Abstract
Development of myelofibrosis (MF) therapeutics has reached fruition as the transformative impact of JAK2 inhibitors in the MPN landscape is complemented/expanded by a profusion of novel monotherapies and rational combinations in the frontline and second line settings. Agents in advanced clinical development span various mechanisms of action (e.g., epigenetic or apoptotic regulation), may address urgent unmet clinical needs (cytopenias), increase the depth/duration of spleen and symptom responses elicited by ruxolitinib, improve other aspects of the disease besides splenomegaly/constitutional symptoms (e.g., resistance to ruxolitinib, bone marrow fibrosis or disease course), provide personalized strategies, and extend overall survival (OS). Ruxolitinib had a dramatic impact on the quality of life and OS of MF patients. Recently, pacritinib received regulatory approval for severely thrombocytopenic MF patients. Momelotinib is advantageously poised among JAK inhibitors given its differentiated mode of action (suppression of hepcidin expression), leading to significant improvements in anemia and OS; momelotinib will likely receive regulatory approval in 2023. An array of other novel agents combined with ruxolitinib, such as pelabresib, navitoclax, parsaclisib, or as monotherapies (navtemadlin) are evaluated in pivotal phase 3 trials. Imetelstat (telomerase inhibitor) is currently evaluated in the second line setting; OS was set as the primary endpoint, marking an unprecedented goal in MF trials, wherein SVR35 and TSS50 at 24 weeks have been typical endpoints heretofore. Transfusion independence may be considered another clinically meaningful endpoint in MF trials given its correlation with OS. Overall, therapeutics are at the cusp of an exponential expansion and advancements that will likely lead to the golden era in MF treatment.
Keywords: Myeloproliferative neoplasms (MPNs), momelotinib, pelabresib, luspatercept, imetelstat
Introduction
Among myeloproliferative neoplasms (MPN), myelofibrosis (MF) is the most aggressive and is characterized by clonal myeloproliferation, bone marrow fibrosis, splenomegaly, worsening anemia, systemic symptoms, and increased production of inflammatory cytokines.1 In addition, MF can transform to the accelerated phase2 and blast phase MPN3,4 or post-MPN acute myeloid leukemia, which are diagnosed when blasts in the peripheral blood/bone marrow are in the range 10–19% and ≥20%, respectively.
Clinical development and regulatory approval (in 2011) of ruxolitinib- a selective JAK1/2 inhibitor targeting the overactivated JAK signal and transducer of transcription (JAK/STAT) pathway- revolutionized treatment of intermediate- and high-risk MF and dramatically improved quality-of-life, splenomegaly and constitutional symptoms in MF patients.5 Ruxolitinib has become the cornerstone treatment for MF, conferring significant prolongation of survival in MF patients6,7 besides other clinical benefits. The survival benefit of ruxolitinib treatment evidenced from clinical data5 was complemented by an analysis of real-world data from patients diagnosed with intermediate- to high-risk MF between 2010 and 2017: the 1-year survival rates for post-approval ruxolitinib-exposed vs. post-approval ruxolitinib-unexposed vs. pre-approval of ruxolitinib unexposed patients were 82.3% vs. 72.5% vs. 55.6%, respectively.7 The tremendous impact of ruxolitinib in prolonging overall survival (OS) of MF patients in the last decade was also demonstrated in another retrospective study of patients stratified in groups by decade (2000–2010 and 2011–2020): the overall survival with ruxolitinib treatment was nine times higher compared to that with other agents during the second decade.6 Notably, the updated multivariate analysis of the real-world ERNEST (European Registry for Myeloproliferative Neoplasms) study, which evaluated the outcomes of 1,010 patients with MF from 13 centers in 5 European countries, demonstrated the significant benefit of ruxolitinib treatment in extending OS compared to hydroxyurea.8 While the survival advantage of ruxolitinib has even been recognized by regulatory bodies (Federal Drug Administration) and is listed as a benefit of ruxolitinib on its label in the US, a few investigators still find this issue controversial.9 The EXPAND study (NCT01317875) showed that ruxolitinib treatment at 10 mg bid elicited meaningful clinical efficacy (reduction of spleen size and symptoms) in patients with MF and baseline platelet counts in the range 50 to <100 × 109/L;10 patients with platelet counts in the former range were included in the EXPAND study because the COMFORT-I and COMFORT-II trials, which compared ruxolitinib efficacy to placebo and best available therapy (BAT), respectively, included patients with platelet counts ≥100×109/L.11,12
Fedratinib, the second JAK2 inhibitor, received regulatory approval for intermediate-2 and high-risk MF in 2019,13,14 and is an option primarily in the post-ruxolitinib setting. Post-hoc analysis of the JAKARTA-2 trial demonstrated spleen volume reduction ≥35% (SVR35) 31% and ≥50% improvement in total symptom score (TSS50) 27%.15 Notwithstanding the notable benefits of ruxolitinib and fedratinib in treatment of MF, disease-related cytopenias that can be exacerbated by JAK-inhibitor induced myelosuppression16- anemia17 and thrombocytopenia18- can develop and are associated with poor prognosis.19,20,21 Patients who discontinued ruxolitinib (primarily due to disease progression, including cytopenias)22,23 or acquired ≥3 non-driver mutations24 had a limited median overall survival (ranging between 11 and 16 months in several studies).25,26,27,28 In February 2022, the field of MPNs was marked by the accelerated regulatory approval of pacritinib, a relatively non-myelosuppressive JAK2 and interleukin-1 receptor-associated kinase 1 (IRAK1) inhibitor, as a treatment for patients with intermediate- or high-risk MF and severe thrombocytopenia (platelet count below 50×109/L).29,30,31,32 Pacritinib’s approval was primarily based on the results of the PERSIST-2 trial (NCT02055781)33 and the PAC203 dose-finding trial (NCT03165734).34 Currently, pacritinib is compared to “physician’s choice” (low dose ruxolitinib, danazol, steroids, hydroxyurea) in the phase 3 PACIFICA trial (NCT03165734) in patients with advanced MF and severe thrombocytopenia.35 Despite the development of pacritinib to treat thrombocytopenic patients with MF, significant areas of unmet need still remain, such as anemia, suboptimal response or resistance to ruxolitinib, modification of disease course, prolongation of survival; and have sparked a plethora of endeavors to develop novel medications and treatment strategies for MF.36,37,38,39,40 Furthermore, in light of the fact that deeper spleen responses with ruxolitinib treatment have been associated with prolonged OS,41,42,43 combination regimens with ruxolitinib may further improve spleen and symptom responses and prolong OS. A collection of novel agents that act through various mechanisms, have been explored as monotherapies and complimentary or synergistic combinations with ruxolitinib.36 In this review, we highlight several new concepts for treatment of MF and the corresponding investigational agents that were recently evaluated (Table 1) or are currently being appraised (Table 2) in the frontline and second line settings, focusing on advanced phase 3 clinical trials.
Table 1.
