
• Anemia (Hb < 10 g/dL)
• Age ≥ 18 years
• Sporadic RBC transfusions, or TD
• ORR = TI + Hb increase ≥ 1.5 g/dL from BL for ≥ 12 consecutive weeks without RBC transfusion
• 6/17 (35%) in sotatercept monotherapy cohort
TRAEs:
• Grade 2 bilateral lower limb pain, n = 2 (1 patient in each cohort)
• Hypertension, n = 1
RUX combination cohort: ≥ 6 months RUX with stable dose for ≥ 2 months
RUX combination cohort: ORR in 1/8 (12.5%)
 
Thalidomide [55]
(immunomodulatory agent)
N = 23 (n = 15 evaluated)
2
(NCT03069326)
• Age ≥ 18 years
• ECOG PS ≤ 2
• ANC ≥ 1 × 109/L
• PLT, ≥ 50 × 109/L
• Suboptimal response, or refractory to RUX single-agent therapy
• RUX treatment for ≥ 3 months, and stable dose for ≥ 4 weeks before enrollment
Cycle 3:
• Trend toward increase in Hb over time
Nonhematologic grade ≥ 3 AEs:
• Limb edema, diverticulitis, hypertension, syncope (n = 1 each)
• Thromboembolic event and grade 3 neutropenia, n = 1
Prednisone, thalidomide, and danazol [46]
N = 72 (n = 53 in combination therapy group)
Pilot study
(ChiCTR1900025219)
• Age ≥ 18 years
• SV ≥ 450 cm3
• Peripheral blood blasts < 10%
• ECOG PS ≤ 2
• DIPSS: int-1, int-2, or high-risk
• ANC ≥ 1 × 109/L
• PLT, ≥ 50 × 109/L
Combo therapy group vs RUX monotherapy:
• Anemia response: 56% vs 0%
• Hb increase ≥ 10 g/L: 66% vs 0%
• Hb increase ≥ 20 g/L: 38% vs 0%
Combo therapy group vs RUX monotherapy:
• New or worsening anemia: 21% vs 58%
• No grade 3/4 nonhematologic AEs occurred
Erythropoiesis-stimulating agents [38]
N = 59
Retrospective study (completed)
• Anemia (Hb < 10 g/dL)
• IPSS: int-2 or high (int-1 for some patients in compassionate use)
• Anemia response: 54%
• Median time to anemia response: 4 mo
• Mild nausea, no other thrombotic events or toxicities reported
AE, adverse event; ANC, absolute neutrophil count; bid, twice daily; BL, baseline; DIPSS, Dynamic International Prognostic Scoring System; ECOG PS, Eastern Cooperative Oncology Group Performance Status; Hb, hemoglobin; int, intermediate; MF, myelofibrosis; NTD, non-transfusion dependence; ORR, overall response rate; PLT, platelet count; PV, polycythemia vera; qd, once daily; qw, once weekly; RBC, red blood cell; RBC-TI, red blood cell transfusion independence; RUX, ruxolitinib; SV, spleen volume; TD, transfusion dependence; TI, transfusion independence; tid, 3 times daily; TRAE, treatment-related adverse event

MF treatment considerations in patients with MF-related anemia

In general, MF treatment is initiated as early as possible for symptomatic patients in my practice, as supported by clinical trial evidence. A pooled analysis of the COMFORT I/II trials suggested that earlier ruxolitinib initiation in patients with intermediate-2 or high-risk MF was associated with improved clinical outcomes including fewer anemia events [47]. In addition, a post hoc analysis of the phase 3 JUMP trial demonstrated that a lower IPSS score at treatment initiation was associated with better spleen response rates, suggesting that ruxolitinib treatment earlier in the disease course improves response [48]. This should be balanced by possible lead-time bias and the known relationship between lower MF disease stage and better spleen response in patients treated with ruxolitinib [49]. Nonetheless, treating MF as early as possible, before the onset of MF-related anemia, should improve outcomes, both because of the direct benefit of early intervention and indirectly due to potentially avoiding the negative outcomes associated with MF-related anemia itself.
For patients who develop MF-related anemia, anemia is not a driver for primary treatment choice and therefore is managed based on my practice’s standard MF treatment algorithm (Fig. 1). In particular, ruxolitinib is not contraindicated in patients with anemia [17]. In the COMFORT I/II trials, ruxolitinib was associated with prolonged survival in patients with MF compared with controls, regardless of baseline anemia status [14]. Regarding the choice of ruxolitinib dose, my practice follows in many patients the approach evaluated in the phase 2 REALISE trial, which established a novel ruxolitinib dosing strategy for patients with anemia based on a lower ruxolitinib starting dose (10 mg twice daily [bid] with up-titration as necessary based on platelet counts and efficacy; Fig. 2) [12]. REALISE demonstrated that patients with baseline anemia experienced improvements in spleen size and MF-related symptoms with ruxolitinib treatment, and median Hb levels remained stable throughout the study, with red blood cell (RBC) transfusion requirements decreasing or remaining stable [12].

Sample patient—Sect. 2

The patient was not interested in undergoing a hematopoietic stem cell transplant right at diagnosis and was prescribed ruxolitinib 15 mg bid, as recommended for platelet counts between 100 and 200 × 109/L. During follow-up 3 weeks after treatment initiation, the patient reported feeling better, eating more, and tolerating the treatment well. Upon examination, the spleen was smaller in size, at 2 cm below the costal margin. Laboratory results showed Hb of 8.7 g/dL, a platelet count of 67 × 109/L, and a WBC count of 14 × 109/L. Due to the decrease observed in platelet count, the patient was now prescribed a decreased dose of ruxolitinib, at 10 mg bid.
After one more month of therapy, the patient reported feeling much better than before ruxolitinib. The spleen size remained at 2 cm below the costal margin, and laboratory reports showed Hb at 7.5 g/dL, a platelet count of 82 × 109/L, and a WBC count of 17 × 109/L.

