Skip to main content
AACR Open Access logoLink to AACR Open Access
. 2022 Mar 3;82(5):749–763. doi: 10.1158/0008-5472.CAN-21-2930

Myelofibrosis: Genetic Characteristics and the Emerging Therapeutic Landscape

Ayalew Tefferi 1,*, Naseema Gangat 1, Animesh Pardanani 1, John D Crispino 2
PMCID: PMC9306313  PMID: 34911786

Abstract

Primary myelofibrosis (PMF) is one of three myeloproliferative neoplasms (MPN) that are morphologically and molecularly inter-related, the other two being polycythemia vera (PV) and essential thrombocythemia (ET). MPNs are characterized by JAK-STAT–activating JAK2, CALR, or MPL mutations that give rise to stem cell–derived clonal myeloproliferation, which is prone to leukemic and, in case of PV and ET, fibrotic transformation. Abnormal megakaryocyte proliferation is accompanied by bone marrow fibrosis and characterizes PMF, while the clinical phenotype is pathogenetically linked to ineffective hematopoiesis and aberrant cytokine expression. Among MPN-associated driver mutations, type 1–like CALR mutation has been associated with favorable prognosis in PMF, while ASXL1, SRSF2, U2AF1-Q157, EZH2, CBL, and K/NRAS mutations have been shown to be prognostically detrimental. Such information has enabled development of exclusively genetic (GIPSS) and clinically integrated (MIPSSv2) prognostic models that facilitate individualized treatment decisions. Allogeneic stem cell transplantation remains the only treatment modality in MF with the potential to prolong survival, whereas drug therapy, including JAK2 inhibitors, is directed mostly at the inflammatory component of the disease and is therefore palliative in nature. Similarly, disease-modifying activity remains elusive for currently available investigational drugs, while their additional value in symptom management awaits controlled confirmation. There is a need for genetic characterization of clinical observations followed by in vitro and in vivo preclinical studies that will hopefully identify therapies that target the malignant clone in MF to improve patient outcomes.

Historical Prelude

William Dameshek (1900–1969) is credited for coining the concept of myeloproliferative disorders (1951; ref. 1), which are now referred to as myeloproliferative neoplasms (MPN; ref. 2). The original MPN members included primary myelofibrosis (PMF), polycythemia vera (PV), essential thrombocythemia (ET), chronic myeloid leukemia, and Di Guglielmo's syndrome (erythroleukemia; ref. 1). Pre-Dameshek PMF luminaries include Gustav Heuck (1854–1940) who first described PMF (1879; ref. 3) and recognized its association with bone marrow fibrosis (BMF), extramedullary hematopoiesis (EMH), and osteosclerosis; Max Askanazy (1865–1940; ref. 4); and Herbert Assmann (1882–1950; ref. 5). Pseudonyms for MF used in the past include agnogenic myeloid metaplasia (6), chronic idiopathic myelofibrosis (7), and myelofibrosis with myeloid metaplasia. In 2006, the International Working Group for Myelofibrosis Research and Treatment (IWG-MRT) selected “PMF” as the preferred term (8). In 1939 (9), Vaughan and Harrison underscored the relationship between PMF, PV, and ET, in terms of their origination from a common mesenchymal reticulum cell, a view subsequently shared by others (10, 11) and ultimately led to the formal description of the MPN concept by Dameshek (1).

Pathogenetic Insights

Dameshek's aforementioned concept of MPN was genetically ratified in 2005 by the seminal discovery of a Janus kinase 2 (JAK2 located on chromosome 9p24) gain of-function mutation (JAK2V617F; a G to T somatic mutation at nucleotide 1849, in exon 14, resulting in the substitution of valine to phenylalanine at codon 617) in PV, ET, and PMF (12–15). Additional MPN driver mutations have since been described and include JAK2 exon 12 described in JAK2V617F-negative PV (16); CALR (calreticulin; located on chromosome 19p13.2; refs. 17, 18) and MPL (myeloproliferative leukemia virus oncogene; located on chromosome 1p34; ref. 19). Among these mutations, JAK2 is the most frequent with frequencies of approximately 98% in PV (95% JAK2V617F and 3% JAK2 exon 12), 50% to 60% in ET, and 55% to 65% in PMF (20, 21). CALR and MPL mutations are often absent in PV, except rare exceptions (22, 23), while their frequencies in ET are estimated at 20% to 25% and 3% to 4%, respectively, and in PMF at 20% to 25% and 6% to 7% (21). Approximately 10% to 15% of patients with PMF or ET do not express any one of the three MPN driver mutations and are operationally referred to as being triple-negative, although that might not be the case during higher sensitivity testing (21, 24). MPN driver mutations have also been reported in other myeloid malignancies, including myelodysplastic syndromes with ring sideroblasts associated with marked thrombocytosis (MDS-RS-T; 50% frequency; refs. 25, 26).

JAK2 and MPLW515K/L/A/R and S505N mutations are believed to directly activate JAK2-STAT, resulting in clonal myeloproliferation that is cytokine independent or hypersensitive (19, 27). Frameshift CALR mutations mostly include type 1 (52-bp deletion in exon 9) or type 2 (5-bp insertion in exon 9), and less frequently a myriad of type 1-like or type 2-like variants (28). The precise mechanism of mutant CALR-induced myeloproliferation is less clear but one possibility includes mutant CALR binding to the extracellular domain of MPL in the endoplasmic reticulum, leading to dimerization and transfer to cell surface and activation of JAK-STAT (29). Mutant CALR-induced mouse models have suggested a primary effect on platelet production that is followed by development of MF (30). Although the central role of JAK-STAT activation in MPN has been highlighted (29, 31), the particular concept is confounded by the coexistence of an inflammatory state in MF with aberrant cytokine expression and the fact that activated JAK-STAT is a nonspecific phenomenon in cancer (32). Furthermore, “targeted therapy” with JAK inhibitors has so far failed to induce selective suppression of the disease clone in MF (33).

The pathogenetic role of MPN driver mutations is highlighted by their origin at the stem cell level and the demonstration of persistent JAK-STAT activation and induction of mutant JAK2/CALR/MPL–driven MPN phenotype in mice (13, 34). Recent studies have highlighted the central pathogenetic role of persistent MPL-driven JAK-STAT activation in MPN; this is presumably accomplished, in addition to activating MPL mutations, by direct activation and altered trafficking of MPL by JAK2 V617F or binding of extracellular domain of MPL by mutant CALR that begins in the endoplasmic reticulum and leads to dimerization of immature receptors and their translocation to the cell surface for activation (29, 35, 36). It is currently assumed that the phenotypic differences between MPN variants are in part contributed by differences in the conformations of specific cytokine receptors that lead to distinct signaling outcomes for EPOR and MPL and further modified by interactions with other cooccurring mutations, including those of epigenetic regulators, and their order of acquisition (29, 34). Additional mechanisms of phenotype diversity associated with MPN-driving mutations include variations in signal intensity (often related to JAK2V617F allele burden) and specificity of downstream signals, including STAT5, STAT1, and STAT3 activation (37). Phenotype-wise, JAK2V617F, versus mutant CALR, is associated with older age, higher hemoglobin level, leukocytosis, lower platelet count and increased risk of thrombosis, while the latter was associated with younger age, male sex, higher platelet count, lower hemoglobin level, lower leukocyte count and lower incidence of thrombotic events (17, 18, 38, 39); type 2 versus type 1 CALR mutations were associated with higher platelet count (40). Patients with CALR-mutated PMF are also younger and present with higher platelet count and lower frequencies of anemia, leukocytosis, and spliceosome mutations (41). Recently published mouse studies appear to recapitulate the phenotypic differences seen among patients with different driver mutations (42, 43). Regardless, the underlying mechanisms that enable single mutations to result in different MPN phenotypes remain not fully understood (44–47).

Other mutations in PMF with frequencies of 10% or higher include ASXL1 (additional sex combs-like 1), TET2 (TET oncogene family member 2), SRSF2 (serine/arginine-rich splicing factor 2), and U2AF1 (U2 small nuclear RNA auxiliary factor 1; refs. 48–50); the latter has been associated with severe anemia and thrombocytopenia in PMF (50) and treatment response to imetelstat (51) while all three mutations (ASXL1, SRSF2, U2AF1) have been associated with inferior survival (52–54). Other mutations who are less frequent in chronic phase disease but with significantly higher frequency in blast phase MPN include IDH1/IDH2 (isocitrate dehydrogenase 1 and 2; refs. 55, 56), TP53 (tumor protein p53; refs. 56, 57), DNMT3A (DNA cytosine methyltransferase 3a; ref. 58), and LNK mutation (59, 60). Other infrequent mutations reported in MPN include SF3B1 (splicing factor 3B subunit 1), which are characteristic and more frequent in MDS-RS and MDS-RS-T (25), EZH2 (enhancer of zeste homolog 2), which were shown to be prognostically relevant (61), Casitas B-lineage lymphoma proto-oncogene (CBL), which are more frequent in juvenile and chronic myelomonocytic leukemia (62) and SETBP1 (SET binding protein 1), which are more frequent in chronic neutrophilic leukemia and atypical chronic myeloid leukemia (63, 64).

The pathogenetic role of mutations other than JAK2/CALR/MPL in MPN is much less understood but believed to involve cooperation with driver mutations, leading to downstream disruption of epigenetic (e.g., ASXL1, TET2, EZH2, IDH1, IDH2, DNMT3A), RNA splicing (e.g., SRSF2, U2AF1, SF3B1), or transcriptional (TP53, IKZF1, NF-E2, CUX1) regulation, which might facilitate disease progression and leukemic transformation (56, 65). The recent demonstration of TET2, ASXL1, and DNMT3A mutations in “normal” elderly individuals has added to the complexity regarding their precise pathogenetic contribution (66, 67). More recent observations in MF include identification of STK11 as a tumor suppressor and that loss of LKB1/STK11 leads to stabilization of HIF1α and thus might promote disease progression (68), impaired megakaryocyte maturation associated with reduced GATA1 expression that might stem from ribosomal deficiency (69), and identification of aurora kinase A (70, 71), peroxisome proliferator-activated receptor gamma (PPARγ; ref. 72) and cyclin-dependent kinase 6 (CDK6; ref. 73), as therapeutic targets in MF.

Among the JAK2 mutation–related MPNs (MF, PV, ET), MF is uniquely characterized by more intense bone marrow and splenic tissue stromal changes, including reticulin/collagen fibrosis and neoangiogenesis (74). The mechanisms of such intense stromal reaction are not fully understood but might involve abnormal proliferation of fibroblasts, endothelial, and other mesenchymal cells mediated by inflammatory cytokines derived from clonal megakaryocytes or mononuclear cells, as well as in situ cell interactions involving microvascular endothelial cells and mesenchymal cells (75–81). Whether or not some of the latter stromal cells are part of the underlying disease clone is currently debated although the presence of their increased proliferation has been demonstrated and in certain cases phenotypically characterized (82–85). The potential role of fibroblasts in bone marrow fibrosis is supported by their morphologic and phenotypic resemblance to hematopoietic stem cells and monocytes and their participation in the production of extracellular matrix components (86). Several studies in MF have suggested increased expression of fibroblast growth factors or their receptors and pathogenetic interaction between megakaryocytes and fibroblasts (75–77). The reactive nature of bone marrow fibrosis in MF is further supported by clinical observations that document its complete resolution under effective targeted therapy of the disease-initiating clone (51, 87).