Investigational Agent | Mechanism of Action | Clinical Trial Identifier and Setting | Enrolled Patients | Phase | SVR35 response | TSS50 response | Anemia response | VAF reduction | BMF reduction |
---|---|---|---|---|---|---|---|---|---|
Concept #1:Combination of anemia therapeutic with a JAK1/2 inhibitor | |||||||||
Luspatercept48 (on a stable dose of ruxolitinib) | Activin receptor IIB ligand trap | ACE-536-MF-001 (NCT03194542) | MF patients who were on a stable dose of ruxolitinib for ≥16 weeks prior to enrollment and required RBC transfusions (Cohort 3B) | 2 | NR | NR | 27% became RBC TI over any12 consecuti ve wks during the first 24 wks; 36% became RBC TI for ≥12 wks over the entire treatment period; 46% had ≥50% reduction in RBC transfusi ons (>4 units) over 12 wks | NR | NR |
Concept #2: Agents in combination with a JAK1/2 inhibitor in the frontline setting | |||||||||
Pelabresib53 (+ Ruxolitinib) | BET inhibitor | MANIFEST (NCT02158858) First-line | MF patients who were JAK inhibitornaive (Arm 3) | 2 | 68%¥ | 56% | 24% had a mean Hb increase ≥1.5 g/dL over 12 wks | NR | 28% of the patient s had ≥ 1 grade reducti on |
Navitoclax58 (+ Ruxolitinib) | Bcl-2/Bcl-xL inhibitor | REFINE (NCT03222609) (Cohort 3) First-line | MF patients who were JAK inhibitornaive and BET inhibitornaive | 2 | 52%¥ 76% at any time post-baseline | 31% 56% at anytime post-baseline | Of those with Hb<10 g/dL or TD; increase of Hb >2 g/dL or TI: 55% | NR | 35% of the patients had a 1 grade reduction at any time post-baseline |
Concept #3:“Add-on” agents to a JAK1/2 inhibitor in the second line setting | |||||||||
Navitoclax66 (+Ruxolitinib) | Bcl-2/Bcl-xL inhibitor | REFINE (NCT03222609)“Add-on” to ruxolitinib (Cohort 1a) | MF patients with suboptimal response to ruxolitinib | 2 | 26.5%¥ | 30% | Of those with Hb<10 g/dL or TD; increas e of Hb ≥2 g/dL or TI: 64% | 46% of the patients had >10% reducti on in VAF of driver mutatio ns | 21% and 33% of the patients had ≥ 1 grade reducti on at 24 wks and at any time, respectively |
Parsaclisib69 (+ Ruxolitinib) | PI3Kδ inhibitor | NCT02718300 “Add-on” to ruxolitinib | MF patients with suboptimal response to ruxolitinib | 3 | 7.1% | 15.6% | NR | NR | NR |
Concept #4: JAK1/2 inhibitor to treat anemia, splenomegaly, and constitutional symptoms in the second line setting | |||||||||
Momelotinib 83 | ACVR 1 and JAK1/2 inhibitor | SIMPLIFY-2 (NCT02101268) Second-line | Anemic patients with MF previously treated with ruxolitinib | 3 | 7%¥ | 26% | RBC TI: 43% at wk 24; Rate of zero transfusions over entire treatment phase: 40% | NR | NR |
Momelotinib 87,88,89 | ACVR 1 and JAK1/2 inhibitor | MOMENTUM (NCT04173494) Second-line | Symptomatic (TSS ≥ 10), anemic (Hb <10 g/dL) MF patients who were previously treated with an approved JAK inhibitor | 3 | 23.1% | 24.6%¥ | RBC TI: 31% at wk 24; zero RBC transfusion rate: 35% | NR | NR |
Concept #5: HDM2 inhibitor in the second line setting | |||||||||
Navtemadlin 91,92,93 | HDM2 inhibitor | BOREAS (NCT03662126) Second-line Phase 2: 240 mg, days 1–7/28-day cycle | Wild type TP53 MF patients who relapsed or are refractory/resistant to JAK inhibitors | 2/3 | Ph. 2: 16%¥ | Ph.2:30% | NR | 34% of the patients had ≥20% VAF decrease in HMR or driver mutations; 29% had VAF<d etection limit | 27% had ≥ 1 grade reduction, and 51% had stable BMF scores |
Concept #6: Telomerase inhibitor to prolong survival in the second line setting | |||||||||
Imetelstat 94 | Telomerase inhibitor | IMbark (NCT02426086) 9.4 mg/kg once every 3 wks |
Patients with intermediate −2 or high-risk MF that relapsed or were refractory to JaK inhibitors | 2 | 10.2%†,§ | 32.2%† | 25% TI for ≥12 wks | 42.1% of the patients had ≥25% reduction in VAF of driver mutations | 40.5% of the patients |
Abbreviations: ACVR1 = activin A receptor type 1; Bcl-2/Bcl-xL = B-cell lymphoma-2/extra-large; BET = bromodomain and extra-terminal; BMF = bone marrow fibrosis; HDM2 = human double minute 2; Hb = hemoglobin; HMR = high molecular risk; JAK = Janus kinase; MF = myelofibrosis; NR = not reported; PI3Kδ = phosphatidylinositol 3-kinase-delta; RBC = red blood cell; SVR35 = spleen volume reduction ≥35% from baseline to week 24; TD = transfusion-dependent; TI = transfusion-independent; TSS50: ≥50% improvement in total symptom score from baseline to week 24; VAF = variant allele frequency; wk = week.
Primary endpoint.
Coprimary endpoints.
Median OS (one of the secondary endpoints) was 29.9 months for the subgroup treated with 9.4 mg/kg of imetelstat.
Table 2.
Investigational Agent | Mechanism of Action | Clinical Trial Identifier and Setting | Enrolled Patients | Phase | Primary Endpoint | Secondary Endpoint(s) |
---|---|---|---|---|---|---|
Concept #1 :Combination of anemia therapeutic with a JAK1/2 inhibitor | ||||||
Luspatercept49 (on a stable dose of ruxolitinib) | Activin receptor IIB ligand trap | INDEPENDENCE (NCT04717414) | Anemic MF patients on a stable dose of ruxolitinib, requiring 4–12 RBC transfusions in the 12 weeks before randomization | 3 | % RBC TI for any 12 consecutive wks during the first 24 wks | % RBC TI for any consecutive 16 wks, ≥50% reduction of transfusion burden by ≥4 units for any 12 consecutive wks and duration of reduction |
Concept #2: Agents in combination with a JAK1/2 inhibitor in the frontline setting | ||||||
Pelabresib56 (+ Ruxolitinib) | BET inhibitor | MANIFEST-2 (NCT04603495) Frontline | MF patients who are JAK inhibitornaive | 3 | SVR35 response | TSS50 response BMF reduction SVR35 duration TSS50 duration PFS, OS, TD to TI at 24 wks |
Navitoclax59 (+ Ruxolitinib) | Bcl-2/Bcl-xL inhibitor | TRANSFORM-1 (NCT04472598) Frontline | MF patients who are JAK in hibitor-naïve a nd BET inhibitor-naïve | 3 | SVR35 response | TSS50, SVR35 duration, anemia response, BMF reduction, OS, LFS |
Parsaclisib64 (+ Ruxolitinib) | PI3Kδ inhibitor | LIMBER-313 (NCT0455106 Frontline | Patients with MF who are JAK inhibitor-naïve | 3 | SVR35 response | Time to first TSS50, OS, SVR35 duration |
Concept #3-“Add- on” agents to a JAK1/2 inhibitor in the second line setting | ||||||
Navitoclax68 (+ Ruxolitinib) | Bcl-2/Bcl-xL inhibitor | TRANSFORM-2 (NCT04468984) “Add-on” to ruxolitinib | MF patients who are refractory/resistant to JAK2 inhibitors | 3 | SVR35 response | TSS50, SVR35 duration, anemia response, OS, LFS, BMF reduction |
Parsaclisib (+ Ruxolitinib) | PI3Kδ inhibitor | LIMBER-304 (NCT04551053) “Add-on” to ruxolitinib | Patients with MF who had suboptimal response to ruxolitinib | 3 | SVR35 response | TSS50, TSS change, time to TSS50, OS |
Navtemadlin72 (+ Ruxolitinib) | HDM2 inhibitor | NCT04485260 “Add-on” to ruxolitinib | TP53-wild type MF patients who had a suboptimal response to ruxolitinib after ≥18 wks, at a stable dose for ≥8 wks | 1b/2 | RP2D of navtemadlin in combination with ruxolitinib | SVR35 response, TSS at any time point, SVR35 duration, TI rate, OS, PFS,LFS |
Enasidenib (+ Ruxolitinib) 77 | IDH2 inhibitor | NCT04281498 | IDH2-mutated, persistent or progressive MF with 4–9% blasts or IDH2-mutated AP/BP-MPN | 2 | Proportion of responders within 6 cycles of combined therapy | Proportion of participants with blast response or any response in 6 months |
Concept #4: JAK1/2 inhibitor to treat anemia, splenomegaly, and constitutional symptoms in the second line setting | ||||||
Momelotinib 87,88,89 | ACVR 1 and JAK1/2 inhibitor | MOMENTUM (NCT04173494) Second-line | Symotonatic (TSS ≥ 10), aremic (Hb <10 g/dL) MF patients who were previously treated with an approved JAK inhibitor | 3 | TSS50 response | Rate of RBC TI patients for ≥12 wks prior to the end of wk 24, SVR35, mean TSS change, zero RBC transfusion rate |
Concept #5: HDM2 inhibitor in the second line setting | ||||||
Navtemadlin 91,93 | HDM2 inhibitor | BOREAS (NCT03662126) Second-line Phase 3: 240 mg, days 1-7/28-day cycle | Wild type TP53 MF patients who relapsed or are refractory/resistant to JAK inhibitors | 2/3 | SVR35 response | TSS50, PFS, OS, best overall SVR35, SVR35 duration |
Concept #6: Telomerase inhibitor to prolong survival in the second line setting | ||||||
Imetelstat 101 | Telomerase inhibitor | IMpactMF (NCT04576156) Second-line | Patients with intermediate-2 or high-risk MF who failed JAK inhibitor therapy | 3 | OS | TSS50, PFS, SVR35, BMF reduction |
Abbreviations: ACVR1 = activin A receptor type 1; AP = accelerated phase; Bcl-2/Bcl-xL = B-cell lymphoma-2/extra-large; BET = bromodomain and extra-terminal; BMF = bone marrow fibrosis; BP = blast phase; HDM2 = human double minute 2; Hb = hemoglobin; JAK = Janus kinase; LFS = leukemia-free survival; MF = myelofibrosis; OS = overall survival; PFS = progression-free survival; PI3Kδ = phosphatidylinositol 3-kinase-delta; RBC = red blood cell; RP2D = recommended phase 2 dose; SVR35 = spleen volume reduction ≥35% from baseline to week 24; TI = transfusion-independent; TSS50: ≥50% improvement in total symptom score from baseline to week 24; wk = week.
Concept #1:Combination of anemia therapeutic with a JAK1/2 inhibitor
One of the cardinal features of MF that affects OS is anemia17,21 and red blood cell (RBC) transfusion dependence, which are associated with substantial clinical burden44 and constitute adverse prognostic factors in MF.45 In a study that was performed at Mayo Clinic, more than 50% of the patients with primary MF were anemic when they were referred to the institution (at diagnosis, within one year of diagnosis and more than 1 year after diagnosis), and approximately one quarter of the patients required RBC transfusions at initial diagnosis;46 nearly all MF patients become anemic over the course of the disease.23 Currently, anemia treatments are not satisfactory, and there is a critical unmet need to improve anemia in MF patients.