Treatment options for anemia resulting from JAK inhibition

Although MF itself can lead to anemia, JAK inhibition may also separately cause or exacerbate anemia, which often occurs early in treatment and gradually improves with long-term exposure [13, 14, 1719, 50]. In the COMFORT studies, the number of patients with grade 3 or 4 anemia was higher for ruxolitinib compared with placebo; however, the lowest Hb levels were observed at weeks 8 to 12 of treatment and recovered to near-baseline levels by week 24 [14, 18, 19]. Furthermore, the number of patients with grade 3 or 4 anemia decreased over 42 months of treatment, with no patients reporting new or worsening grade 3 or 4 anemia after month 42 of treatment [13]. Importantly, new or worsening postbaseline anemia did not affect survival probability during ruxolitinib treatment in the COMFORT I/II pooled analysis [14]. In fact, patients with postbaseline anemia who received ruxolitinib had a survival advantage compared with the overall control group [14]. Likewise, transfusion dependence did not affect the survival benefit observed with ruxolitinib treatment in the COMFORT studies [20]. Similar to observations with ruxolitinib, in the JAKARTA studies of fedratinib in MF, a decrease in Hb levels was observed for 12 to 16 weeks, with a partial recovery observed afterward in the 400-mg group [50]. Taken together, these findings demonstrate that treatment-induced anemia as a result of JAK inhibition can be temporary.
In general, management for treatment-related anemia follows the same pattern described above for MF-related anemia, where contributing factors should first be assessed and treated appropriately. In the absence of non-MF-related contributing factors, primary management includes RBC transfusion and potential addition of secondary anemia treatments (Fig. 3). If transfusions and secondary treatment options are insufficient or burdensome, JAK inhibitor dose reduction can be considered to help improve anemia [17, 21]. After recovery of anemia to acceptable levels, ruxolitinib should be continued at the given dose or with subsequent modifications if necessary. Complete blood counts should be monitored every 2 to 4 weeks until doses are stabilized [16]. I try to avoid interruptions in therapy with ruxolitinib, as it has been reported that patients may have a significant rebound in symptoms within 7 to 10 days upon sudden interruption of ruxolitinib [16].
The JAK1/JAK2 inhibitor momelotinib, currently under investigation for patients with MF and anemia, has potent inhibitory activity against ACVR1/ALK2 and may become a second-line treatment option for patients who have to eventually stop ruxolitinib due to excessive anemia [30, 31]. This mechanism of action includes suppression of aberrant activation of hepcidin transcription in the liver and thus may improve iron homeostasis, facilitating normalized Hb levels and a decrease in transfusion requirements [30, 31]. In the phase 3 MOMENTUM trial of momelotinib versus danazol in patients with intermediate or high-risk MF previously treated with a JAK inhibitor, momelotinib provided superior clinical benefit as assessed by Myelofibrosis Symptom Assessment Form Total Symptom Score (MFSAF TSS) response and spleen response rate, as well as noninferiority for transfusion independence rate [51].

Sample patient—Sect. 3

The patient continued ruxolitinib treatment and underwent a transfusion with packed RBCs. In addition, anemia medication was provided, including an ESA as serum EPO was < 500 mU/mL. Follow-up was scheduled for every 3 to 4 weeks. After 6 months of therapy, the patient’s Hb was 8.4 g/dL, the platelet count was 77 × 109/L, and the WBC count was 12 × 109/L. The spleen was no longer palpable, no transfusions were needed, and the patient reported no symptoms.

Conclusions

Patients with MF endure burdensome symptoms and coexisting conditions as a result of their disease. In particular, patients commonly develop anemia, which can either be secondary to the disease or a result of MF treatment, further complicating disease management. Although MF treatment with JAK inhibitors can exacerbate anemia, evidence suggests that this is typically temporary and, as in the case of ruxolitinib, does not reduce survival contrary to MF-related anemia. MF treatment should be initiated as early as possible for symptomatic patients, ideally before the onset of MF-related anemia, to maximize clinical benefit. For those patients with MF who develop anemia, careful patient management, including RBC transfusions, secondary anemia treatments, JAK inhibitor dose modifications, and monitoring, can improve anemia to prevent further disease complications and improve clinical outcomes.

Acknowledgements

This study was funded by Incyte Corporation (Wilmington, DE, USA). Writing assistance was provided by Nicole Farra, PhD, an employee of ICON (Blue Bell, PA, USA), and was funded by Incyte.