Mutation-Enhanced Morphologic Diagnosis

Over the last several years, we have been involved in the development of both the 2008 (88) and 2016 (89) World Health Organization (WHO) classification system for MPNs. According to these consensus guidelines, PMF is operationally subclassified into “prefibrotic” and “overtly fibrotic” variant (90–92). Diagnosis of overt PMF requires the presence of three major criteria and at least one minor criterion: major criteria include (i) presence of grade ≥2 reticulin/collagen bone marrow fibrosis, associated with megakaryocyte proliferation and dysmorphia, (ii) presence of JAK2/CALR/MPL or other clonal markers, or absence of evidence for reactive bone marrow fibrosis, and (iii) absence of evidence for another myeloid neoplasm; minor criteria include the presence of (i) anemia, (ii) leukocytosis, (iii) palpable splenomegaly, (iv) increased serum level of lactate dehydrogenase (LDH), and (v) leukoerythroblastosis; the latter indicates presence of nucleated red cells, immature granulocytes and/or dacryocytes (teardrop-shaped erythrocytes) in the peripheral blood. The diagnostic criteria for prefibrotic PMF are almost identical to those of overt PMF, with two exceptions: (i) the first major criterion does not require the presence of grade ≥2 reticulin/collagen bone marrow fibrosis, and (ii) leukoerythroblastosis was not listed as a minor criterion. Because more than 80% of patients with PMF harbor mutations other than JAK2/CALR/MPL, additional mutation screening by next-generation DNA sequencing (NGS) might help confirm clonality in triple-negative cases (93). About 15% of patients with ET or PV develop a PMF-like phenotype over time, referred to as post-ET or post-PV MF. The diagnosis of post-PV or post-ET MF should adhere to criteria published by the IWG-MRT; these criteria require documentation of (i) an antecedent WHO-compliant diagnosis of PV or ET, and (ii) presence of grade ≥2 reticulin/collagen bone marrow fibrosis, as well as presence of at least 2 minor criteria, including (i) anemia, (ii) leukoerythroblastosis, (iii) increasing splenomegaly, (iv) development of constitutional symptoms, and, in case of post-ET MF, (v) increased LDH.

Clinical Phenotype

Prominent clinical features in MF include anemia (attributed primarily to ineffective erythropoiesis), hepatosplenomegaly (attributed to EMH and cytokine-mediated organ congestion), constitutional symptoms including fatigue, night sweats, and low-grade fever, progressive cachexia with loss of muscle mass, bone pain, splenic infarct, pruritus, nonhepatosplenic EMH, thrombosis and bleeding (94). Consequences of hepatosplenic EMH include portal hypertension that might lead to variceal bleeding or ascites and those of nonhepatosplenic EMH include spinal cord compression, ascites, pleural effusion, pulmonary hypertension or extremity pain. It is currently assumed that aberrant cytokine production by clonal cells and host immune reaction contribute to PMF-associated bone marrow stromal changes, ineffective erythropoiesis, EMH, cachexia, and constitutional symptoms (95). Causes of death in PMF include leukemic progression that occurs in approximately 20% of patients (96).

Mutation and Karyotype-Enhanced Prognostication

Beginning in 2009, a number of prognostic models in PMF have been described and summarized in Table 1 (52–54, 97–99). The most recently developed prognostic models in PMF incorporate mutations and/or cytogenetic information: MIPSS70, MIPSS70+ version 2.0 (MIPSSv2) and GIPSS (52–54). MIPSS70 (mutation-enhanced international prognostic scoring system for transplant-age patients) utilizes mutations and clinical variables (54); MIPSSv2 (the karyotype-enhanced MIPSS70) utilizes mutations, karyotype and clinical variables (52); GIPSS (the genetically-inspired prognostic scoring system) is based exclusively on mutations and karyotype (53).

Table 1.

Contemporary prognostic scoring systems for primary myelofibrosis.

Risk categories
Models Variables Very low Low Intermediate-1 Intermediate-2 High Very high
IPSS (97)a Age >65 years (1 point) NA (0 points) (1 point) (2 points) (≥3 points) NA
Constitutional symptomsb (1 point) 11.3 years 7.9 years 4 years 2.3 years
Hemoglobin <10 g/dL (1 point)
Leukocytes >25 x 10(9)/L (1 point)
Circulating blasts ≥1% (1 point)
DIPSS (98)c Age >65 years (1 point) NA (0 points) (1–2 points) (3–4 points) (5–6 points) NA
Constitutional symptoms (1 point) Not reached 14.2 years 4 years 1.5 years
Hemoglobin <10 g/dL (2 points)
Leukocytes >25 x 10(9)/L (1 point)
Circulating blasts ≥1% (1 point)
DIPSS-plus (99)d Age >65 years (1 point) NA (0 points) (1 point) (2–3 points) (≥4 points) NA
Constitutional symptomsb (1 point) 15.4 years 6.5 years 2.9 years 1.3 years
Hemoglobin <10 g/dL (1 point)
Leukocytes >25 x 10(9)/L (1 point)
Circulating blasts ≥1% (1 point)
Unfavorable karyotypee (1 point)
Platelet count <100 x 10(9)/L (1 point)
Transfusion needs (1 point)
MIPSS70 (54)c ≥2 HMR mutationsf (2 points) NA (0–1 point) (2–4 points) (≥5 points) NA
Leukocytes >25 × 109/L (2 points) Not reached 6.3 years 3.1 years
Platelets <100 × 109/L (2 points)
Hemoglobin <10 g/dL (1 point)
Circulating blasts ≥2% (1 point)
BM fibrosis grade ≥2 (1 point)
Constitutional symptomsa (1 point)
Type 1/like CALR absent (1 point)
One HMR mutationf (1 point)
MIPSS70+v2 (52)d Very high-risk karyotypeg (4 points) (0 points) (1–2 points) (3–4 points) (5–8 points) (≥9 points)
Unfavorable karyotypeh (3 points) Not reached 16.4 years 7.7 years 4.1 years 1.8 years
≥2 HMR mutationsf (3 points)
One HMR mutationf (2 points)
Type 1/like CALR absent (2 points)
Constitutional symptomsb (2 points)
Severe anemiai (2 points)
Moderate anemiaj (1 point)
Circulating blasts ≥2% (1 point)
GIPSS (53)d Very high-risk karyotypeg (2 points) NA (0 points) (1 point) (2 points) (≥3 points) NA
Unfavorable karyotypeh (1 point) 26.4 years 8 years 4.2 years 2 years
ASXL1 mutation (1 point)
SRSF2 mutation (1 point)
U2AF1Q157 mutation (1 point)
Type 1/like CALR absent (1 point)

Abbreviations: DIPSS, Dynamic International Prognostic Scoring System; GIPSS, Genetically Inspired International Prognostic Scoring System; IPSS, International Prognostic Scoring System; MIPSS, Mutation-enhanced International Prognostic Scoring System (age ≤ 70); NA, not applicable.

aHMR for MIPSSv2 and GIPSS include ASXL1, SRSF2, and U2AF1Q157.

bConstitutional symptoms include weight loss, fever, drenching night sweats.

cParameters used at the time of diagnosis.

dParameters used at any time in the clinical course.

eUnfavorable karyotype in the context of DIPSS-plus = complex karyotype or sole or two abnormalities that include +8, −7/7q-, i(17q), inv(3), −5/5q-, 12p- or 11q23 rearrangement.

fHigh molecular risk (HMR) mutations for MIPSS70 include ASXL1, SRSF2, EZH2, IDH1, IDH2.

gVery high-risk (VHR) karyotype = single/multiple abnormalities of -7, inv(3)/3q21, i(17q), 12p-/12p11.2 or 11q-/11q23, single/multiple autosomal trisomies other than +9 and +8.

hUnfavorable karyotype in the context of GIPSS/MIPSSv2 = any abnormal karyotype other than VHR karyotype, normal karyotype or sole abnormalities of 20q-, 13q-, +9, chr. 1 translocation/duplication, -Y, or sex chromosome abnormality other than -Y.

iSevere anemia: Hemoglobin <8 g/dL in women and <9 g/dL men.

jModerate anemia: Hemoglobin 8–9.9 g/dL in women and 9–10.9 g/dL men.

MIPSSv2 incorporates 5 genetic and 4 clinical risk factors (52); the five genetic risk factors include very high risk (VHR) karyotype (4 adverse points), unfavorable karyotype (3 points), ≥2 high molecular risk (HMR) mutations (3 points), presence of one HMR mutation (2 points), absence of type 1/like CALR mutation (2 points); the four clinical variables in MIPSSv2 include constitutional symptoms (2 points), severe anemia, defined by hemoglobin levels of <8 g/dL in women and <9 g/dL in men (2 points), moderate anemia, defined by hemoglobin levels of 8–9.9 g/dL in women and 9–10.9 g/dL in men (one point) and circulating blasts ≥2% (one point; Fig. 1). MIPSSv2 includes five risk categories: very high risk (≥9 points); high risk (5–8 points); intermediate risk (3–4 points); low risk (1–2 points); and very low risk (zero points); in patients age 70 years or younger, the corresponding median survivals (10-year survival rates) were 1.8 years (<5%), 4.1 years (13%), 7.7 years (37%), 16.4 years (56%) and “median not reached” (92%; Fig. 1). In instances where cytogenetic information is not available, MIPSS70 (54) performs as well as MIPSSv2, whereas GIPSS offers a prognostic model that is exclusively dependent on mutations and karyotype (53). Most recently, RAS/CBL mutations in PMF were associated with poor response to ruxolitinib therapy, poor prognostic features and inferior survival; in the latter regard, however, incorporation of such information to MIPSSv2 did not show additional value (100, 101).

Figure 1.

Figure 1. Current treatment algorithm in myelofibrosis based on risk stratification according to the mutation enhanced international prognostic scoring system (MIPSS70+ version 2.0). Genetic risk factors: very high-risk karyotype, 4 points; unfavorable karyotype, 3 points; ≥2 high risk mutations, 3 points; one high risk mutation, 2 points; type 1 CALR absent, 2 points. Clinical risk factors: constitutional symptoms, 2 points; severe anemia, 2 points; moderate anemia, 1 point; ≥2% circulating blasts, 1 point.

Current treatment algorithm in myelofibrosis based on risk stratification according to the mutation enhanced international prognostic scoring system (MIPSS70+ version 2.0). Genetic risk factors: very high-risk karyotype, 4 points; unfavorable karyotype, 3 points; ≥2 high risk mutations, 3 points; one high risk mutation, 2 points; type 1 CALR absent, 2 points. Clinical risk factors: constitutional symptoms, 2 points; severe anemia, 2 points; moderate anemia, 1 point; ≥2% circulating blasts, 1 point.

Risk-Adjusted Treatment Approach

Allogeneic hematopoietic stem cell transplant (AHSCT) currently remains the only treatment modality in MF that can prolong life and, in some cases, cure the disease (102). However, transplant-related complications are not trivial and might result in death or substantial morbidity in about half of the cases (103). Therefore, for the individual patient, the risk of AHSCT must be balanced against expected survival without AHSCT. On the other hand, current drug therapy in MF, including use of JAK2 inhibitors, is mostly palliative and has not been shown to modify disease natural history or prolong survival (Table 2; refs. 33, 104–108). Figure 1 outlines our current treatment algorithm that is based on aforementioned risk stratification model, MIPSSv2.

Table 2.

Conventional drug treatment strategies in myelofibrosis.

Indications Treatment options Reported response rates Side effects include
Anemia Thalidomide 50 mg QD + Prednisone 20 mg QD (171, 172) 46%–62% Peripheral neuropathy
Constipation
Fatigue
Cutaneous reactions
CNS symptoms/sedation
Sleep disturbances
Anabolic steroids (173, 174) (dose depends on preparation) 44%–57% Virilizing effects
Liver toxicity
Danazol 200 mg BID (175) 30% (175) Liver toxicity
LFT abnormalities
Lenalidomide 10 mg QD ± Prednisone 20 mg QD (176, 177) 20%–25% Myelosuppression
ESAs (114, 178) 0% in Tx-dependent (114) Unremarkable
37% in Tx-independent (114)
Splenomegaly Hydroxyurea 500 mg BID (112) (starting dose) 40%b ↓hemoglobin; ↓platelet count
Skin changes including ulcers
Oral ulcers
Nail discolorations
Ruxolitinib 15 mg BID (117, 120) (starting dose) 32%a ↓hemoglobin; ↓platelet count
Herpes zoster (180)
Reactivation of tuberculosis (180)
Other opportunistic infections (180)
↓COVID-19 vaccine response (126)
Fedratinib 400 mg QD (129, 179) 47%a ↓hemoglobin; ↓platelet count
↑LFTs; ↑pancreatic enzymes
Gastrointestinal symptoms
Encephalopathyb
Constitutional symptoms Ruxolitinib 15 mg BID (117, 120) Majority of patients See above
Fedratinib 400 mg QD (129, 179) Majority of patients See above

Abbreviations: BID, twice-daily; ESAs, erythropoiesis-stimulating agents including darbepoetin 150–300 g every 2 weeks; LFT, liver function test; Tx, red blood cell transfusion; QD, once-daily.

aSeen at 500 mg/day dose.

bResponse rates are not comparable because of reference to different patient populations and different methods and timing of spleen response assessment.