Luspatercept is an activin receptor IIB ligand trap enhancing late-stage erythropoiesis that is being studied in MF patients with anemia. This novel class of fusion proteins ameliorates anemia of diverse etiologies by sequestering ligands of the transforming growth factor beta (TGF-β) superfamily that bind to the activin receptor; thus promoting terminal erythroid differentiation and improvement of anemia (via potent inhibition of downstream Smad2/3 signaling).47 Luspatercept recently received regulatory approval for treatment of anemia in β-thalassemia and myelodysplastic syndromes with ring sideroblasts. The phase 2 trial evaluated luspatercept in MF patients who were on a stable dose of ruxolitinib for ≥16 weeks prior to enrollment and required RBC transfusions (NCT03194542, cohort 3B); in cohort 3B of the study, 27% of the patients became RBC transfusion-independent over any 12 consecutive weeks during the first 24 weeks (primary endpoint), 36% of the patients achieved RBC transfusion-independence for 12 consecutive weeks or more when assessing the entire treatment period, and 46% of the patients achieved ≥50% reduction of RBC transfusion burden (≥4 units) over 12 weeks (Table 1).48 Luspatercept is currently evaluated versus placebo in a registrational, randomized phase 3 trial (INDEPENDENCE; NCT04717414; Table 2) in MF patients who are concomitantly receiving ruxolitinib and require RBC transfusions (4–12 transfusions in the 12 weeks immediately prior to randomization).49
Concept #2: Agents in combination with a JAK1/2 inhibitor in the frontline setting
1. Bromodomain and Extra-terminal (BET) Inhibitor (Pelabresib).
Preclinical studies in JAK2-mutated MPN mouse models demonstrated that BET inhibitor proteins (epigenetic “reader” proteins that bind acetylated lysine residues in histones; acetylation of lysine residues in histones is critical in epigenetic control of gene transcription) in combination with ruxolitinib synergistically suppressed inflammatory cytokine production and megakaryocyte infiltration, and eliminated fibrosis.50,51 In addition, JQ1 (BETg inhibitor) acted synergistically with ruxolitinib to induce apoptosis of primary MPN-BP patient-derived cells in vitro, and prolonged survival of an immunodeficient mouse model transplanted with MPN-BP patient-derived blast progenitor cells.52
In the phase 2 MANIFEST trial (NCT02158858), which comprised 3 Arms, pelabresib (selective BET protein inhibitor of BD1 and BD2 bromodomains) was studied in MF patients who were no longer on ruxolitinib and received pelabresib monotherapy (Arm 1); patients with a suboptimal response to ruxolitinib who received pelabresib as an “add-on” to a stable dose of ruxolitinib (Arm 2); and JAK inhibitor naïve patients who received the combination of pelabresib and ruxolitinib from the start (Arm 3). Patients in Arms 1 and 2 of the study were stratified in transfusion-independent and transfusion-dependent cohorts.
In Arm 3 (frontline) of the MANIFEST study, pelabresib in combination with ruxolitinib elicited SVR35 and TSS50 from baseline in 68% (median change −50%) and 56% (median change −59%) of the JAK inhibitor-naïve MF patients, respectively, at week 24 (Table 1); moreover, 24% of the patients had a mean hemoglobin increase of 1.5 g/dL or more compared to baseline over 12 weeks (without RBC transfusions).53 The spleen and symptom response rates that were recorded in Arm 3 were considerably higher than the historical data reported for ruxolitinib monotherapy in phase 2 and 3 trials; for example, in the COMFORT-1 trial, the SVR35 and TSS50 rates were 42% and 46%, respectively54). In the MANIFEST study, more than half of the patients had primary MF and harbored high molecular risk mutations; notably, clinical efficacy was noted regardless of the mutational profiles, which included high molecular risk mutations, such as ASXL1.55 In addition, bone marrow fibrosis decreased by ≥1 grade compared to baseline in 31% of the patients, in Arm 3.53 Based on the notable findings of pelabresib in combination with ruxolitinib in the frontline setting of the MANIFEST trial (Arm 3), the combination of pelabresib with ruxolitinib versus placebo and ruxolitinib is currently further evaluated in the global, randomized registrational phase 3 MANIFEST-2 trial (NCT04603495) in JAK inhibitor-naïve patients with intermediate or high-risk MF for control of splenomegaly and symptom burden (Table 2).56
2. BcL-xl Inhibitor (Navitoclax).
Navitoclax is an orally bioavailable small molecule that potently inhibits the anti-apoptotic B-cell lymphoma-2/extra-large (Bcl-2/Bcl-xL) family of proteins (primarily Bcl-xL). Preclinical studies demonstrated synergism between ruxolitinib and the non-clinical analog of navitoclax (ABT-737) in inducing a significantly higher apoptotic rate of leukocytes in MF patients.57 In a multicenter, open-label phase 2 study in JAK inhibitor-naïve and BET inhibitor-naïve patients with MF (REFINE, NCT03222609; Cohort 3), treatment with navitoclax in combination with ruxolitinib resulted in SVR35 and TSS50 in 52% and 31% of the patients at week 24, respectively; and improved anemia and bone marrow fibrosis by ≥1 grade in 55% and 35% of the patients (at any time during treatment), respectively.58 SVR35 and TSS50 were noted in 76% and 56% of the patients, respectively, compared to baseline at any time during the study.58 Grade 3 or higher thrombocytopenia was noted in 31% of the patients treated with the combination (platelet counts were ≤150 ×109/L and >150 ×109/L at baseline for patients receiving 100 mg and 200 mg daily, respectively). Navitoclax in combination with ruxolitinib is currently being evaluated in the placebo-controlled phase 3 trial TRANSFORM-1 (NCT04472598) in JAK- and BET-inhibitor-naïve patients with intermediate-2 and high-risk MF.59 The primary endpoint is SVR35 at week 24, and TSS50 at week 24 is included among the secondary endpoints of the trial (Table 2).
3. PI3K Inhibitor (Parsaclisib).
Parsaclisib is a potent, highly selective inhibitor of phosphatidylinositol 3-kinase (PI3K)-delta, an isoform that is important in hematologic malignancies.60 Preclinical studies showed synergism of PI3K and JAK2 inhibitors,61,62 thereby supporting the clinical assessment of parsaclisib in combination with ruxolitinib in MF patients.63 To this effect, parsaclisib is evaluated in combination with ruxolitinib in the placebo-controlled phase 3 trial LIMBER-313 (NCT04551066) in MF patients (with a DIPSS risk category of at least intermediate-1) who are JAK- and PI3K- inhibitor naïve.64 The primary endpoint is evaluation of SVR at week 24; secondary objectives include evaluation of TSS, OS, and duration of spleen response among others.
Concept #3: “Add-on” agents to a JAK1/2 inhibitor in the second line setting
1. BcL-xl Inhibitor (Navitoclax).
In preclinical models, resistance of JAK2 V617F-expressing MPN cells to ruxolitinib was overcome by treatment with ABT-737, the non-clinical analog of navitoclax.65 In the open-label, phase 2 REFINE trial (NCT03222609) that was conducted in patients who progressed or had suboptimal response to ruxolitinib monotherapy, the addition of navitoclax to ruxolitinib resulted in SVR35 and TSS50 in 26.5% and 30% of MF patients, respectively, at week 24 (Cohort 1a); and 41% of the patients achieved SVR35 at any time on the study (Table 1).66 In the same trial, 33% of the evaluable patients showed improvement in bone marrow fibrosis (1–2 grades), and 64% of the patients demonstrated an anemia response (improvement in Hb of ≥2 g/dL).66 Notably, 58% of the patients harbored high molecular risk mutations, and 52% had ≥3 mutations.66 Post-hoc analysis of molecular biomarkers demonstrated that spleen responses were noted regardless of the number of mutated genes and high molecular risk mutations (ASXL1, SRSF2, EZH2, U2AF1, IDH1) at baseline.67 Variant allele frequencies (VAF) of the driver genes JAK2 and CALR were reduced by ≥20% compared to baseline in 23% of the patients; patients with VAF reduction of ≥20% and improvement in bone marrow fibrosis of ≥1 grade exhibited better OS (median not reached) compared to those who did not achieve a reduction in VAF or bone marrow fibrosis (median OS 28.5 months).67The combination of navitoclax with ruxolitinib is currently being evaluated vs. BAT in the randomized phase 3 trial TRANSFORM-2 (NCT04468984) in patients with intermediate-2 and high-risk MF who relapsed or are refractory to JAK2 inhibition; the primary endpoint of the trial is SVR35 at week 24.68
2. PI3K Inhibitor (Parsaclisib).
Besides the frontline setting, parsaclisib was assessed as an “add-on” agent to ruxolitinib in MF patients with suboptimal response to ruxolitinib22 in a phase 2 trial (NCT02718300).69 The final results of the phase 2 trial demonstrated that 28.6% and 7.1% of the patients achieved SVR ≥25% and ≥35%, respectively, at 24 weeks, on a daily dosing schedule of parsaclisib, which were more efficacious compared to daily/weekly dosing in inducing SVR and TSS reduction.69 The combination of ruxolitinib and parsaclisib (as “add on”) compared to ruxolitinib and placebo will be further evaluated in the phase 3 trial LIMBER-304 (NCT04551053) in MF patients who had a suboptimal response to ruxolitinib monotherapy (Table 2).