Declarations

Competing interests

SV received research support from AstraZeneca, Blueprints Medicines Corp., Celgene, CTI BioPharma Corp., Genentech, Gilead, Incyte, ItalPharma, Novartis, NS Pharma, PharmaEssentia, Promedior, Protagonist Therapeutics, Roche, and Sierra Oncology; and is a paid consultant for Celgene, Incyte, Novartis, and Sierra Oncology.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Literatur
2.
Zurück zum Zitat Klampfl T, Gisslinger H, Harutyunyan AS, Nivarthi H, Rumi E, Milosevic JD, Them NC, Berg T, Gisslinger B, Pietra D, Chen D, Vladimer GI, Bagienski K, Milanesi C, Casetti IC, Sant’Antonio E, Ferretti V, Elena C, Schischlik F, Cleary C, Six M, Schalling M, Schonegger A, Bock C, Malcovati L, Pascutto C, Superti-Furga G, Cazzola M, Kralovics R (2013) Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 369(25):2379–2390CrossRefPubMed Klampfl T, Gisslinger H, Harutyunyan AS, Nivarthi H, Rumi E, Milosevic JD, Them NC, Berg T, Gisslinger B, Pietra D, Chen D, Vladimer GI, Bagienski K, Milanesi C, Casetti IC, Sant’Antonio E, Ferretti V, Elena C, Schischlik F, Cleary C, Six M, Schalling M, Schonegger A, Bock C, Malcovati L, Pascutto C, Superti-Furga G, Cazzola M, Kralovics R (2013) Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 369(25):2379–2390CrossRefPubMed
11.
Zurück zum Zitat Badireddy M, Baradhi KM (2022) Chronic anemia. In: StatPearls. StatPearls Publishing, Treasure Island (FL) Badireddy M, Baradhi KM (2022) Chronic anemia. In: StatPearls. StatPearls Publishing, Treasure Island (FL)
12.
Zurück zum Zitat Cervantes F, Ross DM, Radinoff A, Palandri F, Myasnikov A, Vannucchi AM, Zachee P, Gisslinger H, Komatsu N, Foltz L, Mannelli F, Passamonti F, Gilotti G, Sadek I, Tiwari R, Zor E, Al-Ali HK (2021) Efficacy and safety of a novel dosing strategy for ruxolitinib in the treatment of patients with myelofibrosis and anemia: the REALISE phase 2 study. Leukemia 35(12):3455–3465. https://doi.org/10.1038/s41375-021-01261-xCrossRefPubMedCentralPubMed Cervantes F, Ross DM, Radinoff A, Palandri F, Myasnikov A, Vannucchi AM, Zachee P, Gisslinger H, Komatsu N, Foltz L, Mannelli F, Passamonti F, Gilotti G, Sadek I, Tiwari R, Zor E, Al-Ali HK (2021) Efficacy and safety of a novel dosing strategy for ruxolitinib in the treatment of patients with myelofibrosis and anemia: the REALISE phase 2 study. Leukemia 35(12):3455–3465. https://​doi.​org/​10.​1038/​s41375-021-01261-xCrossRefPubMedCentralPubMed
13.
Zurück zum Zitat Verstovsek S, Mesa RA, Gotlib J, Gupta V, DiPersio JF, Catalano JV, Deininger MW, Miller CB, Silver RT, Talpaz M, Winton EF, Harvey JH Jr, Arcasoy MO, Hexner EO, Lyons RM, Paquette R, Raza A, Jones M, Kornacki D, Sun K, Kantarjian H, Comfort-I investigators, (2017) Long-term treatment with ruxolitinib for patients with myelofibrosis: 5-year update from the randomized, double-blind, placebo-controlled, phase 3 COMFORT-I trial. J Hematol Oncol 10(1):55. https://doi.org/10.1186/s13045-017-0417-zCrossRefPubMedCentralPubMed Verstovsek S, Mesa RA, Gotlib J, Gupta V, DiPersio JF, Catalano JV, Deininger MW, Miller CB, Silver RT, Talpaz M, Winton EF, Harvey JH Jr, Arcasoy MO, Hexner EO, Lyons RM, Paquette R, Raza A, Jones M, Kornacki D, Sun K, Kantarjian H, Comfort-I investigators, (2017) Long-term treatment with ruxolitinib for patients with myelofibrosis: 5-year update from the randomized, double-blind, placebo-controlled, phase 3 COMFORT-I trial. J Hematol Oncol 10(1):55. https://​doi.​org/​10.​1186/​s13045-017-0417-zCrossRefPubMedCentralPubMed
15.
Zurück zum Zitat Cervantes F, Vannucchi AM, Kiladjian JJ, Al-Ali HK, Sirulnik A, Stalbovskaya V, McQuitty M, Hunter DS, Levy RS, Passamonti F, Barbui T, Barosi G, Harrison CN, Knoops L, Gisslinger H, COMFORT-II investigators (2013) Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood 122(25):4047–4053. https://doi.org/10.1182/blood-2013-02-485888CrossRefPubMed Cervantes F, Vannucchi AM, Kiladjian JJ, Al-Ali HK, Sirulnik A, Stalbovskaya V, McQuitty M, Hunter DS, Levy RS, Passamonti F, Barbui T, Barosi G, Harrison CN, Knoops L, Gisslinger H, COMFORT-II investigators (2013) Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood 122(25):4047–4053. https://​doi.​org/​10.​1182/​blood-2013-02-485888CrossRefPubMed
17.
Zurück zum Zitat JAKAFI® (ruxolitinib). Full Prescribing Information, Incyte Corporation, Wilmington, DE, USA, 2021 JAKAFI® (ruxolitinib). Full Prescribing Information, Incyte Corporation, Wilmington, DE, USA, 2021
18.
Zurück zum Zitat Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger M, Miller C, Silver RT, Talpaz M, Winton EF, Harvey JH Jr, Arcasoy MO, Hexner E, Lyons RM, Paquette R, Raza A, Vaddi K, Erickson-Viitanen S, Koumenis IL, Sun W, Sandor V, Kantarjian HM (2012) A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med 366(9):799–807. https://doi.org/10.1056/NEJMoa1110557CrossRefPubMedCentralPubMed Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, Catalano JV, Deininger M, Miller C, Silver RT, Talpaz M, Winton EF, Harvey JH Jr, Arcasoy MO, Hexner E, Lyons RM, Paquette R, Raza A, Vaddi K, Erickson-Viitanen S, Koumenis IL, Sun W, Sandor V, Kantarjian HM (2012) A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med 366(9):799–807. https://​doi.​org/​10.​1056/​NEJMoa1110557CrossRefPubMedCentralPubMed