There is no evidence to support the value of specific drug therapy in asymptomatic patients with MIPSSv2 “low” or “very low” risk disease, whose expected 10-year survival rates were reported at 50% and 86%, respectively; expected survival in such patients was retrospectively estimated to be superior in the absence of AHSCT as first-line therapy (109, 110). Therefore, observation alone is a reasonable treatment strategy in such patients, in the absence of treatment-requiring symptoms. AHSCT is the preferred treatment of choice for MIPSSv2 “high” or “very high” risk disease, where 10-year expected survival rates without transplant might be as low as 10% and <3%, respectively (Fig. 1; refs. 109, 110). In transplant-ineligible patients with high/very high-risk disease, as well as in those with symptomatic low/intermediate risk disease, clinical trial participation might be the most appropriate treatment choice, considering the current dearth of disease-modifying drugs.

Symptom-directed conventional therapy

Choice of drugs for combatting MF symptoms is based on the primary clinical indication and is outlined in Table 2 and Fig. 1. For anemia, we consider androgen preparations, prednisone, immunomodulatory drugs (IMiDs; thalidomide, lenalidomide, pomalidomide), or danazol, depending on tolerability of potential side effects for the individual patient (111). For symptomatic splenomegaly, our first-line drug of choice is hydroxyurea (112) and secondarily JAK2 inhibitors (e.g., ruxolitinib, fedratinib). Ruxolitinib is our first-line drug of choice for constitutional symptoms while involved-field radiotherapy is most effective for symptomatic non-hepatosplenic EMH or localized bone pain. Splenectomy might be necessary for drug-refractory spleen-associated symptoms. Anemia response rates to each one of the aforementioned drugs are in the vicinity of 15 to 25% and response durations average about one to two years; lenalidomide works best in the presence of del(5q31) (87). We are less enthusiastic about the use of radiotherapy for hepatosplenic EMH (danger of treatment-induced protracted thrombocytopenia; ref. 113) and the use of erythropoiesis stimulating agents (ESA; refs. 114, 115) or luspatercept (116), because of inadequate efficacy (112).

Ruxolitinib and fedratinib are now FDA approved for use in MF and are considered effective treatment options for countering splenomegaly and constitutional symptoms in hydroxyurea-refractory MF, with response rates estimated at about 30% to 50% (117–120). However, treatment with these two drugs is complicated by treatment-emergent anemia and thrombocytopenia (120). Longer-term experience with ruxolitinib therapy in MF has disclosed a high rate of treatment discontinuation rate (92% after a median time of 9.2 months) and the occurrence of severe withdrawal symptoms during treatment discontinuation (“ruxolitinib withdrawal syndrome”), characterized by acute relapse of disease symptoms, accelerated splenomegaly, worsening of cytopenias and occasional hemodynamic decompensation, including a septic shock-like syndrome (105, 121, 122). In addition, several reports have now associated ruxolitinib with sometimes fatal serious opportunistic infections (123–125), and as of late, poor immune antibody response to COVID-19 vaccination (126). It is important to underscore the fact that neither ruxolitinib nor other JAK2 inhibitors in MF possess antitumor activity and none of them has been shown to reverse bone marrow fibrosis or induce cytogenetic or molecular remissions. Instead, their mechanism of action is based on their broad activity in suppressing inflammatory cytokines.

JAK2 inhibitors other than ruxolitinib that have undergone phase III clinical trials include fedratinib, momelotinib, and pacritinib (127). Fedratinib was recently approved for use in patients intolerant or resistant to ruxolitinib, with approximately a third of patients responding, regardless of the reason for ruxolitinib discontinuation (128). Compared with placebo, the spleen response rate with fedratinib 400 mg/day (36% vs. 1%) was similar to that seen with ruxolitinib (JAKARTA-1; ref. 129); spleen response rates for pacritinib were 19% versus 5% and 18% versus 3%, compared with BAT, which excluded (PERSIST-1) (130) or included (PERSIST-2; ref. 131) JAK inhibitor therapy, the latter involving patients with platelet count of <100 × 106/L. Momelotinib was compared with ruxolitinib (SIMPLIFY-1) with similar spleen response rates (26.5% vs. 29%; ref. 132) and compared with BAT (SIMPLIFY-2) in patients failing treatment with ruxolitinib with meager spleen response rates (7% vs. 6%; ref. 133). The toxicity profiles were noticeably different for the aforementioned JAK inhibitors: fedratinib (Wernicke's encephalopathy, anemia, thrombocytopenia, gastrointestinal distress and elevations in serum liver function tests and pancreatic enzymes); pacritinib (cardiac events, severe diarrhea, nausea, thrombocytopenia, anemia and hemorrhage); momelotinib (peripheral neuropathy, thrombocytopenia, first-dose effect including dizziness, nausea, hypotension, headache, and flushing; refs. 107, 108). Among the currently available JAK inhibitors, momelotinib is uniquely identified as being potentially useful in alleviating anemia, in addition to the expected responses in spleen size and symptoms, and the particular activity has been ascribed to the drug's inhibitory activity against activin receptor type 1 (ACVR1), which upregulates hepcidin synthesis (134).

AHSCT in Myelofibrosis

MIPSSv2-stratifed outcome analysis of AHSCT in MF was assessed in a retrospective study of 110 patients with a median follow-up of 64 months (110); approximately 89% of evaluable patients were MIPSSv2 high or very high risk and the remaining intermediate risk. The 5-year overall survival rate for the entire patient cohort was 65%, progression-free survival 60%, relapse rate 17%, nonrelapse mortality 24%, grade 2–4 acute graft versus host disease (GVHD) 45%, and chronic extensive GVHD 59%. In the particular study (110), somatic mutations did not affect outcome after AHSCT, whereas survival in patients with intermediate or high risk MIPSSv2 risk category (median not reached at last follow-up) was superior to that of patients with very high risk MIPSSv2 risk category (median 25 months; ref. 110). The better than 50% five-year survival was also apparent in our own institutional experience where neither very high risk nor unfavorable karyotype affected post-transplant survival (102). In a much larger study of 2,224 patients with MF who underwent AHSCT, 781 received myeloablative (MAC; median age 53 years) and 1,443 reduced intensity (RIC; median age 58 years) conditioning; outcome in the two groups were mostly similar in regards to engraftment period, rates of grade 2–4 acute GVHD (MAC 28% and RIC 31%), 5-year nonrelapse mortality (MAC 35% and RIC 34%), and 5-year survival (MAC 53% and RIC 51%; ref. 135). More recent studies have confirmed the value of AHSCT in older patients (136), the possibility of using family mismatched donors (137) and newer effective therapies for GVHD (e.g., ruxolitinib, ibrutinib; refs. 138–140). In a recent study of 556 transplanted patients with MF and age ≥65 years (median 67, range 65–76), followed for a median of 3.4 years, 5-year survival, nonrelapse mortality, and relapse rates were 40%, 37%, and 25%, respectively (136). Similarly favorable outcome data have also been reported in patients receiving alternative donor grafts (141). Unresolved issues with AHSCT in MF include the role of splenectomy (142–144), involved field radiotherapy (145, 146), and use of JAK2 inhibitors before and after AHSCT (147–151). A number of recent publications have addressed the issue of pretransplant management of splenomegaly in MF, including splenectomy, splenic irradiation, and use of JAK2 inhibitors (142, 144, 145, 147, 152). There is currently no consensus regarding either the need or specific treatment option in this regard and decisions are best made on a case-by-case basis (152).

New Drugs

In this section, we provide a synopsis of selected novel agents under active clinical investigation in patients with MF (Table 3).

Table 3.

Summary of selected novel therapies in clinical trials in patients with myelofibrosis.

Drug/mechanism Dose Spleen response Anemia response Symptom response Correlative studies Toxicity Current status
Pelabresib (CPI-0610) (153155, 181) 125 mg daily on days 1–14 of a 21-day cycle Week 24 Week 24 Week 24 Non-TD cohort Thrombocytopenia (25.6%),
 Bromodomain and Extraterminal Domain Inhibitor (BETi). SVR≥35% TD cohort TSS≥50% Improvement in fibrosis Anemia (11.6%),
 Phase II trial (MANIFEST) Non-TD cohort 3/14 (21%) Non-TD cohort Nausea (32.6%),
Arm 1 (refractory/intolerant to ruxolitinib). 5/21(23.8%) TD-TI 9/19 (47.4%) Diarrhea (30.2%),
 Non-TD (n = 27) TD cohort none Non-TD cohort TD cohort Respiratory tract
 TD (n = 19) 13/22 (59%), hemoglobin improved ≥1.5 g/dL over 12 weeks 1/12 (8.3%)  infections (27.9%)
 Arm 2 (suboptimal response or progression on ruxolitinib) CPI-0610 + ruxolitinib Week 24 TD cohort 13/36 (36%) TD-TI Week 24 Not provided Thrombocytopenia (40%),
TD (n = 52), Non-TD (n = 26). SVR≥35% TSS ≥ 50% Asthenia (32%),
TD cohort: 5/24 (20.8%) Non-TD cohort TD cohort: 12/26 (46.2%) Diarrhea (46%),
Non-TD cohort: 4/18 (22.2%) 4/23 (17%) Hb improved ≥1.5 g/dL over 12 weeks. Nausea (36%),
Non-TD cohort: 7/19 (36.8%) Respiratory infections (32%)
Arm 3: CPI-0610 + ruxolitinib 10 mg, bid Week 24 None Week 24 Not provided Anemia (23.4%), Phase 3 study CPI-0610 + ruxolitinib vs. placebo + ruxolitinib in JAKi-naïve MF (MANIFEST-2)
JAKi-naïve patients SVR≥35% TSS≥50% Thrombocytopenia (20.3%),
(n = 64) 19/30 (63.3%) 17/29 (58.6%) Diarrhea (26.6%),
Respiratory infections (18.8%),
Nausea (18.8%)
Navitoclax (157, 182) 50 mg to 300 mg daily + ruxolitinib (>10 mg, bid) Week 24 7/11 (64%) with Hb < 10 g/dL or TD improved ≥ 2 g/dL (n = 6) or TI (n = 1) Week 24 Fibrosis reduction AEs ≥20% Phase III study navitoclax + ruxolitinib vs. placebo + ruxolitinib in JAKi naïve (TRANSFORM-1) vs. BAT in relapsed/refractory to JAKi. (TRANSFORM-2)
Antiapoptotic, binds to B-cell lymphoma 2 (BCL2) SVR ≥35% 9 (27%) TSS≥50% 6/20 (30%) ≥1 grade in 11/33 (33%). Thrombocytopenia (88%)
Family, including BCL-XL, BCL2, and BCL-W Diarrhea
Phase II trial in pts with suboptimal response to ruxolitinib (n = 34) Anytime SVR ≥35% 15 (44%) 12/26 (46%) with >10% driver gene VAF reduction. Fatigue
Median duration of SVR- 13.8 months Median overall survival- not reached 24-month survival- 84% Anemia
Nausea
Dizziness
Confusion
Vomiting
Imetelstat (161) Imetelstat (9.4 mg/kg or 4.7 mg/kg) i.v. every 3 weeks Week 24 None Week 24 41% with ≥1 grade reduction in fibrosis. Thrombocytopenia (49%), Phase 3 study 9.4 mg/kg of imetelstat vs. BAT with the exclusion of JAKi in refractory MF
 Telomerase inhibitor SVR ≥35% TSS≥50% 5/24 (20.8%) with ≥50% reduction in abnormal cytogenetic clones (all with sole 13q) Anemia (44%),
 Phase II trial (MYF2001) in relapsed/refractory to JAKi, (n = 107) 6 (10.2%) in 9.4 mg/kg arm with no responses in 4.7 mg/kg arm 19 (32%) in 9.4 mg/kg arm and 3 (6%) in 4.7 mg/kg arm. 12/26 (46.2%) in 9.4 mg/kg vs. (4/23) Neutropenia (36%),
17.4% in 4.7 mg/kg arm with ≥20% VAF reduction of any mutated gene Nausea (34%).
Median overall survival 29.9 months with 9.4 mg/kg.
Luspatercept (159, 160) 1–1.75 mg/kg s.c. every 21 days Not provided Week 24 Not provided Not provided Treatment-related AEs; Phase 3 study in MF-associated anemia with concomitant JAKi therapy and transfusion needs
 Binds to TGFβ superfamily ligand, reduces aberrant Smad2/3 signaling and enhances late-stage erythropoiesis. Cohort 1 (14%) Hypertension (13%),
 Phase II trial  Cohort 3a (21%)) Bone pain (9%),
 Non-TD without ruxolitinib (n = 22) (Cohort 1) Cohort 2 Diarrhea (5%).
 2/21 (10%) 8 (10%) pts had ≥ 1 AE leading to drug discontinuation
 With ruxolitinib (n = 14) Cohort 3b
  (Cohort 3a) TD 6/22 (27%) achieved TI.
Overall
 Without ruxolitinib (n = 21) Cohort 2
  (Cohort 2) 4/21 (19%)
 With ruxolitinib (n = 22) Cohort 3b
  (Cohort 3b) 8/22 (36%) achieved TI.
Parsaclisib (164) 10 or 20 mg QD Week 12 Hemoglobin remained stable Week 12 Not provided 18%/30% Phase 3 randomized study ruxolitinib + parsaclisib in JAK- and PI3K-inhibitor treatment-naïve myelofibrosis.
 PI3Kδ inhibitor oral for 8 wks median % change, median % change grade 3 thrombocytopenia. Phase 3 randomized study ruxolitinib + parsaclisib vs. ruxolitinib + placebo in patients with suboptimal responseto ruxolitinib.
 Phase II trial in MF with suboptimal response to ruxolitinib (n = 53) QW thereafter) or parsaclisib QD (5 or 20 mg QD for 8 wks/5 mg QD thereafter) with stable ruxolitinib dose (5–25 mg bid) −2.3 and −15.4 −14.0 in QD/QW 21%/0%
Week 24 −39.6 in QD grade 4 thrombocytopenia,
−2.5 and −25.4 with QD/QW and QD dosing grade 3/4 events; tuberculosis,
enteritis, fatigue, hypertension,
abnormal liver tests, stomatitis
Bomedemstat (IMG-7289) (163) Dose uptitrated based on platelet count targeting 50–100k/μL (QD orally). Week 24 72% of evaluable patients (n = 36) with stable or improved hemoglobin >1 g/dl. Week 24 Improvement in fibrosis > grade 1 in 26%. 8 SAEs possibly related: Ongoing.
 Lysine-specific demethylase, LSD1 inhibitor 81% (n = 19) 85% (n = 13), Reduction in driver/HMR mutation VAF in 42%  painful splenomegaly,
 Phase II trial in MF resistant to approved therapies. (n = 62) reduction in spleen volume (mean SVR: -8%). TSS reduction (mean change -31%).  rectal bleeding, heart failure,
31% with TSS >50% reduction.  headache, vertigo,
 GI hemorrhage,
 anemia,
 pyoderma gangrenosum
Sotatercept (ACE-011) (183) Single agent 0.75 or 1 mg/kg S/C every 3 weeks (n = 31) and in combination with a stable dose of ruxolitinib (n = 21) Not provided Sotatercept alone. Not provided Not provided Hypertension (n = 7), Ongoing
 activin receptor type IIA ligand trap  4 anemia responses (29%); limb pain (n = 3),
 Phase II trial in MF pts with anemia, (n = 53)  3 TD to TI. headache (n = 1)
combination cohort
5/17 (29%) pts responded,
all non-TD pts
Tagraxofusp (SL-401) (184) Stage 1 (dose escalation), at 7, 9, or 12 mcg/kg i.v. on days 1–3 every 21 days (cycle 1–4), Week 24 None 3 patients by IWG-MRT Not provided hypoalbuminemia (23%), Ongoing
CD123-directed antibody 28 days (cycles 5–7), Spleen response by palpation (n = 10, 45%), 12/26 (46%) with symptom reduction. headache (17%),
Phase I/II trial in relapsed/refractory MF (n = 35) 42 days (cycles 8+). 2 pts with >50% reduction. elevated ALT (17%).
Stage 2 (expansion), Capillary leak syndrome in 3 pts (1 grade 4).
recommended phase II dose (12 mcg/kg).