3. HDM2 Inhibitor (Navtemadlin).
Navtemadlin (formerly KRT-232) is a first-in-class, potent, bioavailable inhibitor of human double minute 2 (HDM2), a key negative regulator of the tumor suppressor p53. HDM2 is overexpressed in MF CD34+ hematopoietic/progenitor cells compared to normal CD34+ cells,70 and TP53 mutations are uncommon in chronic phase MF,71 thus HDM2 inhibition is a rational target in MF. Accordingly, navtemadlin restores p53 function and mediates apoptosis of neoplastic cells. Presently, navtemadlin is evaluated as an “add-on” to ruxolitinib in an open-label, global phase 1b/2 study (NCT04485260) in TP53 wild-type MF patients with a suboptimal response to ruxolitinib monotherapy after ≥18 weeks of treatment on a stable dose in the 8 weeks prior to study entry (Table 2).72 In the first part of the trial, the maximum tolerated dose of navtemadlin in combination with ruxolitinib will be determined; in the dose expansion phase, the maximum tolerated dose will be combined with a stable dose of ruxolitinib to evaluate the safety and efficacy of the regimen (phase 2).72
4. IDH2 Inhibitor (Enasidenib).
Enasidenib is an IDH2 inhibitor that received regulatory approval for treatment of IDH2-mutated patients with relapsed/refractory acute myeloid leukemia (AML) and is currently being evaluated in combination with other regimens for AML.73 Notably, enasidenib in combination with ruxolitinib demonstrated synergistic activity in double-mutant IDH2/JAK2 V617F MPN and blast phase MPN patient-derived cells.74 Treatment with enasidenib-based combinations75 or enasidenib monotherapy76 elicited significant responses, including complete hematological and molecular remissions, negative measurable residual disease, and prolonged survival, in small groups of IDH2-mutated patients with blast phase and accelerated/blast phase MPN. The combination of enasidenib with ruxolitinib is currently being evaluated in patients with IDH2-mutated accelerated/blast phase MPN or chronic-phase MF (4–9% circulating blasts) in a phase 2 trial (NCT04281498; Table 2); preliminary results of the aforementioned trial demonstrated an overall response rate of 50% in the evaluable cohort (2 patients achieved complete remission).77
Concept #4: Unique JAK1/2 inhibitor to improve anemia, splenomegaly, and constitutional symptoms in the second line setting
Besides selectively inhibiting JAK1/2 and thereby treating splenomegaly and constitutional symptoms, momelotinib has a differentiated mode of action because it also suppresses expression of hepcidin, the master regulator of iron metabolism; hepcidin suppression occurs by inhibiting the type 1 activin receptor (ACVR1) or activin receptor-like kinase-2 (ALK2), thereby restoring iron homeostasis, stimulating erythropoiesis, and leading to a remarkable suite of anemia benefits in MF patients.78 In particular, momelotinib inhibits the primary bone morphogenetic protein 6 (BMP6)/ACVR1/SMAD1/5/8 iron-sensing pathway and the inflammation-driven IL-6/STAT3 axis79− two pro-erythropoietic pathways controlling expression of the hepatic-secreted hormone hepcidin. Hepcidin is elevated in MF patients as a result of aberrant hyperactivation of the BMP6-stimulated kinase ACVR1/ALK2 (BMP6 receptor) signaling and inflammatory cytokine signaling via IL-6, which is also elevated in MF patients.78 Preclinical studies in a rat model of anemia of chronic disease demonstrated a dose-dependent decrease in ACVR1-mediated hepcidin expression following momelotinib administration, mediated via selective inhibition of ACVR1/ALK2 and IL-6-induced pSTAT3.79 In the rat model, inhibition of hepcidin expression resulted in increased release of stored iron from the reticuloendothelial system (via the hepcidin-ferroportin axis), higher circulating iron concentrations, and thereby stimulated erythropoiesis.79
In accordance with these preclinical findings and the pro-erythropoietic mechanism of momelotinib, 41 transfusion-dependent MF patients demonstrated declining serum hepcidin levels versus baseline over 24 weeks of treatment with momelotinib in a translational biology phase 2 study.80 In this study, 41% of the patients achieved transfusion-independence for 12 weeks or more at any time during the study, including 34% of the patients who became transfusion-independent by week 24.80 Analogous marked and sustained transfusion-independence benefits were noted in a total of 558 MF patients who participated in the two phase 3 SIMPLIFY trials and were treated with momelotinib, besides experiencing improvements in splenomegaly and constitutional symptoms. In the SIMPLIFY-1 trial, 66.5% of the JAK-inhibitor naïve patients participating in the momelotinib arm achieved or maintained transfusion-independence versus 49.3% in the ruxolitinib arm at week 24.81 Notably, the median duration of transfusion independence was not reached with momelotinib in the SIMPLIFY-1 trial after follow-up of ≥3 years.82 In the SIMPLIFY-2 trial, 43% of the second-line MF patients achieved or maintained transfusion-independence in the momelotinib arm versus 21% in the control arm (BAT), which primarily involved ruxolitinib (89%), at week 24 (Table 1).83 Furthermore, according to the findings of a zero-inflated negative binominal model, the odds of momelotinib-treated patients remaining transfusion-independent were 9.69 times higher versus ruxolitinib-treated patients in the SIMPLIFY-1 trial; and a patient on momelotinib had an 83% chance of not receiving RBC transfusions versus 34% if treated with ruxolitinib.84,85 Moreover, the hazard ratio for transfusion of an RBC unit for patients treated with momelotinib was about one-half as compared to patients receiving ruxolitinib.85 Importantly, momelotinib showed robust OS benefits in both frontline and ruxolitinib-treated MF patients who crossed over to momelotinib after 24-weeks in both the SIMPLIFY-1 and SIMPLIFY-2 trials.82 In particular, in the SIMPLIFY-2 trial, the patients who crossed over to momelotinib after randomization to BAT (ruxolitinib 89%) for 24 weeks had a median OS of 37.5 months, and the patients who were initially randomized to momelotinib had a median OS of 34.3 months;82 the aforementioned median OSs noted in SIMPLIFY-2 compare favorably with the median OS reported for patients who discontinued ruxolitinib.25,26,27,28 In the SIMPLIFY-1 trial, the median OS was not reached, and the 5-year survival probability was ~55% in both arms.82,86 In SIMPLIFY-2, the median OS was ~3 years and the 2-year survival rate was 66% in the momelotinib arm and 61% in the BAT to momelotinib cross-over arm, with extended momelotinib treatment.86 Notably, analysis of the data from the SIMPLIFY-1 and SIMPLIFY-2 trials demonstrated an association of transfusion independence at baseline with prolonged OS in MF patients (HR = 0.474, P =0.0001 in SIMPLIFY-1; HR = 0.226, P = 0.0005 in SIMPLIFY-2).86 Univariate and multivariate analyses of the SIMPLIFY-1 trial data showed that achievement of transfusion-independence at 24 weeks in JAK inhibitor-naïve MF patients randomized to momelotinib was strongly associated with prolonged OS (HR = 0.323, P <0.0001; HR = 0.311, P <0.0001, respectively); in SIMPLIFY-1, the 3-year survival rate for patients who achieved transfusion independence was 77.2% vs. 51.6% for transfusion independence non-responders.86 The aforementioned findings indicate that transfusion independence at week 24 may be a reasonable surrogate endpoint to predict OS in JAK inhibitor-naïve patients treated with momelotinib.86 This finding may expand the primary endpoints of clinical trials for MF to include transfusion-independence beyond SVR35 and TSS50 at week 24, which have been the usual endpoints for JAK inhibitors heretofore. Interestingly, a recent targeted literature review of 15 publications that reported on anemia and RBC transfusion-related outcomes of MF patients participating in clinical trials demonstrated the highest improvement in RBC transfusion-independence rates with momelotinib compared to the other JAK inhibitors (ruxolitinib, fedratinib, pacritinib).44
Notable results showing the superior efficacy of momelotinib in comparison to danazol were recently reported in the pivotal, randomized, double-blind phase 3 trial (MOMENTUM; NCT04173494),87 in JAK-inhibitor treated (195 and 9 patients were treated with ruxolitinib and fedratinib, respectively), symptomatic (TSS≥10) and anemic (Hb <10 g/dL) patients with intermediate- or high-risk MF (and platelet counts ≥25×109/L).88 Danazol was selected as the comparator because it is recommended for treatment of anemia in the guidelines of the National Comprehensive Cancer Network and the European Society of Medical Oncology.78 In the MOMENTUM trial, momelotinib met both the primary (TSS50 response from baseline at week 24) and key secondary endpoints. In particular, the TSS50 response rate was 24.6% for the cohort treated with momelotinib (Table 1) vs. 9.2% for danazol. The respective secondary endpoints, sequentially assessed, for momelotinib and danazol were the following at week 24 (rates): RBC transfusion independence (30.8% vs. 20.0%), SVR ≥25% from baseline (40.0% vs. 6.2%), TSS change from baseline (−9.36 vs. −3.13), SVR ≥35% from baseline (23.1 vs. 3.1%), and zero units transfused compared to baseline (35.4% vs. 16.9%).88 Interim analysis of the data collected in the MOMENTUM trial demonstrated that patients who achieved transfusion-independence with momelotinib at 24 weeks had prolonged OS compared to the patients who remained transfusion-dependent (HR = 0.15, P = 0.0364), suggesting that transfusion-independence at week 24 could be a potential surrogate endpoint to predict improved OS in MF patients.89 Furthermore, data analysis of the broader thrombocytopenic subgroups (baseline platelet counts <150 × 109/L, <100 × 109/L, and <50 ×109/L) at 24 weeks demonstrated the superiority of momelotinib vs. danazol regarding symptom and spleen responses, RBC transfusion independence rate; and favorable OS.88,90 Collectively, the results of the MOMENTUM trial confirmed that momelotinib is a highly promising treatment for MF patients who are RBC transfusion-dependent and will likely result in regulatory approval of momelotinib in the near future.39,78,85 Notably, in August 2022, the Federal Drug Administration accepted a new drug application for momelotinib as a treatment for MF patients with anemia.