19.
Zurück zum Zitat Harrison C, Kiladjian JJ, Al-Ali HK, Gisslinger H, Waltzman R, Stalbovskaya V, McQuitty M, Hunter DS, Levy R, Knoops L, Cervantes F, Vannucchi AM, Barbui T, Barosi G (2012) JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med 366(9):787–798. https://doi.org/10.1056/NEJMoa1110556CrossRefPubMed Harrison C, Kiladjian JJ, Al-Ali HK, Gisslinger H, Waltzman R, Stalbovskaya V, McQuitty M, Hunter DS, Levy R, Knoops L, Cervantes F, Vannucchi AM, Barbui T, Barosi G (2012) JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med 366(9):787–798. https://​doi.​org/​10.​1056/​NEJMoa1110556CrossRefPubMed
20.
Zurück zum Zitat Verstovsek S, Gotlib J, Mesa RA, Vannucchi AM, Kiladjian JJ, Cervantes F, Harrison CN, Paquette R, Sun W, Naim A, Langmuir P, Dong T, Gopalakrishna P, Gupta V (2017) Long-term survival in patients treated with ruxolitinib for myelofibrosis: COMFORT-I and -II pooled analyses. J Hematol Oncol 10(1):156. https://doi.org/10.1186/s13045-017-0527-7 Verstovsek S, Gotlib J, Mesa RA, Vannucchi AM, Kiladjian JJ, Cervantes F, Harrison CN, Paquette R, Sun W, Naim A, Langmuir P, Dong T, Gopalakrishna P, Gupta V (2017) Long-term survival in patients treated with ruxolitinib for myelofibrosis: COMFORT-I and -II pooled analyses. J Hematol Oncol 10(1):156. https://​doi.​org/​10.​1186/​s13045-017-0527-7
21.
Zurück zum Zitat Inrebic® (fedratinib). Full Prescribing Information, Celgene Corporation, Summit, NJ, USA, 2019 Inrebic® (fedratinib). Full Prescribing Information, Celgene Corporation, Summit, NJ, USA, 2019
22.
Zurück zum Zitat VONJO® (pacritinib). Full Prescribing Information, CTI BioPharma Corporation, Seattle, WA, 2022 VONJO® (pacritinib). Full Prescribing Information, CTI BioPharma Corporation, Seattle, WA, 2022
23.
Zurück zum Zitat Mascarenhas J, Hoffman R, Talpaz M, Gerds AT, Stein B, Gupta V, Szoke A, Drummond M, Pristupa A, Granston T, Daly R, Al-Fayoumi S, Callahan JA, Singer JW, Gotlib J, Jamieson C, Harrison C, Mesa R, Verstovsek S (2018) Pacritinib vs best available therapy, including ruxolitinib, in patients with myelofibrosis: a randomized clinical trial. JAMA Oncol 4(5):652–659. https://doi.org/10.1001/jamaoncol.2017.5818CrossRefPubMedCentralPubMed Mascarenhas J, Hoffman R, Talpaz M, Gerds AT, Stein B, Gupta V, Szoke A, Drummond M, Pristupa A, Granston T, Daly R, Al-Fayoumi S, Callahan JA, Singer JW, Gotlib J, Jamieson C, Harrison C, Mesa R, Verstovsek S (2018) Pacritinib vs best available therapy, including ruxolitinib, in patients with myelofibrosis: a randomized clinical trial. JAMA Oncol 4(5):652–659. https://​doi.​org/​10.​1001/​jamaoncol.​2017.​5818CrossRefPubMedCentralPubMed
26.
Zurück zum Zitat Yasuda H, Tsutsui M, Ando J, Inano T, Noguchi M, Yahata Y, Tanaka M, Tsukune Y, Masuda A, Shirane S, Misawa K, Gotoh A, Sato E, Aritaka N, Sekiguchi Y, Sugimoto K, Komatsu N (2019) Vitamin B6 deficiency is prevalent in primary and secondary myelofibrosis patients. Int J Hematol 110(5):543–549. https://doi.org/10.1007/s12185-019-02717-8CrossRefPubMed Yasuda H, Tsutsui M, Ando J, Inano T, Noguchi M, Yahata Y, Tanaka M, Tsukune Y, Masuda A, Shirane S, Misawa K, Gotoh A, Sato E, Aritaka N, Sekiguchi Y, Sugimoto K, Komatsu N (2019) Vitamin B6 deficiency is prevalent in primary and secondary myelofibrosis patients. Int J Hematol 110(5):543–549. https://​doi.​org/​10.​1007/​s12185-019-02717-8CrossRefPubMed
27.
Zurück zum Zitat Birgegard G, Samuelsson J, Ahlstrand E, Ejerblad E, Enevold C, Ghanima W, Hasselbalch H, Nielsen CH, Knutsen H, Pedersen OB, Sørensen A, Andreasson B (2019) Inflammatory functional iron deficiency common in myelofibrosis, contributes to anaemia and impairs quality of life. From the Nordic MPN Study Group. Eur J Haematol 102(3):235–240. https://doi.org/10.1111/ejh.13198CrossRefPubMed Birgegard G, Samuelsson J, Ahlstrand E, Ejerblad E, Enevold C, Ghanima W, Hasselbalch H, Nielsen CH, Knutsen H, Pedersen OB, Sørensen A, Andreasson B (2019) Inflammatory functional iron deficiency common in myelofibrosis, contributes to anaemia and impairs quality of life. From the Nordic MPN Study Group. Eur J Haematol 102(3):235–240. https://​doi.​org/​10.​1111/​ejh.​13198CrossRefPubMed
31.