Abbreviations: ALT, alanine transferase; AEs, adverse events; BAT, best available therapy; BID, twice daily; HMR, high molecular risk mutation; IWG-MRT, International working group-myeloproliferative neoplasms research and treatment; JAKi- Janus kinase inhibitor; MF, myelofibrosis; SAE, serious adverse event; SVR, spleen volume reduction;TSS, total symptom score; TI, transfusion independent; TD, transfusion dependent; VAF, variant allele frequency; QD/QW, once daily/once weekly.

Pelabresib (CPI-0610)

CPI-0610, is an oral inhibitor of bromodomain and extraterminal domain (BET) proteins that inhibits cytokine production and promotes megakaryocytic and erythroid differentiation. In the phase II (MANIFEST) study, CPI-0610 was administered as (i) monotherapy in MF patients that were refractory or intolerant to JAK inhibitors (Arm 1, n = 43; ref. 153), (ii) add on therapy in those with suboptimal response to ruxolitinib (Arm 2, n = 78; ref. 154), or (iii) in combination with ruxolitinib in JAK inhibitor–naïve patients (Arm 3, n = 64; ref. 155); 3/14 (21%) and 13/36 (36%) transfusion-dependent patients achieved transfusion independence in Arm 1 (CPI-0610 monotherapy) and Arm 2 (CPI-0610 with ruxolitinib) cohorts, respectively (153, 154). Correspondingly, among nontransfusion-dependent patients, 13 of 22 (59%) in Arm 1 and 4 of 23 (17%) in Arm 2 had a rise in hemoglobin level of >1.5 g/dL (153, 154). Notably, none of the transfusion dependent patients in Arm 1 versus 21% in Arm 2 demonstrated spleen response. CPI-0610 in combination with ruxolitinib yielded spleen and symptom response in 63% and 59% of JAK inhibitor–naïve patients, respectively (155). Commonly reported adverse events included gastrointestinal toxicity (46%), thrombocytopenia (40%), respiratory infections (32%), and anemia (12%; refs. 153–155). Based on these findings, a randomized placebo-controlled phase III study (MANIFEST-2) with CP-0610 in combination with ruxolitinib in JAK inhibitor–naïve patients is ongoing (156).

Navitoclax

Navitoclax is an oral small-molecule inhibitor of antiapoptotic B-cell lymphoma 2 (BCL2) family proteins, including BCL-XL, BCL2, and BCL-W. In a phase II study with navitoclax in combination with ruxolitinib in patients with MF with suboptimal response to ruxolitinib, 34 patients were enrolled, with ongoing therapy in 17 patients (157, 158). Spleen and symptom response were reported in 27% and 30% of patients, respectively (157). Commonly reported adverse events included thrombocytopenia in 88%, which led to dose reduction in 56% of patients, diarrhea (71%), and fatigue (62%). As a next step, two phase III studies are ongoing, which include a placebo-controlled study of navitoclax in combination with ruxolitinib in JAK inhibitor treatment–naïve patients (TRANSFORM-1), and navitoclax in combination with ruxolitinib versus best available therapy in patients relapsed following JAK inhibitor therapy (TRANSFORM-2).

Luspatercept

Luspatercept, an erythropoiesis maturating agent that binds to TGFβ superfamily ligand, reduces aberrant Smad2/3 signaling and enhances late-stage erythropoiesis, is FDA approved for myelodysplastic syndromes with ringed sideroblasts. In an ongoing phase II investigation in MF, four cohorts that included transfusion-dependent and -independent patients that were either receiving or not on ruxolitinib were enrolled (159). Among transfusion-dependent patients, 36% and 19% of patients receiving or not receiving ruxolitinib were rendered transfusion independent with median duration of response of 55 and 59 weeks, respectively (160). Most common adverse event of hypertension was noted in 13% of patients. Currently, luspatercept is undergoing phase III investigation in the INDEPENDENCE study, which includes patients with MF with transfusion requiring anemia and receiving JAK inhibitor therapy.

Imetelstat

Imetelstat, a telomerase inhibitor, has proven safety and efficacy in a pilot study in patients with treatment-naïve and relapsed MF (51). Recently published data from the randomized phase II study in patients relapsed/refractory to JAK inhibitors showed modest activity in terms of spleen (10%) and symptom (32%) response in patients receiving imetelstat 9.4 mg/kg i.v. every 3 weeks (161). However, improvement in bone marrow fibrosis was noted in 41%, with driver mutation variant allele frequency reduction in 42% of patients, which correlated with superior overall survival (median survival; 29.9 months; ref. 161). Major toxicities included thrombocytopenia recorded in half of patients, followed by anemia (44%) and neutropenia in one-third of patients (161). Based on its selective impact on the malignant clone, a confirmatory phase II study of imetelstat 9.4 mg/kg vs. best available therapy excluding JAK inhibitors in relapsed MF is ongoing.

Bomedemstat (IMG-7289)

An oral inhibitor of lysine-specific demethylase-1 inhibitor (LSD1), which is involved in differentiation and maturation of megakaryocytes, has been shown to improve blood counts, splenomegaly, cytokine profile, mutant allele burden and fibrosis in murine models (162). In a phase II study of IMG-7289 monotherapy in patients with MF resistant to approved therapies, 62 patients have been enrolled, majority had received prior ruxolitinib (n = 56), one-third were transfusion-dependent, and 94% harbored ASXL1 mutation (163). Eighty-one percent of patients demonstrated spleen response, one-third reported symptom response, and 72% of patients had stable or >1 g/dL improvement in hemoglobin levels (163). In addition, 26% of patients demonstrated at least grade 1 improvement in fibrosis, 42% with reduction, and 50% with stability of driver and high-risk mutation allele burden.

Parsaclisib

An oral highly selective PI3Kδ inhibitor is under phase II investigation in patients with MF with suboptimal response to ruxolitinib that may arise due to persistent PI3K/AKT activation (164). Patients with MF underwent randomization to add-on parsaclisib once daily/once weekly or parsaclisib once daily while remaining on a stable ruxolitinib dose. Fifty-three patients have been treated; 33 and 20 patients received parsaclisib once daily/once weekly and once daily, respectively. At 12- and 24-week assessments, median percent spleen change was −2.3 and −15.4 at week 12; and −2.5 and −25.4 at week 24 with once daily/once weekly and once daily dosing, respectively, while median percent change in symptom score at week 12 was −14.0 in once daily/once weekly, and −39.6 in once daily (164). With once daily/once weekly and once daily dose, 18% and 30% developed grade 3 thrombocytopenia; 21% and 0% had grade 4 thrombocytopenia, while hemoglobin levels remained stable. Additional grade 3/4 treatment-related, adverse events included disseminated tuberculosis, enteritis, fatigue, hypertension, abnormal liver tests and stomatitis. Importantly, half of patients interrupted parsaclisib due to adverse events (164). On the basis of the above findings, a randomized study of add-on parsaclisib versus placebo in patients with suboptimal response to ruxolitinib is underway (165), together with first-line phase III investigation in patients with JAK and PI3K inhibitor—naïve MF (166).

Concluding Remarks

At present, AHSCT remains the only treatment modality in MF that secures disease-free remission state and prolonged survival; furthermore, palliative value beyond ruxolitinib has remained out of reach for most of the drugs that are currently under investigation, and the possibility of incremental value is likely to be countered by additional side effects (Table 1). The scenario warrants urgent attention to newer therapeutic targets and, more importantly, identification of repurposable drug candidates, to accelerate the discovery process (167). In a recent edition of Cancer Research, Dutta and colleagues described their observations from JAK2V617F knock-in mice, which included upregulation of CDK6 expression in hematopoietic progenitors and in vivo therapeutic activity for CDK4/6 inhibitor palbociclib alone or in combination with ruxolitinib, including reversal of bone marrow fibrosis and reduction of spleen size (73). The observations regarding PPARγ and CDK6 as therapeutic targets are particularly noteworthy considering the current availability of FDA-approved drug antagonists (168, 169). A similar approach might be needed to address the unmet need in the treatment of blast-phase MF, which occurs in approximately 10%–20% of patients during their clinical course and where outcome under current therapy, including AHSCT, is dismal (170). Now more than ever, there is a need for genetic characterization of clinical observations followed by the relevant in vitro and in vivo animal studies that hopefully lead to development of novel therapies that target the malignant clone, and not only its secondary constitutional effects.

Authors' Disclosures

A. Pardanani reports personal fees from Adelphi Values outside the submitted work. J.D. Crispino reports grants from ALSAC during the conduct of the study. No disclosures were reported by the other authors.