Concept #5: HDM2 inhibitor in the second line setting
In a phase 2 study, navtemadlin (administered once daily 240 mg, days 1–7/28-day cycle) demonstrated encouraging clinical efficacy (16% and 30% of the patients achieved SVR35 and TSS50, respectively; and CD34+ cells were reduced by 87% in the peripheral blood at week 24)91 and tolerability in TP53-wild type MF patients who had failed ruxolitinib treatment (median prior treatments were 2) and had poor prognosis. In this study, a notable correlation was found between SVR responses and ≥20% VAF decrease in high molecular risk (ASXL1, EZH2, IDH1/2, SRSF2, U2AF1) or driver (JAK2, CALR, MPL) mutations (34% of evaluable patients; 29% had VAF below the detection limit), lower peripheral CD34+ cell counts, improved bone marrow fibrosis scores, and reduction in the serum levels of the inflammatory cytokine TNFα.92 On the basis of the phase 2 study results, navtemadlin (dosing: 240 mg, days 1–7/28-day cycle) is being assessed in comparison to BAT (chemotherapy, hydroxyurea, and supportive care, excluding JAK inhibitors) in a global phase 3 trial (BOREAS, NCT03662126) in MF patients who are refractory or resistant to JAK inhibitors (Table 2).93 In this trial, the primary endpoint is the proportion of patients who reach SVR35 compared to baseline at week 24; the secondary endpoints are progression-free survival, OS, duration of spleen response; and the proportion of patients who achieve TSS50 and RBC transfusion-independence at 24 weeks.93
Concept #6: Telomerase inhibitor to prolong survival in the second line setting
Imetelstat is a first-in-class potent telomerase inhibitor that was evaluated in two doses (4.7 and 9.4 mg/kg, administered intravenously once every three weeks) in patients with intermediate-2 or high-risk MF relapsed/refractory to JAK inhibitors in the phase 2 study IMbark (NCT02426086).94 Importantly, a notable median OS (one of the secondary endpoints) of 29.9 months was noted with the higher dose of imetelstat (9.4 mg/kg); furthermore, 40.5% of the patients treated with 9.4 mg/kg of imetelstat demonstrated improvement in bone marrow fibrosis by ≥1 grade, the VAFs of the 3 driver mutations (JAK2 V617F, CALR, MPL) decreased by ≥ 25% in 42.1% of evaluable patients compared to baseline, and 32.2% of the patients experienced TSS50 at week 24 (Table 1).94 In this study, the reduced telomerase activity and lower expression levels of human telomerase reverse transcriptase (hTERT) correlated with spleen and symptom responses and longer median OS. Also, prolonged OS was noted in patients who had ≥20% decrease in VAF compared to patients without VAF reduction; dose-dependent reductions of mutation burden (≥20%) compared to baseline by imetelstat correlated with higher spleen (12.3% vs. 3%) and symptom (31.3% vs. 24.2%) responses, improvement in bone marrow fibrosis (54.4% vs. 21.5%), and prolongation of OS (31.6 vs. 21.5 months).95 Importantly, the higher dose of imetelstat conferred a survival benefit that compared favorably with matched historical data for treatment with BAT; in particular, analysis of the data from 59 MF patients treated with 9.4 mg/kg of imetelstat in the IMbark trial and 38 propensity score-matched MF patients treated with BAT (after JAK inhibitor failure) from real world data collected at a major academic center demonstrated the significant survival benefit that imetelstat conferred vs. BAT (30 vs. 12 months, respectively).96 Notably, higher-risk “triple-negative” MF patients (who are known to have a poor prognosis97,98,99 exhibited superior OS and clinical outcomes compared to the non “triple-negative” cohort.100 On the basis of the promising results noted in the IMbark trial, the international, registrational phase 3 trial (IMpactMF; NCT04576156) is underway to assess the OS (primary endpoint) advantage− an unprecedented endpoint in MF clinical trials− that imetelstat (at 9.4 mg/kg as an intravenous infusion every 21 days) compared to BAT (excluding JAK inhibitors) may confer to intermediate-2 or high-risk MF patients who are refractory to JAK-inhibitors (Table 2).101 Spleen and symptom responses at week 24, progression-free survival and reduction of bone marrow fibrosis are included in the secondary objectives; molecular responses and other biomarkers are included in the exploratory objectives of the trial.101
Future Outlook
Despite the transformative impact of JAK inhibitors in the MPN landscape and their pivotal role in MF treatment,102 there are limitations in their activity. The recent regulatory approval of pacritinib for MF patients with severe thrombocytopenia and the plausible approval of momelotinib for MF patients with anemia78,103 in the near future are welcome developments in the therapeutic landscape of MF. Several promising novel agents with various mechanisms of action beyond the JAK-STAT pathway, administered as monotherapies or in combination with ruxolitinib, have been in advanced clinical development, in the frontline and second line settings;36,104 these strategies are poised to expand the arsenal of MF therapeutics with novel disease-modifying agents that complement or expand the clinical benefits of ruxolitinib. For example, novel agents in clinical development are aimed to increase the depth and duration of spleen and symptom responses via ruxolitinib-based synergistic combinations; improve other aspects of the disease besides splenomegaly and constitutional symptom burden (for example, decrease driver mutation burden); address unmet therapeutic needs of MF patients (for example, anemia, resistance to ruxolitinib); prolong survival beyond ruxolitinib; modify the proclivity of the disease; and thwart leukemic transformation.2,3,4 In clinical trials assessing promising investigational agents and strategies for treatment of MF, new endpoints beyond the usual ones (SVR35 and TSS50 at week 24), such as achievement of RBC transfusion independence and OS, may be considered.105,106 Notably, deeper spleen responses (for example, with ruxolitinib treatment41,42,43) and achievement of RBC transfusion independence (for example, with momelotinib treatment86,89) have been correlated with increased OS.
An array of novel agents, such as BET inhibitors (pelabresib), BcL-xl inhibitors (navitoclax) and PI3K inhibitors (parsaclisib) in the frontline and the second-line settings are promising agents that may be paired with a JAK inhibitor to benefit certain cohorts of patients. The extent of cytopenias, driver/non-driver mutations and allele burden influence the phenotype and could potentially guide the choice of JAK inhibitors for first and second-line treatment.30,107 or targeted treatments (for example, IDH2 inhibitors for treatment of IDH2-mutated patients with advanced myelofibrosis.75,76,77 Novel immunotherapies, such as neoepitope-directed vaccines and monoclonal antibodies directed against mutant-driven MPNs (CALR exon 9 frameshift and JAK2V617F mutations) are explored in preclinical studies and in clinical trials in patients with MPNs (for example, trials NCT03566446, NCT05444530); both CALR exon 9 and JAK2V617 driver mutations, which lead to constitutive activation of the JAK/STAT pathway and malignant cell proliferation, have been reported to be immunogenic and are promising targets for immunotherapy (for example, mCALR vaccine treatments) in MPN patients harboring these mutations..108,109,110 111,112,113,114 Furthermore, monotherapy with imetelstat or navtemadlin may be particularly suited for treatment of high-risk MF patients in the second line setting who do not require RBC transfusions.39 The aforementioned treatment options could result in personalized treatment of MF patients that will take into consideration the hematological and molecular profiles and result in optimum responses and prolonged survival in the near future. Following JAK inhibitors as the sole medications for MF treatment heretofore, clinical development of many promising agents36 is a welcome advancement in the field.
Funding
This work was supported, in part, by the MD Anderson Cancer Center Support Grant P30 CA016672 from the National Cancer Institute (National Institutes of Health).