Zurück zum Zitat Verstovsek S, Chen CC, Egyed M, Ellis M, Fox L, Goh YT, Gupta V, Harrison C, Kiladjian JJ, Lazaroiu MC, Mead A, McLornan D, McMullin MF, Oh ST, Perkins A, Platzbecker U, Scheid C, Vannucchi A, Yoon SS, Kowalski MM, Mesa RA (2021) MOMENTUM: momelotinib vs danazol in patients with myelofibrosis previously treated with JAKi who are symptomatic and anemic. Future Oncol 17(12):1449–1459. https://doi.org/10.2217/fon-2020-1048CrossRefPubMed Verstovsek S, Chen CC, Egyed M, Ellis M, Fox L, Goh YT, Gupta V, Harrison C, Kiladjian JJ, Lazaroiu MC, Mead A, McLornan D, McMullin MF, Oh ST, Perkins A, Platzbecker U, Scheid C, Vannucchi A, Yoon SS, Kowalski MM, Mesa RA (2021) MOMENTUM: momelotinib vs danazol in patients with myelofibrosis previously treated with JAKi who are symptomatic and anemic. Future Oncol 17(12):1449–1459. https://​doi.​org/​10.​2217/​fon-2020-1048CrossRefPubMed
32.
Zurück zum Zitat Mohan SR, Oh ST, Ali H, Hunter AM, Palandri F, Lamothe B, Cui Y, Seguy F, McBride A, Savona MR, Kiladjian J-J, Verstovsek S (2022) A phase 1/2 study of INCB000928 as monotherapy or combined with ruxolitinib (RUX) in patients (Pts) with anemia due to myelofibrosis (MF). Blood 140(suppl 1):3943–3944. https://doi.org/10.1182/blood-2022-169210CrossRef Mohan SR, Oh ST, Ali H, Hunter AM, Palandri F, Lamothe B, Cui Y, Seguy F, McBride A, Savona MR, Kiladjian J-J, Verstovsek S (2022) A phase 1/2 study of INCB000928 as monotherapy or combined with ruxolitinib (RUX) in patients (Pts) with anemia due to myelofibrosis (MF). Blood 140(suppl 1):3943–3944. https://​doi.​org/​10.​1182/​blood-2022-169210CrossRef
34.
Zurück zum Zitat Warner MJ, Kamran MT (2021) Iron deficiency anemia. StatPearls Publishing, Treasure Island, FL Warner MJ, Kamran MT (2021) Iron deficiency anemia. StatPearls Publishing, Treasure Island, FL
37.
Zurück zum Zitat Hernández-Boluda JC, Correa JG, García-Delgado R, Martínez-López J, Alvarez-Larrán A, Fox ML, García-Gutiérrez V, Pérez-Encinas M, Ferrer-Marín F, Mata-Vázquez MI, Raya JM, Estrada N, García S, Kerguelen A, Durán MA, Albors M, Cervantes F (2017) Predictive factors for anemia response to erythropoiesis-stimulating agents in myelofibrosis. Eur J Haematol 98(4):407–414. https://doi.org/10.1111/ejh.12846CrossRefPubMed Hernández-Boluda JC, Correa JG, García-Delgado R, Martínez-López J, Alvarez-Larrán A, Fox ML, García-Gutiérrez V, Pérez-Encinas M, Ferrer-Marín F, Mata-Vázquez MI, Raya JM, Estrada N, García S, Kerguelen A, Durán MA, Albors M, Cervantes F (2017) Predictive factors for anemia response to erythropoiesis-stimulating agents in myelofibrosis. Eur J Haematol 98(4):407–414. https://​doi.​org/​10.​1111/​ejh.​12846CrossRefPubMed
38.
Zurück zum Zitat Crisà E, Cilloni D, Elli EM, Martinelli V, Palumbo GA, Pugliese N, Beggiato E, Frairia C, Cerrano M, Lanzarone G, Marchetti M, Mezzabotta M, Boccadoro M, Ferrero D (2018) The use of erythropoiesis-stimulating agents is safe and effective in the management of anaemia in myelofibrosis patients treated with ruxolitinib. Br J Haematol 182(5):701–704. https://doi.org/10.1111/bjh.15450CrossRefPubMed Crisà E, Cilloni D, Elli EM, Martinelli V, Palumbo GA, Pugliese N, Beggiato E, Frairia C, Cerrano M, Lanzarone G, Marchetti M, Mezzabotta M, Boccadoro M, Ferrero D (2018) The use of erythropoiesis-stimulating agents is safe and effective in the management of anaemia in myelofibrosis patients treated with ruxolitinib. Br J Haematol 182(5):701–704. https://​doi.​org/​10.​1111/​bjh.​15450CrossRefPubMed
39.
Zurück zum Zitat Gerds AT, Vannucchi AM, Passamonti F, Kremyanskaya M, Gotlib JR, Palmer JM, McCaul K, Ribrag V, Mead AJ, Harrison CN, Mesa RA, Kiladjian J-J, Barosi G, Gale RP, Laadem A, Pariseau J, Gerike T, Zhang J, Linde PG, Reynolds JG, Verstovsek S (2019) A phase 2 study of luspatercept in patients with myelofibrosis-associated anemia. Blood 134(suppl 1):557. https://doi.org/10.1182/blood-2019-122546CrossRef Gerds AT, Vannucchi AM, Passamonti F, Kremyanskaya M, Gotlib JR, Palmer JM, McCaul K, Ribrag V, Mead AJ, Harrison CN, Mesa RA, Kiladjian J-J, Barosi G, Gale RP, Laadem A, Pariseau J, Gerike T, Zhang J, Linde PG, Reynolds JG, Verstovsek S (2019) A phase 2 study of luspatercept in patients with myelofibrosis-associated anemia. Blood 134(suppl 1):557. https://​doi.​org/​10.​1182/​blood-2019-122546CrossRef
40.
Zurück zum Zitat Komrokji RS, Platzbecker U, Fenaux P, Garcia-Manero G, Mufti GJ, Santini V, Diez-Campelo M, Finelli C, Jurcic JG, Greenberg PL, Sekeres MA, Zeidan AM, DeZern AE, Savona MR, Shetty JK, Ito R, Zhang G, Ha X, Sinsimer D, Backstrom JT, Verma A (2020) Efficacy and safety of luspatercept treatment in patients with myelodysplastic syndrome/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T): a retrospective analysis from the Medalist study. Blood 136(suppl 1):13–15. https://doi.org/10.1182/blood-2020-137232CrossRef Komrokji RS, Platzbecker U, Fenaux P, Garcia-Manero G, Mufti GJ, Santini V, Diez-Campelo M, Finelli C, Jurcic JG, Greenberg PL, Sekeres MA, Zeidan AM, DeZern AE, Savona MR, Shetty JK, Ito R, Zhang G, Ha X, Sinsimer D, Backstrom JT, Verma A (2020) Efficacy and safety of luspatercept treatment in patients with myelodysplastic syndrome/myeloproliferative neoplasm with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T): a retrospective analysis from the Medalist study. Blood 136(suppl 1):13–15. https://​doi.​org/​10.​1182/​blood-2020-137232CrossRef