References

  • 1. Dameshek W. Some speculations on the myeloproliferative syndromes. Blood 1951;6:372–5. [PubMed] [Google Scholar]
  • 2. Swerdlow HS, Campo E, Haris NL, Jaffe ES, Pileri SA, Stein H, et al. WHO classification of tumours of haematopoietic and lymphoid tissues. Lyon, France: IARC; 2017. [Google Scholar]
  • 3. Heuck G. Zwei Falle von Leukamie mit eigenthumlichem Blut- resp. Knochenmarksbefund (Two cases of leukemia with peculiar blood and bone marrow findings, respectively). Arch Pathol Anat Physiol Virchows 1879;78:475–96. [Google Scholar]
  • 4. Askanazy M. Ueber extrauterine Bildung von Blutzellen in der Leber. Verh Dtsch Pathol Ges 1904;7:58–65. [Google Scholar]
  • 5. Assmann H. Beitrage zur osteosklerotischen anamie. Beitr Pathol Anat Allgemeinen Pathologie 1907;41:565–95. [Google Scholar]
  • 6. Jackson H Jr, Parker F Jr, Lemon HM. Agnogenic myeloid metaplasia of the spleen: a syndrome simulating other more definite hematological disorders. N Engl J Med 1940;222:985–94. [Google Scholar]
  • 7. World Health Organization. World Health Organization classification of tumours of haematopoietic and lymphoid tissues. Lyon, France: IARC Press; 2001. [Google Scholar]
  • 8. Mesa RA, Verstovsek S, Cervantes F, Barosi G, Reilly JT, Dupriez B, et al. Primary myelofibrosis (PMF), post polycythemia vera myelofibrosis (post-PV MF), post essential thrombocythemia myelofibrosis (post-ET MF), blast phase PMF (PMF-BP): consensus on terminology by the international working group for myelofibrosis research and treatment (IWG-MRT). Leuk Res 2007;31:737–40. [DOI] [PubMed] [Google Scholar]
  • 9. Vaughan JM, Harrison CV. Leuco-erythrobalstic anaemia and myelosclerosis. J Pathol Bacteriol 1939;48:339–52. [Google Scholar]
  • 10. Rosenthal N, Erf LA. Clinical observations on osteopetrosis and myelofibrosis. Arch Intern Med 1943;71:793–813. [Google Scholar]
  • 11. Heller EL, Lewisohn MG, Palin WE. Aleukemic myelosis: chronic nonleukemic myelosis, agnogenic myeloid metaplasia, osteosclerosis, leuko-erythroblastic anemia, and synonymous designations. Am J Pathol 1947;23:327–65. [PMC free article] [PubMed] [Google Scholar]
  • 12. Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJP, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 2005;7:387–97. [DOI] [PubMed] [Google Scholar]
  • 13. James C, Ugo V, Le Couédic J-P, Staerk J, Delhommeau F, Lacout C, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005;434:1144–8. [DOI] [PubMed] [Google Scholar]
  • 14. Kralovics R, Passamonti F, Buser AS, Teo S-S, Tiedt R, Passweg JR, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 2005;352:1779–90. [DOI] [PubMed] [Google Scholar]
  • 15. Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 2005;365:1054–61. [DOI] [PubMed] [Google Scholar]
  • 16. Scott LM, Tong W, Levine RL, Scott MA, Beer PA, Stratton MR, et al. JAK2 exon 12 mutations in polycythemia vera and idiopathic erythrocytosis. N Engl J Med 2007;356:459–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Nangalia J, Massie CE, Baxter EJ, Nice FL, Gundem G, Wedge DC, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med 2013;369:2391–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Klampfl T, Gisslinger H, Harutyunyan AS, Nivarthi H, Rumi E, Milosevic JD, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med 2013;369:2379–90. [DOI] [PubMed] [Google Scholar]
  • 19. Pikman Y, Lee BH, Mercher T, McDowell E, Ebert BL, Gozo M, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med 2006;3:e270. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Pardanani A, Lasho TL, Finke C, Hanson CA, Tefferi A. Prevalence and clinicopathologic correlates of JAK2 exon 12 mutations in JAK2V617F-negative polycythemia vera. Leukemia 2007;21:1960–3. [DOI] [PubMed] [Google Scholar]
  • 21. Tefferi A, Guglielmelli P, Larson DR, Finke C, Wassie EA, Pieri L, et al. Long-term survival and blast transformation in molecularly annotated essential thrombocythemia, polycythemia vera, and myelofibrosis. Blood 2014;124:2507–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Broséus J, Park J-H, Carillo S, Hermouet S, Girodon F. Presence of calreticulin mutations in JAK2-negative polycythemia vera. Blood 2014;124:3964–6. [DOI] [PubMed] [Google Scholar]
  • 23. Pardanani A, Lasho TL, Finke CM, Tefferi A. Infrequent occurrence of MPL exon 10 mutations in polycythemia vera and post-polycythemia vera myelofibrosis. Am J Hematol 2011;86:701–2. [DOI] [PubMed] [Google Scholar]
  • 24. Alimam S, Villiers W, Dillon R, Simpson M, Runglall M, Smith A, et al. Patients with triple-negative, JAK2V617F- and CALR-mutated essential thrombocythemia share a unique gene expression signature. Blood Adv 2021;5:1059–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Broséus J, Alpermann T, Wulfert M, Florensa Brichs L, Jeromin S, Lippert E, et al. Age, JAK2(V617F) and SF3B1 mutations are the main predicting factors for survival in refractory anaemia with ring sideroblasts and marked thrombocytosis. Leukemia 2013;27:1826–31. [DOI] [PubMed] [Google Scholar]
  • 26. Swerdlow SHCE, Harris NL, Jaffe ES, Pileri SA, Stein H, Thiele J, et al. WHO classification of tumours of haematopoietic and lymhoid tissues. Lyon, France: IARC: 2008. [Google Scholar]
  • 27. Dupont S, Masse A, James C, Teyssandier I, Lécluse Y, Larbret F, et al. The JAK2 617V>F mutation triggers erythropoietin hypersensitivity and terminal erythroid amplification in primary cells from patients with polycythemia vera. Blood 2007;110:1013–21. [DOI] [PubMed] [Google Scholar]
  • 28. Lasho TL, Finke CM, Tischer A, Pardanani A, Tefferi A. Mayo CALR mutation type classification guide using alpha helix propensity. Am J Hematol 2018;93:E128–9. [DOI] [PubMed] [Google Scholar]
  • 29. Constantinescu SN, Vainchenker W, Levy G, Papadopoulos N. Functional consequences of mutations in myeloproliferative neoplasms. Hemasphere 2021;5:e578. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Marty C, Harini N, Pecquet C, Chachoua I, Gryshkova V, Villeval J-L, et al. Calr mutants retroviral mouse models lead to a myeloproliferative neoplasm mimicking an essential thrombocythemia progressing to a myelofibrosis. Blood 2014ASH abstract #157. [Google Scholar]
  • 31. Rampal R, Al-Shahrour F, Abdel-Wahab O, Patel JP, Brunel J-P, Mermel CH, et al. Integrated genomic analysis illustrates the central role of JAK-STAT pathway activation in myeloproliferative neoplasm pathogenesis. Blood 2014;123:e123–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Constantinescu SN, Girardot M, Pecquet C. Mining for JAK-STAT mutations in cancer. Trends Biochem Sci 2008;33:122–31. [DOI] [PubMed] [Google Scholar]
  • 33. Tefferi A. Challenges facing JAK inhibitor therapy for myeloproliferative neoplasms. N Engl J Med 2012;366:844–6. [DOI] [PubMed] [Google Scholar]
  • 34. Vainchenker W, Kralovics R. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood 2017;129:667–79. [DOI] [PubMed] [Google Scholar]
  • 35. Pecquet C, Chachoua I, Roy A, Balligand T, Vertenoeil G, Leroy E, et al. Calreticulin mutants as oncogenic rogue chaperones for TpoR and traffic-defective pathogenic TpoR mutants. Blood 2019;133:2669–81. [DOI] [PubMed] [Google Scholar]
  • 36. Nivarthi H, Chen D, Cleary C, Kubesova B, Jäger R, Bogner E, et al. Thrombopoietin receptor is required for the oncogenic function of CALR mutants. Leukemia 2016;30:1759–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Vainchenker W, Constantinescu SN. JAK/STAT signaling in hematological malignancies. Oncogene 2013;32:2601–13. [DOI] [PubMed] [Google Scholar]
  • 38. Rotunno G, Mannarelli C, Guglielmelli P, Pacilli A, Pancrazzi A, Pieri L, et al. Impact of calreticulin mutations on clinical and hematological phenotype and outcome in essential thrombocythemia. Blood 2014;123:1552–5. [DOI] [PubMed] [Google Scholar]
  • 39. Rumi E, Pietra D, Ferretti V, Klampfl T, Harutyunyan AS, Milosevic JD, et al. JAK2 or CALR mutation status defines subtypes of essential thrombocythemia with substantially different clinical course and outcomes. Blood 2014;123:1544–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Tefferi A, Wassie EA, Guglielmelli P, Gangat N, Belachew AA, Lasho TL, et al. Type 1 versus Type 2 calreticulin mutations in essential thrombocythemia: a collaborative study of 1027 patients. Am J Hematol 2014;89:E121–4. [DOI] [PubMed] [Google Scholar]
  • 41. Tefferi A, Lasho TL, Finke CM, Knudson RA, Ketterling R, Hanson CH, et al. CALR vs JAK2 vs MPL-mutated or triple-negative myelofibrosis: clinical, cytogenetic and molecular comparisons. Leukemia 2014;28:1472–7. [DOI] [PubMed] [Google Scholar]
  • 42. Benlabiod C, Cacemiro MDC, Nédélec A, Edmond V, Muller D, Rameau P, et al. Calreticulin del52 and ins5 knock-in mice recapitulate different myeloproliferative phenotypes observed in patients with MPN. Nat Commun 2020;11:4886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43. Toppaldoddi KR, da Costa Cacemiro M, Bluteau O, Panneau-Schmaltz B, Pioch A, Muller D, et al. Rare type 1-like and type 2-like calreticulin mutants induce similar myeloproliferative neoplasms as prevalent type 1 and 2 mutants in mice. Oncogene 2019;38:1651–60. [DOI] [PubMed] [Google Scholar]
  • 44. Tiedt R, Hao-Shen H, Sobas MA, Looser R, Dirnhofer S, Schwaller J, et al. Ratio of mutant JAK2-V617F to wild-type Jak2 determines the MPD phenotypes in transgenic mice. Blood 2008;111:3931–40. [DOI] [PubMed] [Google Scholar]
  • 45. Prick J, de Haan G, Green AR, Kent DG. Clonal heterogeneity as a driver of disease variability in the evolution of myeloproliferative neoplasms. Exp Hematol 2014;42:841–51. [DOI] [PubMed] [Google Scholar]
  • 46. Li J, Kent DG, Godfrey AL, Manning H, Nangalia J, Aziz A, et al. JAK2V617F homozygosity drives a phenotypic switch in myeloproliferative neoplasms, but is insufficient to sustain disease. Blood 2014;123:3139–51. [DOI] [PubMed] [Google Scholar]
  • 47. Chen E, Beer PA, Godfrey AL, Ortmann CA, Li J, Costa-Pereira AP, et al. Distinct clinical phenotypes associated with JAK2V617F reflect differential STAT1 signaling. Cancer Cell 2010;18:524–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Vannucchi AM, Lasho TL, Guglielmelli P, Biamonte F, Pardanani A, Pereira A, et al. Mutations and prognosis in primary myelofibrosis. Leukemia 2013;27:1861–9. [DOI] [PubMed] [Google Scholar]
  • 49. Lasho TL, Jimma T, Finke CM, Patnaik M, Hanson CA, Ketterling RP, et al. SRSF2 mutations in primary myelofibrosis: significant clustering with IDH mutations and independent association with inferior overall and leukemia-free survival. Blood 2012;120:4168–71. [DOI] [PubMed] [Google Scholar]
  • 50. Tefferi A, Finke CM, Lasho TL, Wassie EA, Knudson R, Ketterling RP, et al. U2AF1 mutations in primary myelofibrosis are strongly associated with anemia and thrombocytopenia despite clustering with JAK2V617F and normal karyotype. Leukemia 2014;28:431–3. [DOI] [PubMed] [Google Scholar]
  • 51. Tefferi A, Lasho TL, Begna KH, Patnaik MM, Zblewski DL, Finke CM, et al. A pilot study of the telomerase inhibitor imetelstat for myelofibrosis. N Engl J Med 2015;373:908–19. [DOI] [PubMed] [Google Scholar]