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Bose P, Masarova L, Amin HM, Verstovsek S. Philadelphia chromosome-negative myeloproliferative neoplasms (Chapter 6) In: The MD Anderson Manual of Medical Oncology, Kantarjian HM, Wolff RA, Rieber AG. Eds., 4th edition, McGraw-Hill Education. 2022, pp. 119–162. [Google Scholar]
- 2.Shahin OA, Chifotides HT, Bose P, Masarova L, Verstovsek S. Accelerated phase of myeloproliferative neoplasms. Acta Haematologica 2021;144(5):484–499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Pasca S, Chifotides HT, Verstovsek S, Bose P. Mutational landscape of blast phase myeloproliferative neoplasms (BP-MPN) and antecedent MPN. Int. Rev. Cell Mol. Biol 2022;366:83–124. [DOI] [PubMed] [Google Scholar]
- 4.Dunbar AJ, Rampal RK, Levine R. Leukemia secondary to myeloproliferative neoplasms. Blood 2020;136(1):61–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Verstovsek S, Gotlib J, Mesa RA, Vannucchi AM, Kiladjian JJ, Cervantes F, Harrison CN, Paquette R, Sun W, Naim A, Langmuir P, Dong T, Gopalakrishna P, Gupta V. Long-term survival in patients treated with ruxolitinib for myelofibrosis: COMFORT-I and -II pooled analyses. J. Hematol Oncol 2017;10(1):156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Masarova L, Bose P, Pemmaraju N, Daver NG, Sasaki K, Chifotides HT, Zhou L, Kantarjian HM, Estrov Z, Verstovsek S. Improved survival of patients with myelofibrosis in the last decade: Single-center experience. Cancer 2022;128(8):1658–1665. [DOI] [PubMed] [Google Scholar]
- 7.Verstovsek S, Parasuraman S, Yu J, Shah A, Kumar S, Xi A, Harrison C. Real-world survival of US patients with intermediate- to high-risk myelofibrosis: impact of ruxolitinib approval. Ann Hematol. 2022;101(1):131–137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Guglielmelli P Ghirardi A, Carobbio A et al. , Impact of ruxolitinib on survival of patients with myelofibrosis in the real world: update of the ERNEST study. Blood Adv. 2022;6(2):373–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Barosi G Gale RP. Does ruxolitinib really prolong survival in individuals with myelofibrosis? The never-ending story. Blood Adv. 2022;6(7):2331–2333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Guglielmelli P, Kiladjian J-J, Vannucchi AM, Duan M, Meng H, Pan L, He G, Verstovsek S, et al. Efficacy and safety of ruxolitinib in patients with myelofibrosis and low platelet count (50 × 109/L to <100 × 109/L) at baseline: the final analysis of EXPAND. Ther Adv Hematol. 2022. Sep 10;13:20406207221118429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366(9):799–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Harrison C, Kiladjian JJ, Al-Ali HK, et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med. 2012;366(9):787–798. [DOI] [PubMed] [Google Scholar]
- 13.Harrison CN, Schaap N, Vannucchi AM, Kiladjian JJ, Jourdan E, Silver RT, Schouten HC, Passamonti F, Zweegman S, Talpaz M, Verstovsek S, Rose S, Shen J, Berry T, Brownstein C, Mesa RA. 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(6):594–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Harrison C, Kiladjian J, Verstovsek S. et al. Overall and progression-free survival in patients treated with fedratinib as first-line myelofibrosis (MF) therapy and after prior ruxolitinib (RUX): results from the JAKARTA and JAKARTA-2 trials. HemaSphere 2021;5:S203. [Google Scholar]
- 15.Harrison CN, Schaap N, Vannucchi AM. 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(6):594–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bose P, Verstovsek S. Management of myelofibrosis-related cytopenias. Curr. Hematol. Malig. Rep 2018;13(3):164–172. [DOI] [PubMed] [Google Scholar]
- 17.Naymagon L, Mascarenhas J. Myelofibrosis-related anemia: current and emerging therapeutic strategies. HemaSphere 2017;1(1):e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Sastow D, Mascarenhas J, Tremblay D. Thrombocytopenia in patients with myelofibrosis: pathogenesis, prevalence, prognostic impact, and treatment. Clin. Lymphoma Myeloma Leuk 2022;22(7):e507–e520. [DOI] [PubMed] [Google Scholar]
- 19.Marcellino BK, Verstovsek S, Mascarenhas J. The myelodepletive phenotype in myelofibrosis: clinical relevance and therapeutic implication. Clin. Lymphoma Myeloma Leuk 2020;20:415–421. [DOI] [PubMed] [Google Scholar]
- 20.Masarova L, Alhuraiji A, Bose P, Daver N, Pemmaraju N, Cortes J, Pierce S, Kantarjian H, Verstovsek S. Significance of thrombocytopenia in patients with primary and post-essential thrombocythemia/polycythemia vera myelofibrosis. Eur. J. Haematol 2018;100(3):257–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Elena C, Passamonti F, Rumi E, et al. Red blood cell transfusion-dependency implies a poor survival in primary myelofibrosis irrespective of IPSS and DIPSS. Haematologica 2011;96(1):167–170. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Bose P, Verstovsek S. SOHO State of the Art Updates and Next Questions: Identifying and treating “progression” in myelofibrosis. Clin Lymphoma Myeloma Leuk. 2021;21(10):641–649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Scherber RM, Mesa R. Management of challenging myelofibrosis after JAK inhibitor failure and/or progression. Blood Reviews 2020;42:100716. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Newberry KJ, Patel K, Masarova L, Luthra R, Manshouri T, Jabbour E. et al. Clonal evolution and outcomes in myelofibrosis after ruxolitinib discontinuation. Blood 2017;130(9):1125–1131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Palandri F, Breccia M, Bonifacio M, Polverelli N, Elli EM, Benevolo G. et al. Life after ruxolitinib: Reasons for discontinuation, impact of disease phase, and outcomes in 218 patients with myelofibrosis. Cancer 2020;126(6):1243–1252. [DOI] [PubMed] [Google Scholar]
- 26.Mascarenhas J, Mehra M, He J, Potluri R, Loefgren C. Patient characteristics and outcomes after ruxolitinib discontinuation in patients with myelofibrosis. J. Med. Econ 2020;23(7):721–727. [DOI] [PubMed] [Google Scholar]
- 27.Schain F, Vago E, Song C, et al. Survival outcomes in myelofibrosis patients treated with ruxolitinib: a population-based cohort study in Sweden and Norway. Eur. J. Haematol 2019;103:614– 619. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mascarenhas JO, Verstovsek S. The clinical dilemma of JAK inhibitor failure in myelofibrosis: Predictive characteristics and outcomes. Cancer 2022;128(14):2717–2727. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Verstovsek S, Mesa R, Talpaz M, et al. Retrospective analysis of pacritinib in patients with myelofibrosis and severe thrombocytopenia. Haematologica 2022;107(7):1599–1607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Tremblay D, Mesa R, Scott B, Buckley S, Torres-Roman K, Verstovsek S, Mascarenhas J. Pacritinib demonstrates spleen volume reduction in patients with myelofibrosis independent of JAK2 V617F allele burden. Blood Adv. 2020;4(23):5929–5935. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Venugopal S, Mascarenhas J. The Odyssey of pacritinib in myelofibrosis. Blood Adv. 2022;6(16):4905–4913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Mascarenhas J Pacritinib for the treatment of patients with myelofibrosis and thrombocytopenia. Exp. Review Hematol 2022;15(8):671–684. [DOI] [PubMed] [Google Scholar]
- 33.Mascarenhas J, Hoffman R, Talpaz M, Gerds AT, Stein B, Gupta V, Szoke A, Drummond M, Pristupa A, Granston T, Daly R, Al-Fayoumi S, Callahan JA, Singer JW, Gotlib J, Jamieson C, Harrison C, Mesa R, Verstovsek S. Pacritinib vs best available therapy, including ruxolitinib, in patients with myelofibrosis: A randomized clinical trial. JAMA Oncol. 2018;4(5):652–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Gerds AT, Savona MR, Scott BL, Talpaz M, Egyed M, Harrison C, et al. Determining the recommended dose of pacritinib: Results from the PAC203 dose-finding study in advanced myelofibrosis. Blood Adv. 2020;4(22):5825–5835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Mascarenhas J, Gerds AT, Kiladjian J-J, et al. PACIFICA: A randomized, controlled phase 3 study of pacritinib versus physician’s choice in patients with primary or secondary myelofibrosis and severe thrombocytopenia. Blood 2022;140(Suppl. 1):9592–9594. [Google Scholar]
- 36.Chifotides HT, Bose P, Masarova L, Pemmaraju N, Verstovsek S. SOHO State of the Art Updates and Next Questions: Novel therapies in development for myelofibrosis. Clin. Lymphoma Myeloma Leuk 2021;22(4):210–223. [DOI] [PubMed] [Google Scholar]
- 37.Bose P, Masarova L, Verstovsek S. Novel concepts of treatment for patients with myelofibrosis and related neoplasms. Cancers (Basel) 2020;12(10):2891. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Bose P, Verstovsek S. JAK inhibition for the treatment of myelofibrosis: Limitations and future perspectives. HemaSphere 2020;4(4):e424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Tremblay D, Mesa R. Momelotinib for treatment of myelofibrosis with anemia. Future Oncol. 2022;18(20):2559–2571. [DOI] [PubMed] [Google Scholar]