43.
Zurück zum Zitat Marchetti M, Barosi G, Balestri F, Viarengo G, Gentili S, Barulli S, Demory JL, Ilariucci F, Volpe A, Bordessoule D, Grossi A, Le Bousse-Kerdiles MC, Caenazzo A, Pecci A, Falcone A, Broccia G, Bendotti C, Bauduer F, Buccisano F, Dupriez B (2004) Low-dose thalidomide ameliorates cytopenias and splenomegaly in myelofibrosis with myeloid metaplasia: a phase II trial. J Clin Oncol 22(3):424–431. https://doi.org/10.1200/JCO.2004.08.160CrossRefPubMed Marchetti M, Barosi G, Balestri F, Viarengo G, Gentili S, Barulli S, Demory JL, Ilariucci F, Volpe A, Bordessoule D, Grossi A, Le Bousse-Kerdiles MC, Caenazzo A, Pecci A, Falcone A, Broccia G, Bendotti C, Bauduer F, Buccisano F, Dupriez B (2004) Low-dose thalidomide ameliorates cytopenias and splenomegaly in myelofibrosis with myeloid metaplasia: a phase II trial. J Clin Oncol 22(3):424–431. https://​doi.​org/​10.​1200/​JCO.​2004.​08.​160CrossRefPubMed
44.
Zurück zum Zitat Abgrall JF, Guibaud I, Bastie JN, Flesch M, Rossi JF, Lacotte-Thierry L, Boyer F, Casassus P, Slama B, Berthou C, Rodon P, Leporrier M, Villemagne B, Himberlin C, Ghomari K, Larosa F, Rollot F, Dugay J, Allard C, Maigre M, Isnard F, Zerbib R, Cauvin JM, Groupe Ouest-Est Leucemies et Maladies du Sang (GOELAMS) (2006) Thalidomide versus placebo in myeloid metaplasia with myelofibrosis: a prospective, randomized, double-blind, multicenter study. Haematologica 91(8):1027–1032PubMed Abgrall JF, Guibaud I, Bastie JN, Flesch M, Rossi JF, Lacotte-Thierry L, Boyer F, Casassus P, Slama B, Berthou C, Rodon P, Leporrier M, Villemagne B, Himberlin C, Ghomari K, Larosa F, Rollot F, Dugay J, Allard C, Maigre M, Isnard F, Zerbib R, Cauvin JM, Groupe Ouest-Est Leucemies et Maladies du Sang (GOELAMS) (2006) Thalidomide versus placebo in myeloid metaplasia with myelofibrosis: a prospective, randomized, double-blind, multicenter study. Haematologica 91(8):1027–1032PubMed
45.
Zurück zum Zitat Tefferi A, Al-Ali HK, Barosi G, Devos T, Gisslinger H, Jiang Q, Kiladjian JJ, Mesa R, Passamonti F, McMullin MF, Ribrag V, Schiller G, Vannucchi AM, Zhou D, Reiser D, Zhong J, Gale RP (2017) A randomized study of pomalidomide vs placebo in persons with myeloproliferative neoplasm-associated myelofibrosis and RBC-transfusion dependence. Leukemia 31(5):1252. https://doi.org/10.1038/leu.2017.2CrossRefPubMedCentralPubMed Tefferi A, Al-Ali HK, Barosi G, Devos T, Gisslinger H, Jiang Q, Kiladjian JJ, Mesa R, Passamonti F, McMullin MF, Ribrag V, Schiller G, Vannucchi AM, Zhou D, Reiser D, Zhong J, Gale RP (2017) A randomized study of pomalidomide vs placebo in persons with myeloproliferative neoplasm-associated myelofibrosis and RBC-transfusion dependence. Leukemia 31(5):1252. https://​doi.​org/​10.​1038/​leu.​2017.​2CrossRefPubMedCentralPubMed
46.
48.
49.
Zurück zum Zitat Palandri F, Palumbo GA, Bonifacio M, Tiribelli M, Benevolo G, Martino B, Abruzzese E, D’Adda M, Polverelli N, Bergamaschi M, Tieghi A, Cavazzini F, Ibatici A, Crugnola M, Bosi C, Latagliata R, Di Veroli A, Scaffidi L, de Marchi F, Cerqui E, Anaclerico B, De Matteis G, Spinsanti M, Sabattini E, Catani L, Aversa F, Di Raimondo F, Vitolo U, Lemoli RM, Fanin R, Merli F, Russo D, Cuneo A, Bacchi Reggiani ML, Cavo M, Vianelli N, Breccia M (2017) Baseline factors associated with response to ruxolitinib: an independent study on 408 patients with myelofibrosis. Oncotarget 8(45):79073–79086. https://doi.org/10.18632/oncotarget.18674CrossRefPubMedCentralPubMed Palandri F, Palumbo GA, Bonifacio M, Tiribelli M, Benevolo G, Martino B, Abruzzese E, D’Adda M, Polverelli N, Bergamaschi M, Tieghi A, Cavazzini F, Ibatici A, Crugnola M, Bosi C, Latagliata R, Di Veroli A, Scaffidi L, de Marchi F, Cerqui E, Anaclerico B, De Matteis G, Spinsanti M, Sabattini E, Catani L, Aversa F, Di Raimondo F, Vitolo U, Lemoli RM, Fanin R, Merli F, Russo D, Cuneo A, Bacchi Reggiani ML, Cavo M, Vianelli N, Breccia M (2017) Baseline factors associated with response to ruxolitinib: an independent study on 408 patients with myelofibrosis. Oncotarget 8(45):79073–79086. https://​doi.​org/​10.​18632/​oncotarget.​18674CrossRefPubMedCentralPubMed
50.
Zurück zum Zitat Pardanani A, Harrison C, Cortes JE, Cervantes F, Mesa RA, Milligan D, Masszi T, Mishchenko E, Jourdan E, Vannucchi AM, Drummond MW, Jurgutis M, Kuliczkowski K, Gheorghita E, Passamonti F, Neumann F, Patki A, Gao G, Tefferi A (2015) Safety and efficacy of fedratinib in patients with primary or secondary myelofibrosis: a randomized clinical trial. JAMA Oncol 1(5):643–651. https://doi.org/10.1001/jamaoncol.2015.1590CrossRefPubMed Pardanani A, Harrison C, Cortes JE, Cervantes F, Mesa RA, Milligan D, Masszi T, Mishchenko E, Jourdan E, Vannucchi AM, Drummond MW, Jurgutis M, Kuliczkowski K, Gheorghita E, Passamonti F, Neumann F, Patki A, Gao G, Tefferi A (2015) Safety and efficacy of fedratinib in patients with primary or secondary myelofibrosis: a randomized clinical trial. JAMA Oncol 1(5):643–651. https://​doi.​org/​10.​1001/​jamaoncol.​2015.​1590CrossRefPubMed