  • 52. Tefferi A, Guglielmelli P, Lasho TL, Gangat N, Ketterling RP, Pardanani A, et al. MIPSS70+ version 2.0: mutation and karyotype-enhanced international prognostic scoring system for primary myelofibrosis. J Clin Oncol 2018;36:1769–70. [DOI] [PubMed] [Google Scholar]
  • 53. Tefferi A, Guglielmelli P, Nicolosi M, Mannelli F, Mudireddy M, Bartalucci N, et al. GIPSS: genetically inspired prognostic scoring system for primary myelofibrosis. Leukemia 2018;32:1631–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. Guglielmelli P, Lasho TL, Rotunno G, Mudireddy M, Mannarelli C, Nicolosi M, et al. MIPSS70: mutation-enhanced international prognostic score system for transplantation-age patients with primary myelofibrosis. J Clin Oncol 2018;36:310–8. [DOI] [PubMed] [Google Scholar]
  • 55. Tefferi A, Lasho TL, Abdel-Wahab O, Guglielmelli P, Patel J, Caramazza D, et al. IDH1 and IDH2 mutation studies in 1473 patients with chronic-, fibrotic- or blast-phase essential thrombocythemia, polycythemia vera or myelofibrosis. Leukemia 2010;24:1302–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Rampal R, Ahn J, Abdel-Wahab O, Nahas M, Wang K, Lipson D, et al. Genomic and functional analysis of leukemic transformation of myeloproliferative neoplasms. Proc Natl Acad Sci U S A 2014;111:E5401–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Harutyunyan A, Klampfl T, Cazzola M, Kralovics R. p53 lesions in leukemic transformation. N Engl J Med 2011;364:488–90. [DOI] [PubMed] [Google Scholar]
  • 58. Abdel-Wahab O, Pardanani A, Rampal R, Lasho TL, Levine RL, Tefferi A. DNMT3A mutational analysis in primary myelofibrosis, chronic myelomonocytic leukemia and advanced phases of myeloproliferative neoplasms. Leukemia 2011;25:1219–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59. Pardanani A, Lasho T, Finke C, Oh ST, Gotlib J, Tefferi A. LNK mutation studies in blast-phase myeloproliferative neoplasms, and in chronic-phase disease with TET2, IDH, JAK2 or MPL mutations. Leukemia 2010;24:1713–8. [DOI] [PubMed] [Google Scholar]
  • 60. Tefferi A. Novel mutations and their functional and clinical relevance in myeloproliferative neoplasms: JAK2, MPL, TET2, ASXL1, CBL, IDH and IKZF1. Leukemia 2010;24:1128–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61. Guglielmelli P, Biamonte F, Score J, Hidalgo-Curtis C, Cervantes F, Maffioli M, et al. EZH2 mutational status predicts poor survival in myelofibrosis. Blood 2011;118:5227–34. [DOI] [PubMed] [Google Scholar]
  • 62. Grand FH, Hidalgo-Curtis CE, Ernst T, Zoi K, Zoi C, McGuire C, et al. Frequent CBL mutations associated with 11q acquired uniparental disomy in myeloproliferative neoplasms. Blood 2009;113:6182–92. [DOI] [PubMed] [Google Scholar]
  • 63. Pardanani A, Lasho TL, Laborde RR, Elliott M, Hanson CA, Knudson RA, et al. CSF3R T618I is a highly prevalent and specific mutation in chronic neutrophilic leukemia. Leukemia 2013;27:1870–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Piazza R, Valletta S, Winkelmann N, Redaelli S, Spinelli R, Pirola A, et al. Recurrent SETBP1 mutations in atypical chronic myeloid leukemia. Nat Genet 2013;45:18–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Chen E, Schneider RK, Breyfogle LJ, Rosen EA, Poveromo L, Elf S, et al. Distinct effects of concomitant Jak2V617F expression and Tet2 loss in mice combine to promote disease progression in myeloproliferative neoplasms. Blood 2015;125:327–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66. Genovese G, Kähler AK, Handsaker RE, Lindberg J, Rose SA, Bakhoum SF, et al. Clonal hematopoiesis and blood-cancer risk inferred from blood DNA sequence. N Engl J Med 2014;371:2477–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Jaiswal S, Fontanillas P, Flannick J, Manning A, Grauman PV, Mar BG, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 2014;371:2488–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Marinaccio C, Suraneni P, Celik H, Volk A, Wen QJ, Ling T, et al. LKB1/STK11 is a tumor suppressor in the progression of myeloproliferative neoplasms. Cancer Discov 2021;11:1398–410. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69. Gilles L, Arslan AD, Marinaccio C, Wen QJ, Arya P, McNulty M, et al. Downregulation of GATA1 drives impaired hematopoiesis in primary myelofibrosis. J Clin Invest 2017;127:1316–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Wen QJ, Yang Q, Goldenson B, Malinge S, Lasho T, Schneider RK, et al. Targeting megakaryocytic-induced fibrosis in myeloproliferative neoplasms by AURKA inhibition. Nat Med 2015;21:1473–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Gangat N, Marinaccio C, Swords R, Watts JM, Gurbuxani S, Rademaker A, et al. Aurora kinase a inhibition provides clinical benefit, normalizes megakaryocytes, and reduces bone marrow fibrosis in patients with myelofibrosis: a phase I trial. Clin Cancer Res 2019;25:4898–906. [DOI] [PubMed] [Google Scholar]
  • 72. Lambert J, Saliba J, Calderon C, Sii-Felice K, Salma M, Edmond V, et al. PPARgamma agonists promote the resolution of myelofibrosis in preclinical models. J Clin Invest 2021;131:e136713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Dutta A, Nath D, Yang Y, Le BT, Mohi G. CDK6 is a therapeutic target in myelofibrosis. Cancer Res 2021;81:4332–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74. Mesa RA, Hanson CA, Rajkumar SV, Schroeder G, Tefferi A. Evaluation and clinical correlations of bone marrow angiogenesis in myelofibrosis with myeloid metaplasia. Blood 2000;96:3374–80. [PubMed] [Google Scholar]
  • 75. Schmitz B, Thiele J, Otto F, Farahmand P, Henze F, Frimpong S, et al. Evidence for integrin receptor involvement in megakaryocyte-fibroblast interaction: a possible pathomechanism for the evolution of myelofibrosis. J Cell Physiol 1998;176:445–55. [DOI] [PubMed] [Google Scholar]
  • 76. Martyré M‐C, Le Bousse-Kerdiles M-C, Romquin N, Chevillard S, Praloran V, Demory J-L, et al. Elevated levels of basic fibroblast growth factor in megakaryocytes and platelets from patients with idiopathic myelofibrosis. Br J Haematol 1997;97:441–8. [DOI] [PubMed] [Google Scholar]
  • 77. Le Bousse-Kerdiles M, Chevillard S, Charpentier A, Romquin N, Clay D, Smadja-Joffe F, et al. Differential expression of transforming growth factor-beta, basic fibroblast growth factor, and their receptors in CD34+ hematopoietic progenitor cells from patients with myelofibrosis and myeloid metaplasia. Blood 1996;88:4534–46. [PubMed] [Google Scholar]
  • 78. Erba BG, Gruppi C, Corada M, Pisati F, Rosti V, Bartalucci N', et al. Endothelial-to-mesenchymal transition in bone marrow and spleen of primary myelofibrosis. Am J Pathol 2017;187:1879–92. [DOI] [PubMed] [Google Scholar]
  • 79. Pieri L, Guglielmelli P, Bogani C, Bosi A, Vannucchi AM. Mesenchymal stem cells from JAK2(V617F) mutant patients with primary myelofibrosis do not harbor JAK2 mutant allele. Leuk Res 2008;32:516–7. [DOI] [PubMed] [Google Scholar]
  • 80. Bacher U, Asenova S, Badbaran A, Zander AR, Alchalby H, Fehse B, et al. Bone marrow mesenchymal stromal cells remain of recipient origin after allogeneic SCT and do not harbor the JAK2V617F mutation in patients with myelofibrosis. Clin Exp Med 2010;10:205–8. [DOI] [PubMed] [Google Scholar]
  • 81. Martinaud C, Desterke C, Konopacki J, Pieri L, Torossian F, Golub R, et al. Osteogenic potential of mesenchymal stromal cells contributes to primary myelofibrosis. Cancer Res 2015;75:4753–65. [DOI] [PubMed] [Google Scholar]
  • 82. Rosti V, Villani L, Riboni R, Poletto V, Bonetti E, Tozzi L, et al. Spleen endothelial cells from patients with myelofibrosis harbor the JAK2V617F mutation. Blood 2013;121:360–8. [DOI] [PubMed] [Google Scholar]
  • 83. Reilly JT, Nash JRG, Mackie MJ, McVerry BA. Endothelial cell proliferation in myelofibrosis. Br J Haematol 1985;60:625–30. [DOI] [PubMed] [Google Scholar]
  • 84. Massa M, Rosti V, Ramajoli I, Campanelli R, Pecci A, Viarengo G, et al. Circulating CD34+, CD133+, and vascular endothelial growth factor receptor 2-positive endothelial progenitor cells in myelofibrosis with myeloid metaplasia. J Clin Oncol 2005;23:5688–95. [DOI] [PubMed] [Google Scholar]
  • 85. Bock O, Loch G, Schade U, Busche G, Wasielewski R, Wiese B, et al. Osteosclerosis in advanced chronic idiopathic myelofibrosis is associated with endothelial overexpression of osteoprotegerin. Br J Haematol 2005;130:76–82. [DOI] [PubMed] [Google Scholar]
  • 86. Bellini A, Mattoli S. The role of the fibrocyte, a bone marrow-derived mesenchymal progenitor, in reactive and reparative fibroses. Lab Invest 2007;87:858–70. [DOI] [PubMed] [Google Scholar]
  • 87. Tefferi A, Lasho TL, Mesa RA, Pardanani A, Ketterling RP, Hanson CA. Lenalidomide therapy in del(5)(q31)-associated myelofibrosis: cytogenetic and JAK2V617F molecular remissions. Leukemia 2007;21:1827–8. [DOI] [PubMed] [Google Scholar]
  • 88. Vardiman JW, Thiele J, Arber DA, Brunning RD, Borowitz MJ, Porwit A, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 2009;114:937–51. [DOI] [PubMed] [Google Scholar]
  • 89. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM, et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016;127:2391–405. [DOI] [PubMed] [Google Scholar]
  • 90. Mudireddy M, Shah S, Lasho T, Barraco D, Hanson CA, Ketterling RP, et al. Prefibrotic versus overtly fibrotic primary myelofibrosis: clinical, cytogenetic, molecular and prognostic comparisons. Br J Haematol 2018;182:594–7. [DOI] [PubMed] [Google Scholar]
  • 91. Guglielmelli P, Pacilli A, Rotunno G, Rumi E, Rosti V, Delaini F, et al. Presentation and outcome of patients with 2016 WHO diagnosis of prefibrotic and overt primary myelofibrosis. Blood 2017;129:3227–36. [DOI] [PubMed] [Google Scholar]
  • 92. Guglielmelli P, Vannucchi AM, Investigators A. The prognostic impact of bone marrow fibrosis in primary myelofibrosis. Am J Hematol 2016;91:E454–5. [DOI] [PubMed] [Google Scholar]
  • 93. Tefferi A, Lasho TL, Finke CM, Elala Y, Hanson CA, Ketterling RP, et al. Targeted deep sequencing in primary myelofibrosis. Blood Adv 2016;1:105–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Tefferi A. Myelofibrosis with myeloid metaplasia. N Engl J Med 2000;342:1255–65. [DOI] [PubMed] [Google Scholar]
  • 95. Tefferi A. Pathogenesis of myelofibrosis with myeloid metaplasia. J Clin Oncol 2005;23:8520–30. [DOI] [PubMed] [Google Scholar]
  • 96. Tefferi A, Mudireddy M, Mannelli F, Begna KH, Patnaik MM, Hanson CA, et al. Blast phase myeloproliferative neoplasm: Mayo-AGIMM study of 410 patients from two separate cohorts. Leukemia 2018;32:1200–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Cervantes F, Dupriez B, Pereira A, Passamonti F, Reilly JT, Morra E, et al. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. Blood 2009;113:2895–901. [DOI] [PubMed] [Google Scholar]