- 40.Kuykendall AT, Komrokji RS. JAK be nimble: reviewing the development of JAK inhibitors and JAK inhibitor combinations for special populations of patients with myelofibrosis. J. Immunother. Precis. Oncol 2021;4(3):129–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Verstovsek S, Kantarjian HM, Estrov Z, et al. Long-term outcomes of 107 patients with myelofibrosis receiving JAK1/JAK2 inhibitor ruxolitinib: survival advantage in comparison to matched historical controls. Blood 2012;120(6):1202–1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Vannucchi AM, Kantarjian HM, Kiladjian JJ, et al. A pooled analysis of overall survival in COMFORT-I and COMFORT-II, 2 randomized phase III trials of ruxolitinib for the treatment of myelofibrosis. Haematologica 2015;100:1139–1145. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Miller CB, Komrokji RS, Mesa RA, et al. Practical measures of clinical benefit with ruxolitinib therapy: An exploratory analysis of COMFORT-I. Clin. Lymphoma Myeloma Leuk 2017;17:479–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Gerds AT, Bose P, Hobbs GB, et al. Treating anemic patients with myelofibrosis in the new Janus kinase inhibitor era: Current evidence and real-world implications. HemaSphere 2022;6(10):e778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Nicolosi M, Mudireddy M, Lasho TL, et al. Sex and degree of severity influence the prognostic impact of anemia in primary myelofibrosis: analysis based on 1109 consecutive patients. Leukemia 2018;32:1254–1258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Tefferi A, Lasho TL, Jimma T, et al. One thousand patients with primary myelofibrosis: The Mayo Clinic experience. Mayo Clin. Proc 2012;87:25–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Suragani RN, Cadena SM, Cawley SM, et al. Transforming growth factor-beta superfamily ligand trap ACE-536 corrects anemia by promoting late-stage erythropoiesis. Nat. Med 2014;20(4):408–414. [DOI] [PubMed] [Google Scholar]
- 48.Gerds AT, Vannucchi AM, Passamonti F, 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(Suppl. 1):47–48. [Google Scholar]
- 49.Mesa R, Barosi G, Harrison C, Kiladjian J-J, Verstovsek S, Gerike TG, Chia V, Shetty JK, Wang Y, Marks H, Passamonti F. Efficacy and safety of luspatercept versus placebo in patients with myeloproliferative neoplasm-associated myelofibrosis on JAK2 inhibitor therapy and requiring RBC transfusions (INDEPENDENCE trial). HemaSphere 2021;6(Suppl. 2):805–806, abstract PB1702. [Google Scholar]
- 50.Kleppe M, Koche R, Zou L, van Galen P, Hill CE, Dong L. et al. Dual targeting of oncogenic activation and inflammatory signaling increases therapeutic efficacy in myeloproliferative neoplasms. Cancer Cell 2018;33(1):29–43.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Mascarenhas J, Gerds A, Verstovsek S. Paradigm shift: combination BET and JAK inhibition in myelofibrosis. Leukemia 2021;35(12):3361–3363. [DOI] [PubMed] [Google Scholar]
- 52.Saenz DT, Fiskus W, Manshouri T, Rajapakshe K, Krieger S, Sun B, et al. BET protein bromodomain inhibitor-based combinations are highly active against post-myeloproliferative neoplasm secondary AML cells. Leukemia 2017;31(3):678–687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Mascarenhas J, Kremyanskaya M, Patriarca A, Harrison C, Bose P. et al. BET inhibitor pelabresib (CPI-0610) combined with ruxolitinib in patients with myelofibrosis – JAK-inhibitor naïve or with suboptimal response to ruxolitinib – Preliminary data from the MANIFEST study. HemaSphere 2022;6:S3:99–100, abstract S198. [Google Scholar]
- 54.Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N. Engl. J. Med 2012;366(9):799–807. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Gupta V, Kremyanskaya M, Mascarenhas J, et al. Clinical benefit of pelabresib (CPI-0610) in combination with ruxolitinib in JAK inhibitor treatment naïve myelofibrosis patients: Interim efficacy subgroup analysis from Arm 3 of the MANIFEST phase 2 study. Clin. Lymphoma Myeloma Leuk 2021;21(Suppl. 1):S362. [Google Scholar]
- 56.Harrison C, Gupta V, Gerds AT, et al. Phase III MANIFEST-2: pelabresib + ruxolitinib vs. placebo + ruxolitinib in JAK-inhibitor treatment-naïve myelofibrosis. Future Oncol. 2022;18(27):2987–2997. [DOI] [PubMed] [Google Scholar]
- 57.Petiti J, Lo Iacono M, Rosso V, et al. Bcl-xL represents a therapeutic target in Philadelphia negative myeloproliferative neoplasms. J. Cell Mol. Med 2020;24:10978–10986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Passamonti F, Foran J, Tandra A, et al. Navitoclax plus ruxolitinib in JAK-inhibitor naïve patients with myelofibrosis: Preliminary safety and efficacy in a multicenter, open-label phase 2 study. HemaSphere 2022;6:S3:98–99; abstract S197. [Google Scholar]
- 59.Potluri J, Harb J, Masud AA, Hutti JE. A phase 3, double-blind, placebo-controlled, randomized study evaluating navitoclax in combination with ruxolitinib in patients with myelofibrosis (TRANSFORM-1). Blood 2020;136(Suppl. 1):4.32614961 [Google Scholar]
- 60.Gerds AT, Bartalucci N, Assad A, Yacoub A. Targeting the PI3K pathway in myeloproliferative neoplasms. Exp. Review Anticancer Therapy 2022;22(8):835–843. [DOI] [PubMed] [Google Scholar]
- 61.Fiskus W, Verstovsek S, Manshouri T, et al. Dual PI3K/AKT/mTOR inhibitor BEZ235 synergistically enhances the activity of JAK2 inhibitor against cultured and primary human myeloproliferative neoplasm cells. Mol. Cancer Ther 2013;12(5):577–588. [DOI] [PubMed] [Google Scholar]
- 62.Bartalucci N, Tozzi L, Bogani C, et al. Co-targeting the PI3K/mTOR and JAK2 signaling pathways produces synergistic activity against myeloproliferative neoplasms. J. Cell Mol. Med 2013;17(11):1385–1396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Gerds AT, Bartalucci N, Assad A, Yacoub A. Targeting the PI3K pathway in myeloproliferative neoplasms. Exp. Rev. Anticancer Ther 2022;22(8):835–843. [DOI] [PubMed] [Google Scholar]
- 64.Yacoub A, Erickson-Viitanen S, Zhou F, et al. 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(15)Suppl.,TPS7058. [Google Scholar]
- 65.Waibel M, Solomon VS., Knight DA, et al. Combined targeting of JAK2 and Bcl-2/Bcl-xL to cure mutant JAK2-driven malignancies and overcome acquired resistance to JAK2 inhibitors. Cell Rep. 2013;5(4):1047–1059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Harrison CN, Garcia JS, Somervaille TCP, et al. Addition of navitoclax to ongoing ruxolitinib therapy in patients with myelofibrosis with progression or suboptimal response: Phase II safety and efficacy. J. Clin. Oncol 2022;40(15):1671–1680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Pemmaraju N, Garcia JS, Potluri J, et al. Addition of navitoclax to ongoing ruxolitinib treatment in patients with myelofibrosis (REFINE): a post-hoc analysis of molecular biomarkers in a phase 2 study. Lancet Haematol. 2022;9(6):E434–E444. [DOI] [PubMed] [Google Scholar]
- 68.Dilley K, Harb J, Jalaluddin M, et al. A phase 3, open-label, randomized study evaluating the efficacy and safety of navitoclax plus ruxolitinib versus best available therapy in patients with relapsed/refractory myelofibrosis (TRANSFORM-2). Blood 2020;136(Suppl. 1):8.32614959 [Google Scholar]
- 69.Yacoub A, Borate U, Rampal R, et al. Efficacy and safety of add-on” parsaclisib to ruxolitinib therapy in myelofibrosis patients with suboptimal response to ruxolitinib: Final results from a phase 2 study. Blood 2022;140(Suppl. 1):579–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Lu M, Xia L, Salama ME, Hoffman R. Combination treatment with an MDM2 antagonist and a BET inhibitor targets both myelofibrosis hematopoietic stem/progenitor cells and their tumor promoting microenvironment. Blood 2017;130 (Suppl. 1):4225. [Google Scholar]
- 71.Tefferi A, Lasho TR, Finke CM, et al. Targeted deep sequencing in primary myelofibrosis. Blood Adv. 2016;1(2):105–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Mascarenhas J, Vannucchi AM, Mead AJ, et al. An open-label, global, multicenter phase 1b/2 study of KRT-232, a first-in-class, oral small-molecule inhibitor of murine double minute 2 (MDM2), combined with ruxolitinib in patients who have myelofibrosis and a suboptimal response to ruxolitinib. Blood 2020;136(Suppl. 1):44–45. [Google Scholar]
- 73.Lachowiez C, DiNardo CD, Stein E. Combining isocitrate dehydrogenase inhibitors with existing regimens in acute myeloid leukemia. Cancer J. 2022;28(1):21–28. [DOI] [PubMed] [Google Scholar]
- 74.McKenney AS, Lau AN, Somasundara AVH, et al. JAK2/IDH-mutant-driven myeloproliferative neoplasm is sensitive to combined targeted inhibition. J. Clin. Invest 2018;128(2):789–804. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Chifotides HT, Masarova L, Alfayez M, Daver N, Alvarado Y, Jabbour E, Konopleva M, Kantarjian HM, Patel KP, DiNardo CD, Verstovsek S. Outcome of patients with IDH1/2-mutated post-myeloproliferative neoplasm AML in the era of IDH inhibitors. Blood Adv. 2020; 4(21):5336–5342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Patel AA, Cahill K, Charnot-Katsikas A, et al. Clinical outcomes of IDH2-mutated advanced-phase Ph-negative myeloproliferative neoplasms treated with enasidenib. Br. J. Haematol 2020;190(1):e48–e51. [DOI] [PubMed] [Google Scholar]
