Abstract
Purpose of Review
To provide a contemporary update of novel agents and targets under investigation in myelofibrosis in the JAK inhibitor era.
Recent findings
Myelofibrosis (MF) is a clonal stem cell disease characterized by marrow fibrosis and a heterogeneous disease phenotype with a variable degree of splenomegaly, cytopenias, and constitutional symptoms that significantly impact quality of life and survival. Overactive JAK/STAT signaling is a hallmark of MF. The only approved therapy for MF, JAK1/2 inhibitor ruxolitinib, can ameliorate splenomegaly, improve symptoms, and prolong survival in some patients. Therapeutic challenges remain, however. Myelosuppression limits the use of ruxolitinib in some patients, eventual drug resistance is common, and the underlying malignant clone persists despite therapy. A deeper understanding of the pathogenesis of MF has informed the development of additional agents.
Summary
Promising targets under investigation include JAK1 and JAK2, downstream intermediates in related signaling pathways, epigenetic modifiers, pro-inflammatory cytokines, and immune regulators.
Keywords: myelofibrosis, myeloproliferative neoplasms, novel therapies, JAK inhibitors, drug development
Introduction
Myelofibrosis (MF) is a heterogeneous disease within the family of BCR-ABL negative myeloproliferative neoplasms (MPNs) characterized by dysregulated proliferation of myeloid cells, aberrant deposition of reticulin and collagen in the bone marrow, and excess production of pro-inflammatory cytokines. The resulting clinical manifestations vary between individuals and include progressive cytopenias, extramedullary hematopoiesis resulting in splenomegaly, constitutional symptoms (i.e. fatigue, pruritus, and night sweats), psychosocial symptoms, acute leukemic transformation, and shortened life expectancy (1-3). Current MF therapies are often ineffective in controlling symptoms or altering the natural history of the disease.
Insights into molecular mechanisms of MPN pathogenesis have spurred drug development in the field. Dysregulation of the JAK/STAT pathway is central to MPN development, and driven by activating mutations in Janus kinase 2 (JAK2), calreticulin (CALR), or myeloproliferative leukemia virus (MPL) in over 90% of MF cases (4, 5). Alterations in additional cellular processes such as DNA methylation (i.e. TET2, DNMT3A mutations), histone modification (ASXL1, EZH2 mutations), RNA splicing (U2AF1, SF3B1, SRSF2 mutations), and signaling through other pathways (RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, LNK) further contribute to MF initiation or progression and may explain some of the variability in the disease phenotype (6). This complexity and heterogeneity in disease biology provides both challenges and opportunities for drug development in MF.
Current Risk-Adapted Approach to Treatment
There are currently few standard treatment options for patients with MF. Allogeneic hematopoietic stem cell transplantation (alloHSCT) provides the only potentially curative treatment modality, however its use in the MF population is marred by potential toxicities due to advanced age (median age at diagnosis is 67 years), comorbidities, and poor functional status resulting from disease symptomatology (7). Other treatment modalities are aimed at reducing symptoms and improving blood counts, with little effect on the underlying malignant clone or on patient survival. Ruxolitinib is an oral inhibitor of JAK1/2 with the ability to reduce spleen size and improve symptoms in some patients, and has been associated with a modest survival advantage (8, 9). A number of other agents have been used to improve cytopenias or reduce splenomegaly with variable success, including erythropoietin stimulating agents, androgens (i.e. danazol), immunomodulators (i.e. thalidomide), and hydroxyurea.
Management decisions for patients with MF are dictated by individual patient symptoms and the risk of disease transformation or patient death. These risks are assessed using either the Dynamic International Prognostic Scoring System (DIPSS) which incorporates age, white blood cell count, hemoglobin level, circulating blast cells, and constitutional symptoms, or the newer DIPSS-Plus that adds karyotype, red blood cell transfusion requirement, and thrombocytopenia (10, 11). Using the DIPSS-Plus tool, patients are assigned low, intermediate-1, intermediate-2, or high risk scores corresponding to median overall survival times of 15.4 years, 6.5 years, 2.9 years, and 1.3 years, respectively. These disparate outcomes highlight the heterogeneity among patients with MF, and underscore the importance of risk-directed treatment algorithms (12). Presently, there is no evidence that early treatment of asymptomatic patients improves survival, and therefore management of asymptomatic low risk patients is generally supportive and expectant. Ruxolitinib is approved by the United States Food and Drug Administration (FDA) for intermediate or high risk MF, however it is often used in lower risk patients with significant disease-related symptoms and has been included in the 2017 inaugural National Comprehensive Cancer Network (NCCN) guidelines for MF for any symptomatic patient, provided that the platelet count is >50 109/L (12). AlloHSCT has been shown to improve long-term outcomes among those with intermediate-2 or high-risk disease, and should therefore be offered to those deemed eligible (13). For those with low risk disease, alloHSCT is associated with inferior 5-year survival rates when compared to those treated without transplant. For those with intermediate-1 risk disease, the risk-benefit ratio of alloHSCT remains unclear, and may be considered on a case-by-case basis. Despite improvements in risk stratification and their application to treatment algorithms for patients with MF, the current therapies prove inadequate for many. Development of novel agents and approaches for treatment of MF therefore remains a significant area of unmet need. In this review, we summarize the contemporary drug therapies for MF, with a focus on novel agents and approaches.