  • 98. Passamonti F, Cervantes F, Vannucchi AM, Morra E, Rumi E, Pereira A, et al. A dynamic prognostic model to predict survival in primary myelofibrosis: a study by the IWG-MRT (International Working Group for Myeloproliferative Neoplasms Research and Treatment). Blood 2010;115:1703–8. [DOI] [PubMed] [Google Scholar]
  • 99. Gangat N, Caramazza D, Vaidya R, George G, Begna K, Schwager S, et al. DIPSS plus: a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. J Clin Oncol 2011;29:392–7. [DOI] [PubMed] [Google Scholar]
  • 100. Coltro G, Rotunno G, Mannelli L, Mannarelli C, Fiaccabrino S, Romagnoli S, et al. RAS/CBL mutations predict resistance to JAK inhibitors in myelofibrosis and are associated with poor prognostic features. Blood Adv 2020;4:3677–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Santos FPS, Getta B, Masarova L, Famulare C, Schulman J, Datoguia TS, et al. Prognostic impact of RAS-pathway mutations in patients with myelofibrosis. Leukemia 2020;34:799–810. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Tefferi A, Partain DK, Palmer JM, Slack JL, Roy V, Hogan WJ, et al. Allogeneic hematopoietic stem cell transplant overcomes the adverse survival effect of very high risk and unfavorable karyotype in myelofibrosis. Am J Hematol 2018;93:649–54. [DOI] [PubMed] [Google Scholar]
  • 103. Ballen KK, Shrestha S, Sobocinski KA, Zhang M-J, Bashey A, Bolwell BJ, et al. Outcome of transplantation for myelofibrosis. Biol Blood Marrow Transplant 2010;16:358–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Cervantes F, Vannucchi AM, Kiladjian J-J, Al-Ali HK, Sirulnik A, Stalbovskaya V, et al. Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood 2013;122:4047–53. [DOI] [PubMed] [Google Scholar]
  • 105. Tefferi A. JAK inhibitors for myeloproliferative neoplasms: clarifying facts from myths. Blood 2012;119:2721–30. [DOI] [PubMed] [Google Scholar]
  • 106. Tefferi A, Litzow MR, Pardanani A. Long-term outcome of treatment with ruxolitinib in myelofibrosis. N Engl J Med 2011;365:1455–7. [DOI] [PubMed] [Google Scholar]
  • 107. Tefferi A, Barraco D, Lasho TL, Shah S, Begna KH, Al-Kali A, et al. Momelotinib therapy for myelofibrosis: a 7-year follow-up. Blood Cancer J 2018;8:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108. Pardanani A, Gotlib J, Roberts AW, Wadleigh M, Sirhan S, Kawashima J, et al. Long-term efficacy and safety of momelotinib, a JAK1 and JAK2 inhibitor, for the treatment of myelofibrosis. Leukemia 2018;32:1035–8. [DOI] [PubMed] [Google Scholar]
  • 109. Kröger N, Giorgino T, Scott BL, Ditschkowski M, Alchalby H, Cervantes F, et al. Impact of allogeneic stem cell transplantation on survival of patients less than 65 years of age with primary myelofibrosis. Blood 2015;125:3347–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Ali H, Aldoss I, Yang D, Mokhtari S, Khaled S, Aribi A, et al. MIPSS70+ v2.0 predicts long-term survival in myelofibrosis after allogeneic HCT with the Flu/Mel conditioning regimen. Blood Adv 2019;3:83–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Tefferi A. Primary myelofibrosis: 2021 update on diagnosis, risk-stratification and management. Am J Hematol 2021;96:145–62. [DOI] [PubMed] [Google Scholar]
  • 112. Martínez-Trillos A, Gaya A, Maffioli M, Arellano-Rodrigo E, Calvo X, Díaz-Beyá M, et al. Efficacy and tolerability of hydroxyurea in the treatment of the hyperproliferative manifestations of myelofibrosis: results in 40 patients. Ann Hematol 2010;89:1233–7. [DOI] [PubMed] [Google Scholar]
  • 113. Elliott MA, Tefferi A. Splenic irradiation in myelofibrosis with myeloid metaplasia: a review. Blood Rev 1999;13:163–70. [DOI] [PubMed] [Google Scholar]
  • 114. Huang J, Tefferi A. Erythropoiesis stimulating agents have limited therapeutic activity in transfusion-dependent patients with primary myelofibrosis regardless of serum erythropoietin level. Eur J Haematol 2009;83:154–5. [DOI] [PubMed] [Google Scholar]
  • 115. Tefferi A, Silverstein MN. Recombinant human erythropoietin therapy in patients with myelofibrosis with myeloid metaplasia. Br J Haematol 1994;86:893. [DOI] [PubMed] [Google Scholar]
  • 116. Tefferi A. New drugs for myeloid neoplasms with ring sideroblasts: luspatercept vs imetelstat. Am J Hematol 2021;96:761–3. [DOI] [PubMed] [Google Scholar]
  • 117. Verstovsek S, Kantarjian H, Mesa RA, Pardanani AD, Cortes-Franco J, Thomas DA, et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med 2010;363:1117–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. Pieri L, Paoli C, Arena U, Marra F, Mori F, Zucchini M, et al. Safety and efficacy of ruxolitinib in splanchnic vein thrombosis associated with myeloproliferative neoplasms. Am J Hematol 2017;92:187–95. [DOI] [PubMed] [Google Scholar]
  • 119. Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med 2012;366:799–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120. Harrison C, Kiladjian J-J, Al-Ali HK, Gisslinger H, Waltzman R, Stalbovskaya V, et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med 2012;366:787–98. [DOI] [PubMed] [Google Scholar]
  • 121. Coltro G, Mannelli F, Guglielmelli P, Pacilli A, Bosi A, Vannucchi AM. A life-threatening ruxolitinib discontinuation syndrome. Am J Hematol 2017;92:833–8. [DOI] [PubMed] [Google Scholar]
  • 122. Palandri F, Palumbo GA, Elli EM, Polverelli N, Benevolo G, Martino B, et al. Ruxolitinib discontinuation syndrome: incidence, risk factors, and management in 251 patients with myelofibrosis. Blood Cancer J 2021;11:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Heine A, Brossart P, Wolf D. Ruxolitinib is a potent immunosuppressive compound: is it time for anti-infective prophylaxis? Blood 2013;122:3843–4. [DOI] [PubMed] [Google Scholar]
  • 124. Tsukamoto Y, Kiyasu J, Tsuda M, Ikeda M, Shiratsuchi M, Ogawa Y, et al. Fatal disseminated tuberculosis during treatment with ruxolitinib plus prednisolone in a patient with primary myelofibrosis: a case report and review of the literature. Intern Med 2018;57:1297–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Eyal O, Flaschner M, Ben Yehuda A, Rund D. Varicella-zoster virus meningoencephalitis in a patient treated with ruxolitinib. Am J Hematol 2017;92:E74–5. [DOI] [PubMed] [Google Scholar]
  • 126. Guglielmelli P, Mazzoni A, Maggi L, Kiros ST, Zammarchi L, Pilerci S, et al. Impaired response to first SARS-CoV-2 dose vaccination in myeloproliferative neoplasm patients receiving ruxolitinib. Am J Hematol 2021;96:E408–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Gangat N, Tefferi A. Myelofibrosis biology and contemporary management. Br J Haematol 2020;191:152–70. [DOI] [PubMed] [Google Scholar]
  • 128. Harrison CN, Schaap N, Vannucchi AM, Kiladjian J‐J, Jourdan E, Silver RT, et al. Fedratinib in patients with myelofibrosis previously treated with ruxolitinib: An updated analysis of the JAKARTA2 study using stringent criteria for ruxolitinib failure. Am J Hematol 2020;95:594–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Pardanani A, Harrison C, Cortes JE, Cervantes F, Mesa RA, Milligan D, et al. Safety and efficacy of fedratinib in patients with primary or secondary myelofibrosis: a randomized clinical trial. JAMA Oncol 2015;1:643–51. [DOI] [PubMed] [Google Scholar]
  • 130. Mesa RA, Vannucchi AM, Mead A, Egyed M, Szoke A, Suvorov A, et al. Pacritinib versus best available therapy for the treatment of myelofibrosis irrespective of baseline cytopenias (PERSIST-1): an international, randomised, phase 3 trial. Lancet Haematol 2017;4:e225–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131. Mascarenhas J, Hoffman R, Talpaz M, Gerds AT, Stein B, Gupta V, et al. Pacritinib vs best available therapy, including ruxolitinib, in patients with myelofibrosis: a randomized clinical trial. JAMA Oncol 2018;4:652–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132. Mesa RA, Kiladjian J-J, Catalano JV, Devos T, Egyed M, Hellmann A, et al. SIMPLIFY-1: a phase III randomized trial of momelotinib versus ruxolitinib in janus kinase inhibitor-naive patients with myelofibrosis. J Clin Oncol 2017;35:3844–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133. Harrison CN, Vannucchi AM, Platzbecker U, Cervantes F, Gupta V, Lavie D, et al. Momelotinib versus best available therapy in patients with myelofibrosis previously treated with ruxolitinib (SIMPLIFY 2): a randomised, open-label, phase 3 trial. Lancet Haematol 2018;5:e73–81. [DOI] [PubMed] [Google Scholar]
  • 134. Oh ST, Talpaz M, Gerds AT, Gupta V, Verstovsek S, Mesa R, et al. ACVR1/JAK1/JAK2 inhibitor momelotinib reverses transfusion dependency and suppresses hepcidin in myelofibrosis phase 2 trial. Blood Adv 2020;4:4282–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. McLornan D, Szydlo R, Koster L, Chalandon Y, Robin M, Wolschke C, et al. Myeloablative and reduced-intensity conditioned allogeneic hematopoietic stem cell transplantation in myelofibrosis: a retrospective study by the chronic malignancies working party of the European society for blood and marrow transplantation. Biol Blood Marrow Transplant 2019;25:2167–71. [DOI] [PubMed] [Google Scholar]
  • 136. Hernández-Boluda J-C, Pereira A, Kröger N, Cornelissen JJ, Finke J, Beelen D, et al. Allogeneic hematopoietic cell transplantation in older myelofibrosis patients: A study of the chronic malignancies working party of EBMT and the Spanish Myelofibrosis Registry. Am J Hematol 2021;96:1186–94. [DOI] [PubMed] [Google Scholar]
  • 137. Raj K, Eikema D-J, McLornan DP, Olavarria E, Blok H-J, Bregante S, et al. Family mismatched allogeneic stem cell transplantation for myelofibrosis: report from the chronic malignancies working party of European society for blood and marrow transplantation. Biol Blood Marrow Transplant 2019;25:522–8. [DOI] [PubMed] [Google Scholar]
  • 138. Zeiser R, Polverelli N, Ram R, Hashmi SK, Chakraverty R, Middeke JM, et al. Ruxolitinib for glucocorticoid-refractory chronic graft-versus-host disease. N Engl J Med 2021;385:228–38. [DOI] [PubMed] [Google Scholar]
  • 139. Zeiser R, von Bubnoff N, Butler J, Mohty M, Niederwieser D, Or R, et al. Ruxolitinib for glucocorticoid-refractory acute graft-versus-host disease. N Engl J Med 2020;382:1800–10. [DOI] [PubMed] [Google Scholar]
  • 140. Miklos D, Cutler CS, Arora M, Waller EK, Jagasia M, Pusic I, et al. Ibrutinib for chronic graft-versus-host disease after failure of prior therapy. Blood 2017;130:2243–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141. Bregante S, Dominietto A, Ghiso A, Raiola AM, Gualandi F, Varaldo R, et al. Improved outcome of alternative donor transplantations in patients with myelofibrosis: from unrelated to haploidentical family donors. Biol Blood Marrow Transplant 2016;22:324–9. [DOI] [PubMed] [Google Scholar]
  • 142. Bossard J-B, Beuscart J-B, Robin M, Mohty M, Barraco F, Chevallier P, et al. Splenectomy before allogeneic hematopoietic cell transplantation for myelofibrosis: A French nationwide study. Am J Hematol 2021;96:80–8. [DOI] [PubMed] [Google Scholar]
  • 143. Robin M, Zine M, Chevret S, Meignin V, Munoz-Bongrand N, Moatti H, et al. The impact of splenectomy in myelofibrosis patients before allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant 2017;23:958–64. [DOI] [PubMed] [Google Scholar]