51.
Zurück zum Zitat Mesa RA, Gerds AT, Vannucchi A, Al-Ali HK, Lavie D, Kuykendall AT, Grosicki S, Iurlo A, Goh YT, Lazaroiu MC, Egyed M, Fox ML, McLornan DP, Perkins A, Yoon S-S, Gupta V, Kiladjian J-J, Donahue R, Kawashima J, Verstovsek S (2022) MOMENTUM: phase 3 randomized study of momelotinib (MMB) versus danazol (DAN) in symptomatic and anemic myelofibrosis (MF) patients previously treated with a JAK inhibitor. J Clin Oncol 40(16 suppl):7002–7002. https://doi.org/10.1200/JCO.2022.40.16_suppl.7002CrossRef Mesa RA, Gerds AT, Vannucchi A, Al-Ali HK, Lavie D, Kuykendall AT, Grosicki S, Iurlo A, Goh YT, Lazaroiu MC, Egyed M, Fox ML, McLornan DP, Perkins A, Yoon S-S, Gupta V, Kiladjian J-J, Donahue R, Kawashima J, Verstovsek S (2022) MOMENTUM: phase 3 randomized study of momelotinib (MMB) versus danazol (DAN) in symptomatic and anemic myelofibrosis (MF) patients previously treated with a JAK inhibitor. J Clin Oncol 40(16 suppl):7002–7002. https://​doi.​org/​10.​1200/​JCO.​2022.​40.​16_​suppl.​7002CrossRef
52.
Zurück zum Zitat Gowin K, Kosiorek H, Dueck A, Mascarenhas J, Hoffman R, Reeder C, Camoriano J, Tibes R, Gano K, Palmer J, Mesa R (2017) Multicenter phase 2 study of combination therapy with ruxolitinib and danazol in patients with myelofibrosis. Leuk Res 60:31–35CrossRefPubMed Gowin K, Kosiorek H, Dueck A, Mascarenhas J, Hoffman R, Reeder C, Camoriano J, Tibes R, Gano K, Palmer J, Mesa R (2017) Multicenter phase 2 study of combination therapy with ruxolitinib and danazol in patients with myelofibrosis. Leuk Res 60:31–35CrossRefPubMed
53.
Zurück zum Zitat Gerds AT, Vannucchi AM, Passamonti F, Kremyanskaya M, Gotlib J, Palmer JM, McCaul K, Ribrag V, Mead AJ, Harrison C, Mesa R, Kiladjian J-J, Barosi G, Gerike TG, Shetty JK, Pariseau J, Miranda G, Schwickart M, Giuseppi AC, Zhang J, Backstrom JT, Verstovsek S (2020) Duration of response to luspatercept in patients (pts) requiring red blood cell (RBC) transfusions with myelofibrosis (MF) - updated data from the phase 2 ACE-536-MF-001 study. Blood 136(suppl 1):47–48. https://doi.org/10.1182/blood-2020-137265CrossRef Gerds AT, Vannucchi AM, Passamonti F, Kremyanskaya M, Gotlib J, Palmer JM, McCaul K, Ribrag V, Mead AJ, Harrison C, Mesa R, Kiladjian J-J, Barosi G, Gerike TG, Shetty JK, Pariseau J, Miranda G, Schwickart M, Giuseppi AC, Zhang J, Backstrom JT, Verstovsek S (2020) Duration of response to luspatercept in patients (pts) requiring red blood cell (RBC) transfusions with myelofibrosis (MF) - updated data from the phase 2 ACE-536-MF-001 study. Blood 136(suppl 1):47–48. https://​doi.​org/​10.​1182/​blood-2020-137265CrossRef
54.
Zurück zum Zitat Bose P, Daver N, Pemmaraju N, Jabbour EJ, Estrov Z, Pike A, Huynh-Lu J, Nguyen-Cao M, Wang X, Zhou L, Pierce S, Kantarjian HM, Verstovsek S (2017) Sotatercept (ACE-011) alone and in combination with ruxolitinib in patients (pts) with myeloproliferative neoplasm (MPN)-associated myelofibrosis (MF) and anemia. Blood 130(suppl 1):255. https://doi.org/10.1182/blood.V130.Suppl_1.255.255CrossRef Bose P, Daver N, Pemmaraju N, Jabbour EJ, Estrov Z, Pike A, Huynh-Lu J, Nguyen-Cao M, Wang X, Zhou L, Pierce S, Kantarjian HM, Verstovsek S (2017) Sotatercept (ACE-011) alone and in combination with ruxolitinib in patients (pts) with myeloproliferative neoplasm (MPN)-associated myelofibrosis (MF) and anemia. Blood 130(suppl 1):255. https://​doi.​org/​10.​1182/​blood.​V130.​Suppl_​1.​255.​255CrossRef
55.
Zurück zum Zitat Rampal RK, Verstovsek S, Devlin SM, King AC, Stein EM, Pemmaraju N, Mauro MJ, Kadia TM, Montalban-Bravo G, Alvarez K, Ard N, Goodman T, Taylor B, Bose P (2019) Safety and efficacy of combined ruxolitinib and thalidomide in patients with myelofibrosis: a phase II study. Blood 134(suppl 1):4163. https://doi.org/10.1182/blood-2019-127661CrossRef Rampal RK, Verstovsek S, Devlin SM, King AC, Stein EM, Pemmaraju N, Mauro MJ, Kadia TM, Montalban-Bravo G, Alvarez K, Ard N, Goodman T, Taylor B, Bose P (2019) Safety and efficacy of combined ruxolitinib and thalidomide in patients with myelofibrosis: a phase II study. Blood 134(suppl 1):4163. https://​doi.​org/​10.​1182/​blood-2019-127661CrossRef
Metadaten
Titel
How I manage anemia related to myelofibrosis and its treatment regimens
verfasst von
Srdan Verstovsek
Publikationsdatum
14.02.2023
Verlag
Springer Berlin Heidelberg
Erschienen in
Annals of Hematology / Ausgabe 4/2023
Print ISSN: 0939-5555
Elektronische ISSN: 1432-0584
DOI
https://doi.org/10.1007/s00277-023-05126-4