JAK Inhibitors
Ruxolitinib
JAK1/2 inhibitor ruxolitinib remains the only FDA approved agent for MF, and sets the standard against which novel agents are measured. The COMFORT-I and COMFORT-II trials demonstrated clinical benefit from ruxolitinib compared to placebo (COMFORT-I) or best available therapy (COMFORT-II), including spleen volume reduction (SVR), decrease in total symptom score (TSS), improvement in quality of life measures, and improvement or stabilization of bone marrow fibrosis (table 1) (14-16). Follow up at 5 years revealed sustained responses with median response duration among the ruxolitinib-randomized patients of 3.2 years in both studies (8, 9). Improvement in overall survival (OS) was also shown in the ruxolitinib groups even after crossover (not reached versus 3.8 years in COMFORT-I; not reached versus 4.1 years in COMFORT-II).
Table 1.
Results of Select JAK Inhibitor Clinic Trials
Agent | Target(s) | Clinical Trial | Patient Characteristics | Key Results | Toxicities |
---|---|---|---|---|---|
Ruxolitinib (RUX) | JAK 1/2 | COMFORT-1 Randomized phase 3 study of RUX vs placebo |
Intermediate-2 or high-risk MF RUX (n=155) Placebo (n=154) |
Primary endpoint: SVR ≥35% at 24 weeks - Reached in 41.9% of RUX cohort vs 0.7% in placebo cohort Reduction in TSS ≥50% at 24 weeks - 45.9% (RUX) vs 5.3% (placebo) Median spleen response duration 168.3 weeks (RUX) Median OS at 5 years not reached (RUX) vs 200 weeks (placebo) (HR 0.69; 95% CI 0.50-0.96, p=0.025) |
G3/4 anemia 45.2% G3/4 thrombocytopenia 12.9% G3/4 neutropenia 7.1% Rate of non-hematologic toxicities similar between RUX and placebo groups |
COMFORT-II Randomized phase 3 study of RUX vs BAT |
Intermediate-2 or high-risk MF RUX (n=146) BAT (n=73) |
Primary endpoint: SVR ≥35% at 48 weeks - Reached in 28% (RUX) vs 0% (BAT) At 5 years, probability of maintaining spleen response 0.48 (95% CI, 0.35-0.60), median duration of spleen response 3.2 years Median OS at 5 years not reached (RUX) vs 4.1 years (BAT) |
Similar to COMFORT-I Any-grade diarrhea 23% |
||
Pacritinib | JAK2/FLT3 | PERSIST-I Randomized phase 3 study of pacritinib vs BAT (excluding JAK inhibitors) |
Intermediate-1, intermediate-2 or high-risk MF JAK inhibitor naïve No exclusions for cytopenias Pacritinib 400 mg daily (n=220) BAT (n=107) |
Primary endpoint: SVR ≥35% at 24 weeks - Reached in 19% (pacritinib) vs 5% (BAT) |
G3/4 anemia 17% G3/4 thrombocytopenia 12% G3/4 diarrhea 5% Heart failure 2% |
PERSIST-II Randomized phase 3 study of pacritinib vs BAT (including RUX) |
Intermediate-1, intermediate-2, or high-risk MF with platelets <100×109/L Previously treated or JAK inhibitor naïve Pacritinib 400 mg daily (n=104) Pacritinib 200 mg BID (n=107) BAT (n=100) |
Primary endpoints: SVR ≥35% at 24 weeks -Reached in 18% (pacritinib) vs 3% (BAT) Reduction in TSS ≥50% at 24 weeks - Reached in 25% (pacritinib) vs 14% (BAT) |
Toxicities were less frequent in pacritinib BID dosing compared to daily dosing Cardiac AEs in 7% (pacritinib BID), 13% (pacritinib daily), and 9% (BAT) Intracranial hemorrhage 1% (pacritinib daily) *Pacritinib was on full clinical hold 2/2016-1/2017 for fatal toxicity concerns. Further dose finding studies are now planned. |
||
Momelotinib | JAK1/2 | SIMPLIFY-I Randomized phase 3 study of momelotinib vs RUX |
Intermediate-1 (symptomatic), intermediate-2, or high-risk MF, JAK inhibitor naïve Momelotonib (n=215) RUX (n=217) |
Primary endpoint: SVR ≥35% at 24 weeks - Momelotinib non-inferior to RUX for spleen reduction [26.9% (momelotinib) vs 29% (RUX)] Reduction in TSS ≥50% at 24 weeks - Momelotinib was inferior to RUX Transfusion requirements - Momelotinib was associated with decreased transfusion requirements |
G3/4 thrombocytopenia (7%) G3/4 anemia (6%) All grade peripheral neuropathy 10% (momelotinib) vs 5% (RUX) |
SIMPLIFY-II Randomized phase 3 study of momelotinib vs BAT (including RUX) |
Intermediate-1 (symptomatic), intermediate-2, or high-risk MF previously treated with RUX Momelotinib (n=104) BAT (included RUX in 88%) (n=52) |
Primary endpoint: SVR ≥35% at 24 weeks - Momelotinib was not superior to BAT (including RUX) in improving spleen size in patients previously treated with RUX Reduction in TSS ≥50% at 24 weeks - Momelotinib superior to BAT [26.2% (momelotinib) vs 5.9% (BAT)] Transfusion requirements - Momelotinib was associated with decreased transfusion requirements |
G3/4 anemia (13%) G3/4 thrombocytopenia (7%) All grade peripheral neuropathy 11% (momelotinib) vs 0% (BAT) |
||