  • 144. Polverelli N, Mauff K, Kroger N, Robin M, Beelen D, Beauvais D, et al. Impact of spleen size and splenectomy on outcomes of allogeneic hematopoietic cell transplantation for myelofibrosis: a retrospective analysis by the chronic malignancies working party on behalf of European society for blood and marrow transplantation (EBMT). Am J Hematol 2021;96:69–79. [DOI] [PubMed] [Google Scholar]
  • 145. Helbig G, Wieczorkiewicz-Kabut A, Markiewicz M, Krzemień H, Wójciak M, Białas K, et al. Splenic irradiation before allogeneic stem cell transplantation for myelofibrosis. Med Oncol 2019;36:16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Kalman NS, Mukhopadhyay ND, Roberts CH, Chung HM, Clark WB, McCarty JM, et al. Low-dose splenic irradiation prior to hematopoietic cell transplantation in hypersplenic patients with myelofibrosis. Leuk Lymphoma 2017;58:2983–4. [DOI] [PubMed] [Google Scholar]
  • 147. Shahnaz Syed Abd Kadir S, Christopeit M, Wulf G, Wagner E, Bornhauser M, Schroeder T, et al. Impact of ruxolitinib pretreatment on outcomes after allogeneic stem cell transplantation in patients with myelofibrosis. Eur J Haematol 2018;101:305–17. [DOI] [PubMed] [Google Scholar]
  • 148. Stübig T, Alchalby H, Ditschkowski M, Wolf D, Wulf G, Zabelina T, et al. JAK inhibition with ruxolitinib as pretreatment for allogeneic stem cell transplantation in primary or post-ET/PV myelofibrosis. Leukemia 2014;28:1736–8. [DOI] [PubMed] [Google Scholar]
  • 149. Hanif A, Hari PN, Atallah E, Carlson K-S, Pasquini MC, Michaelis LC. Safety of ruxolitinib therapy prior to allogeneic hematopoietic stem-cell transplantation for myeloproliferative neoplasms. Bone Marrow Transplant 2016;51:617–8. [DOI] [PubMed] [Google Scholar]
  • 150. Shanavas M, Popat U, Michaelis LC, Fauble V, McLornan D, Klisovic R, et al. Outcomes of allogeneic hematopoietic cell transplantation in patients with myelofibrosis with prior exposure to janus kinase 1/2 inhibitors. Biol Blood Marrow Transplant 2016;22:432–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151. Kröger N, Sbianchi G, Sirait T, Wolschke C, Beelen D, Passweg J, et al. Impact of prior JAK-inhibitor therapy with ruxolitinib on outcome after allogeneic hematopoietic stem cell transplantation for myelofibrosis: a study of the CMWP of EBMT. Leukemia 2021;35:3551–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Ho VT, Rambaldi A. Splenectomy before allogeneic hematopoietic stem-cell transplantation for myelofibrosis: tumor burden matters! Am J Hematol 2021;96:12–13. [DOI] [PubMed] [Google Scholar]
  • 153. Mascarenhas J, Harrison C, Patriarca A, Devos T, Palandri F, Rampal RK, et al. CPI-0610, a bromodomain and extraterminal domain protein (BET) inhibitor, as monotherapy in advanced myelofibrosis patients refractory/intolerant to JAK inhibitor: update from phase 2 MANIFEST study. Blood 2020;136:55. [Google Scholar]
  • 154. Verstovsek S, Mascarenhas J, Kremyanskaya M, et al. CPI-0610, bromodomain and extraterminal domain protein (BET) inhibitor, as “add-on” to ruxolitinib, in advanced myelofibrosis patients with suboptimal response: update of MANIFEST phase 2 study. Blood 2020;136. [Google Scholar]
  • 155. Mascarenhas J, Harrison C, Luptakova K, Christo J, Wang J, Mertz JA, et al. CPI-0610, a bromodomain and extraterminal domain protein (BET) inhibitor, in combination with ruxolitinib, in JAK-inhibitor-naïve myelofibrosis patients: update of MANIFEST phase 2 study. Blood 2020;136:56. [Google Scholar]
  • 156. Mascarenhas J, Harrison C, Luptakova K, Christo J, Wang J, Mertz JA, et al. MANIFEST-2, a global, phase 3, randomized, double-blind, active-control study of CPI-0610 and ruxolitinib vs. placebo and ruxolitinib in JAK-inhibitor-naive myelofibrosis patients. Blood 2020;136:43. [Google Scholar]
  • 157. Pemmaraju N, Garcia JS, Potluri J, Holes L, Harb J, Jung P, et al. The addition of navitoclax to ruxolitinib demonstrates efficacy within different high-risk populations in patients with relapsed/refractory myelofibrosis. Blood 2020;136:49–50. [Google Scholar]
  • 158. Harrison CN, Garcia JS, Mesa RA, Somervaille TC, Komrokji RS, Pemmaraju N, et al. Results from a phase 2 study of navitoclax in combination with ruxolitinib in patients with primary or secondary myelofibrosis. Blood 2019;134:671. [Google Scholar]
  • 159. Gerds AT, Vannucchi AM, Passamonti F, Kremyanskaya M, Gotlib JR, Palmer JM, et al. A phase 2 study of luspatercept in patients with myelofibrosis-associated anemia. Blood 2019;134:557. [Google Scholar]
  • 160. Gerds AT, Vannucchi AM, Passamonti F, Kremyanskaya M, Gotlib J, Palmer JM, et al. 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 2020;136:47–48. [Google Scholar]
  • 161. Mascarenhas J, Komrokji RS, Palandri F, Martino B, Niederwieser D, Reiter A, et al. Randomized, single-blind, multicenter phase II study of two doses of imetelstat in relapsed or refractory myelofibrosis. J Clin Oncol 2021;39:2881–92. [DOI] [PubMed] [Google Scholar]
  • 162. Jutzi JS, Kleppe M, Dias J, Staehle HF, Shank K, Teruya-Feldstein J, et al. LSD1 inhibtion prolongs survival in mouse models of MPN by selectively targeting the disease clone. HemaSphere 2018;2:e54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163. Gill H, Yacoub A, Pettit K, Bradley T, Gerds A, Tatarczuch M, et al. A phase 2 study of the LSD-1 inhibitor IMG-7289 (Bomedemstat) for the treatment of advanced myelofibrosis. EHA2021 Virtual Congress Abstract Book. HemaSphere 2021;5:e566. [Google Scholar]
  • 164. Yacoub A, Wang ES, Rampal RK, Borate U, Kremyanskaya M, Ali H, et al. Abstract CT162: Addition of parsaclisib (INCB050465), a PI3Kδ inhibitor, in patients with suboptimal response to ruxolitinib: a phase 2 study in patients with myelofibrosis. Cancer Res 2021;81:CT162. [Google Scholar]
  • 165. Yacoub A, Stouffs M, Zhou F, Assad A. Abstract CT253: a randomized, double-blind, placebo-controlled phase 3 study of parsaclisib plus ruxolitinib in patients with myelofibrosis who have suboptimal response to ruxolitinib. Cancer Res 2021;81:CT253. [Google Scholar]
  • 166. Yacoub A, Erickson-Viitanen S, Zhou F, Assad A. A phase 3, randomized, double-blind, placebo-controlled study of ruxolitinib plus parsaclisib in patients with JAK- and PI3K-inhibitor treatment-naïve myelofibrosis. J Clin Oncol 2021;39:TPS7058. [Google Scholar]
  • 167. Pushpakom S, Iorio F, Eyers PA, Escott KJ, Hopper S, Wells A, et al. Drug repurposing: progress, challenges and recommendations. Nat Rev Drug Discov 2019;18:41–58. [DOI] [PubMed] [Google Scholar]
  • 168. Beaver JA, Amiri-Kordestani L, Charlab R, Chen W, Palmby T, Tilley A, et al. FDA approval: palbociclib for the treatment of postmenopausal patients with estrogen receptor-positive, HER2-negative metastatic breast cancer. Clin Cancer Res 2015;21:4760–6. [DOI] [PubMed] [Google Scholar]
  • 169. Miller JL. FDA approves pioglitazone for diabetes. Am J Health Syst Pharm 1999;56:1698. [DOI] [PubMed] [Google Scholar]
  • 170. Gangat N, Guglielmelli P, Szuber N, Begna KH, Patnaik MM, Litzow MR, et al. Venetoclax with azacitidine or decitabine in blast-phase myeloproliferative neoplasm: a multicenter series of 32 consecutive cases. Am J Hematol 2021;96:781–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171. Mesa RA, Steensma DP, Pardanani A, Li C-Y, Elliott M, Kaufmann SH, et al. A phase 2 trial of combination low-dose thalidomide and prednisone for the treatment of myelofibrosis with myeloid metaplasia. Blood 2003;101:2534–41. [DOI] [PubMed] [Google Scholar]
  • 172. Luo X, Xu Z, Li B, Qin T, Zhang P, Zhang H, et al. Thalidomide plus prednisone with or without danazol therapy in myelofibrosis: a retrospective analysis of incidence and durability of anemia response. Blood Cancer J 2018;8:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173. Shimoda K, Shide K, Kamezaki K, Okamura T, Harada N, Kinukawa N, et al. The effect of anabolic steroids on anemia in myelofibrosis with myeloid metaplasia: retrospective analysis of 39 patients in Japan. Int J Hematol 2007;85:338–43. [DOI] [PubMed] [Google Scholar]
  • 174. Besa EC, Nowell PC, Geller NL, Gardner FH. Analysis of the androgen response of 23 patients with agnogenic myeloid metaplasia: the value of chromosomal studies in predicting response and survival. Cancer 1982;49:308–13. [DOI] [PubMed] [Google Scholar]
  • 175. Cervantes F, Isola IM, Alvarez-Larrán A, Hernández-Boluda J-C, Correa J-G, Pereira A. Danazol therapy for the anemia of myelofibrosis: assessment of efficacy with current criteria of response and long-term results. Ann Hematol 2015;94:1791–6. [DOI] [PubMed] [Google Scholar]
  • 176. Mesa RA, Yao X, Cripe LD, Li CY, Litzow M, Paietta E, et al. Lenalidomide and prednisone for myelofibrosis: Eastern Cooperative Oncology Group (ECOG) phase 2 trial E4903. Blood 2010;116:4436–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177. Tefferi A, Cortes J, Verstovsek S, Mesa RA, Thomas D, Lasho TL, et al. Lenalidomide therapy in myelofibrosis with myeloid metaplasia. Blood 2006;108:1158–64. [DOI] [PubMed] [Google Scholar]
  • 178. Cervantes F, Alvarez-Larrán A, Hernández-Boluda J-C, Sureda A, Torrebadell M, Montserrat E. Erythropoietin treatment of the anaemia of myelofibrosis with myeloid metaplasia: results in 20 patients and review of the literature. Br J Haematol 2004;127:399–403. [DOI] [PubMed] [Google Scholar]
  • 179. Pardanani A, Tefferi A, Masszi T, Mishchenko E, Drummond M, Jourdan E, et al. Updated results of the placebo-controlled, phase III JAKARTA trial of fedratinib in patients with intermediate-2 or high-risk myelofibrosis. Br J Haematol 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180. Lussana F, Cattaneo M, Rambaldi A, Squizzato A. Ruxolitinib-associated infections: a systematic review and meta-analysis. Am J Hematol 2018;93:339–47. [DOI] [PubMed] [Google Scholar]
  • 181. Verstovsek S, Kremyanskaya M, Masacarenhas J, Talpaz M, Harrison C, Rampal RK, et al. Pelabresib (CPI-0610) improved anemia associated withy myelofibrosis: Interim results from MANIFEST Phase 2 Study. EHA2021 Virtual Congress Abstract Book. HemaSphere 2021;5:e566. [Google Scholar]
  • 182. Harrison C, Garcia J, Somervaille T, Foran JM, Verstovsek S, Jamieson C, et al. Navitoclax and ruxolitinib for patients with myelofibrosis and JAK inhibtor experience: response duration in Phase 2 study. EHA2021 Virtual Congress Abstract Book. HemaSphere 2021;5:e566. [Google Scholar]
  • 183. Bose P, Pemmaraju N, Masarova L, Bledsoe SD, Daver N, Jabbour E, et al. Sotatercept (ACE-011) for anemia of myelofibrosis: a phase 2 study. Blood 2020;136:10–11. [Google Scholar]
  • 184. Pemmaraju N, Gupta V, Ali H, Yacoub A, Wang ES, Lee S, et al. A multicenter phase 1/2 clinical trial of tagraxofusp, a CD123-targeted therapy, in patients with poor-risk primary and secondary myelofibrosis. Blood 2020;136:39–40. [Google Scholar]

Articles from Cancer Research are provided here courtesy of American Association for Cancer Research

RESOURCES