Weitere Artikel der Ausgabe 4/2023

Annals of Hematology 4/2023 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Positiver FIT: Die Ursache liegt nicht immer im Dickdarm

27.05.2024 Blut im Stuhl Nachrichten

Immunchemischer Stuhltest positiv, Koloskopie negativ – in solchen Fällen kann die Blutungsquelle auch weiter proximal sitzen. Ein Forschungsteam hat nachgesehen, wie häufig und in welchen Lokalisationen das der Fall ist.

GLP-1-Agonisten können Fortschreiten diabetischer Retinopathie begünstigen

24.05.2024 Diabetische Retinopathie Nachrichten

Möglicherweise hängt es von der Art der Diabetesmedikamente ab, wie hoch das Risiko der Betroffenen ist, dass sich sehkraftgefährdende Komplikationen verschlimmern.

Mehr Lebenszeit mit Abemaciclib bei fortgeschrittenem Brustkrebs?

24.05.2024 Mammakarzinom Nachrichten

In der MONARCHE-3-Studie lebten Frauen mit fortgeschrittenem Hormonrezeptor-positivem, HER2-negativem Brustkrebs länger, wenn sie zusätzlich zu einem nicht steroidalen Aromatasehemmer mit Abemaciclib behandelt wurden; allerdings verfehlte der numerische Zugewinn die statistische Signifikanz.

ADT zur Radiatio nach Prostatektomie: Wenn, dann wohl länger

24.05.2024 Prostatakarzinom Nachrichten

Welchen Nutzen es trägt, wenn die Strahlentherapie nach radikaler Prostatektomie um eine Androgendeprivation ergänzt wird, hat die RADICALS-HD-Studie untersucht. Nun liegen die Ergebnisse vor. Sie sprechen für länger dauernden Hormonentzug.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.