- 77.Bar-Natan M, Mascarenhas J, Gerds AT., et al. Molecularly targeted combination therapy for advanced phase myeloproliferative neoplasm: MPN-RC 119. Blood 2022;140(Suppl. 1):3988–3990. [Google Scholar]
- 78.Chifotides HT, Bose P, Verstovsek S. Momelotinib: An emerging treatment for myelofibrosis patients with anemia. J. Hematol. Oncol 2021;15(1):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Asshoff M, Petzer V, Warr MR, et al. Momelotinib inhibits ACVR1/ALK2, decreases hepcidin production, and ameliorates anemia of chronic disease in rodents. Blood 2017;129(13):1823–1830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Oh ST, Talpaz M, Gerds AT, et al. ACVR1/JAK1/JAK2 inhibitor momelotinib reverses transfusion dependency and suppresses hepcidin in myelofibrosis phase 2 trial. Blood 2020;4(18):4282–4291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Mesa RA, Kiladjian J-J, Catalano JV, et al. SIMPLIFY-1: A phase III randomized trial of momelotinib versus ruxolitinib in Janus kinase inhibitor-naïve patients with myelofibrosis. J. Clin. Oncol 2017;35(34):3844–3850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Verstovsek S, Egyed M, Lech-Maranda E, et al. Robust overall survival and sustained efficacy outcomes during long term exposure to momelotinib in JAK inhibitor naïve and previously JAK inhibitor treated intermediate/high risk myelofibrosis patients. Blood 2020;136(Suppl.1):51–52. [Google Scholar]
- 83.Harrison CN, Vannucchi AM, Platzbecker U, et al. Momelotinib versus best available therapy in patients with myelofibrosis previously treated with ruxolitinib (SIMPLIFY 2): A randomized, open-label, phase 3 trial. Lancet Haematol. 2018;5(2):e73–e81. [DOI] [PubMed] [Google Scholar]
- 84.Mesa RA, Catalano J, Cervantes F, et al. Dynamic and time-to-event analyses demonstrate marked reduction in transfusion requirements for Janus kinase inhibitor-naïve myelofibrosis patients treated with momelotinib compared head-to-head with ruxolitinib. Blood 2019;134(Suppl. 1):1663. [Google Scholar]
- 85.Mesa R, Gerds AT, Gupta V, et al. Momelotinib reduces transfusion requirements in patients with myelofibrosis. Leuk. Lymphoma 2022;63(7):1718–1722. [DOI] [PubMed] [Google Scholar]
- 86.Mesa R, Harrison C, Oh S, et al. Overall survival in the SIMPLIFY-1 and SIMPLIFY-2 phase 3 trials of momelotinib in patients with myelofibrosis. Leukemia 2022;36(9):2261–2268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Verstovsek S, Chen C-C, Egyed M, et al. MOMENTUM: Momelotinib vs. danazol in patients with myelofibrosis previously treated with JAKi who are symptomatic and anemic. Future Oncol. 2021;17(12):1449–1458. [DOI] [PubMed] [Google Scholar]
- 88.Verstovsek S, Gerds A, Vannucchi AM, et al. Momelotinib versus danazol in symptomatic patients with anaemia and myelofibrosis (MOMENTUM): Results from an international, double-blind, randomised, controlled phase 3 study. The Lancet 2023;401(10373):269–280. [DOI] [PubMed] [Google Scholar]
- 89.Verstovsek S, Oh ST, Kiladjian J-J, et al. Transfusion independence response as a potential surrogate for overall survival in Jaki-experienced patients with myelofibrosis from MOMENTUM. Blood 2022;140(Suppl.1):6803–6805. [Google Scholar]
- 90.Gerds AT, Verstovsek S, Vannucchi A, et al. Thrombocytopenic myelofibrosis (MF) patients previously treated with a JAK inhibitor in a phase 3 randomized study of momelotinib (MMB) versus danazol (DAN) [MOMENTUM]. J. Clin. Oncol 2022; 40(16), Suppl., abstract 7061. [Google Scholar]
- 91.Al-Ali HK, Delgado RG, Lange A, et al. KRT-232, a first-in-class, murine double minute 2 inhibitor (MDM2i), for myelofibrosis (MF) relapsed or refractory (R/R) to Janus-associated kinase inhibitor (JAKi) treatment. HemaSphere 2020;4:S215. [Google Scholar]
- 92.Vachhani P, Lange A, Delgado RG, et al. Potential disease-modifying activity of navtemadlin (KRT-232), a first-in-class MDM2 inhibitor, correlates with clinical benefits in relapsed/refractory myelofibrosis. Blood 2021;138(Suppl. 1):3581. [Google Scholar]
- 93.Verstovsek S, Al-Ali HK, Mascarenhas J, et al. BOREAS: A global phase III study of the MDM2 inhibitor navtemadlin KRT-232 in relapsed/refractory myelofibrosis. Future Oncol. 2022;18(37):4059–4069. [DOI] [PubMed] [Google Scholar]
- 94.Mascarenhas J Komrokji RS, Palandri F, et al. Randomized, single-blind, multicenter phase II study of two doses of imetelstat in relapsed or refractory myelofibrosis. J. Clin. Oncol 2021;39(26):2881–2892. [DOI] [PubMed] [Google Scholar]
- 95.Mascarenhas J, Komrojki RS, Cavo M, et al. Potential disease-modifying activity of imetelstat demonstrated by reduction in cytogenetically abnormal clones and mutation burden leads to clinical benefits in relapsed/refractory myelofibrosis patients. Blood 2020;136(Suppl. 1):39–40. [Google Scholar]
- 96.Kuykendall A, Sun L, Mascarenhas J, et al. Favorable overall survival with imetelstat in relapsed/refractory myelofibrosis patients compared with real world data. Ann. Haematol 2022;101(1):139–146. [DOI] [PubMed] [Google Scholar]
- 97.Rumi E, Pietra D, Pascutto C, et al. Clinical effect of driver mutations of JAK2, CALR, or MPL in primary myelofibrosis. Blood 2014;124:1062(7)-1069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Tefferi A, Lasho TL, Finke CM, et al. CALR vs JAK2 vs MPL-mutated or triple-negative myelofibrosis: clinical, cytogenetic and molecular comparisons. Leukemia 2014;28(7):1472–1477. [DOI] [PubMed] [Google Scholar]
- 99.Tefferi A, Lasho TR, Finke CM, et al. Targeted deep sequencing in primary myelofibrosis. Blood Adv. 2016;1(2):105–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kiladjian J-J, Mascarenhas JO, Komrokji RS, et al. Imetelstat treatment results in clinical benefits, including improved overall survival, in patients with higher-risk triple-negative myelofibrosis relapsed/refractory to Janus kinase inhibitors (JAKi). HemaSphere 2020;4(Suppl. 1):508, abstract 1101. [Google Scholar]
- 101.Mascarenhas J, Harrison C, Kiladjian J-J, et al. Imetelstat in intermediate-2 or high-risk myelofibrosis (MF) refractory to Janus kinase inhibitor: IMpactMF phase III study design. Future Oncol. 2022;18(22):2393–2402. [DOI] [PubMed] [Google Scholar]
- 102.Venugopal S, Mascarenhas J. Novel therapeutics in myeloproliferative neoplasms. J Hematol Oncol. 2020;13(1):162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Verstovsek S, Mesa R, Gupta V. et al. Momelotinib (MMB) long-term safety: pooled data from three phase 3 randomized-controlled trials (RCTs). Blood 2022;140(Suppl. 1):9670–9672. [Google Scholar]
- 104.Kuykendall AT, Komrokji RS. JAK be nimble: Reviewing the development of JAK inhibitors and JAK inhibitor combinations for special populations of patients with myelofibrosis. J. Immun. Precis. Oncol 2021;4(3):129–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Vachhani P, Verstovsek S, Bose P. Disease modification in myelofibrosis: An elusive goal? J. Clin. Oncol 2022;40(11):1147–1154. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Pemmaraju N, Verstovsek S, Mesa R. et al. Defining disease modification in myelofibrosis in the era of targeted therapy. Cancer 2022;128(13):2420–2432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Mascarenhas J, Gleitz HFE, Chifotides HT. et al. Biological drivers of clinical phenotype in myelofibrosis. Leukemia 2023;37:255–264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Handlos Grauslund J, Orebo-Holmström M, Grønne Jørgensen N, et al. Therapeutic cancer vaccination with a peptide derived from the calreticulin exon 9 mutations induces strong cellular immune responses in patients with CALR-mutant chronic myeloproliferative neoplasms. Front Oncol. 2021;11:637420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Holmström MO, Hasselbalch HC, Andersen MH. Cancer immune therapy for Philadelphia chromosome-negative chronic myeloproliferative neoplasms. Cancers (Basel) 2022;12:1763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Holmström MO, Martinenaite E, Ahmad SM, et al. The calreticulin (CALR) exon 9 mutations are promising targets for cancer immune therapy. Leukemia 2018;32:429–437. [DOI] [PubMed] [Google Scholar]
- 111.Holmström MO, Hjortsø MD, Ahmad SM, et al. The JAK2V617F mutation is a target for specific T cells in the JAK2V617F-positive myeloproliferative neoplasms. Leukemia 2017;31:495–498. [DOI] [PubMed] [Google Scholar]
- 112.Cimen-Bozkus C, Roudko V, Finnigan JP. Immune checkpoint blockade enhances shared neoantigen-induced T cell immunity directed against mutated calreticulin in myeloproliferative neoplasms. Cancer Discov 2019;9(9):1192–1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Tvorogov D, Thompson-Peach CAL, Foßelteder J. et al. Targeting human CARL-mutated MPN progenitors with a neoepitope-directed monoclonal antibody. EMBO Reports. 2022;23(4):e52904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.How J, Hobbs GS, Mullally A. Mutant calreticulin in myeloproliferative neoplasms. Blood 2019;134(25):2242–2248. [DOI] [PMC free article] [PubMed] [Google Scholar]