NS-018 | JAK2/Src | Phase 1/2 study of 2 dosing schedules of NS-018 (once daily or BID) | Intermediate-1, intermediate-2, or high-risk MF Previously treated or treatment naive Phase 1 n=48 Phase 2 n=29 (ongoing) |
20/36 (56%) evaluable patients with >50% reduction in spleen size by palpation - Includes 9/19 (47%) previously treated with a JAK inhibitor RP2D 300 mg BID. Phase 2 is ongoing. |
G3/4 anemia (21%) G3/4 thrombocytopenia (17%) |
MF, myelofibrosis; RUX, ruxolitinib; SVR, spleen volume reduction; TSS, total symptom score; OS, overall survival; HR, hazard ratio; CI, confidence interval; G3/4, grade 3/4; RP2D, recommended phase 2 dose
Despite meaningful clinical benefits conferred by ruxolitinb, challenges remain. First, the effects on the malignant clone appear to be minimal. Molecular responses as measured by JAK2 mutant allele burden are uncommon (17). Second, anemia and thrombocytopenia limit the use and dose of ruxolitinib in certain populations. Both thrombopoietin and erythropoietin signaling involve JAK2, and therefore thrombocytopenia and anemia are expected and dose-related (18). Despite this limitation, low-dose ruxolitinib has proven to be relatively safe in those with baseline platelet counts of 50–100 × 109/L, and the agent is still associated with a favorable response profile even at low doses in this population (18). Third, the eventual development of resistance to JAK inhibitors presents a therapeutic challenge. Long term follow up from the COMFORT-II trial showed that the probability of maintaining a response to ruxolitinib at 5 years was 0.48 (95% confidence interval 0.35-0.60), and the median response duration was 3.2 years (8). Multiple mechanisms of resistance to JAK inhibition have been described, including up-regulation of parallel pathways, heterodimerization of activated JAK2 with other JAK kinases including JAK1 and TYK2, and point mutations in the kinase domain of JAK2 that have been identified in cell lines but have not yet been seen in patients (19-23). Investigation of rationally designed combination therapies to prevent or overcome resistance is therefore warranted.
Other JAK Inhibitors
The number of other JAK inhibitors in development has unfortunately dwindled over time due to toxicity concerns and failure to meet efficacy endpoints in larger trials. However, several investigational JAK inhibitors of interest persist. Table 1 describes features and key results of clinical trials with select JAK inhibitors.
Pacritinib, a JAK2/FLT3 inhibitor, garnered interest due to its lack of myelosuppression noted in early clinical trials (24). Two phase 3 trials, PERSIST-1 and PERSIST-2, have been performed. In PERSIST-1, patients with higher-risk MF were randomized to pacritinib versus best available therapy (BAT) (25). The primary endpoint of ≥35% reduction in SVR was met by 19% in the pacritinib arm versus 5% in the BAT arm, with minimal myelosuppression (Table 1) even among patients with baseline cytopenias. PERSIST-2 focused exclusively on patients with platelets <100 × 109/L, and randomized patients to two doses of pacritinib (200 mg BID or 400 mg once daily) or BAT, which could include ruxolitinib (26). Prior treatment with ruxolitinib was allowed as well. Patients in the pacritinib arm achieved greater reductions in spleen volume, TSS, and transfusion requirements at 24 weeks (Table 1). The FDA imposed a full clinical hold on pacritinib in February 2016 due to concerns regarding excess fatalities, cardiac events, and hemorrhagic events. The clinical hold was lifted in January 2017. Pacritinib remains an attractive agent due to potential for use in thrombocytopenic patients, however further studies to clarify the safe and effective dose and schedule are warranted.
Momelotinib is a JAK1/2 inhibitor with the attractive feature of improving anemia, likely due to reduction in hepcidin production by the liver (27). SIMPLIFY-1, a phase 3 head-to-head trial of momelotinib versus ruxolitinib in JAK inhibitor-naïve patients with MF met its primary endpoint in demonstrating noninferiority in SVR responses at 24 weeks, however failed to meet its secondary endpoint of TSS reduction (28). In a second phase 3 study, SIMPLIFY-2, patients previously exposed to ruxolitinib were randomized to momelotinib versus BAT, which included ruxolitinib in most (29). This trial failed to meet its primary endpoint of superiority of momelotinib in terms of SVR responses, however did show a reduction in TSS and improvement in anemia. As a result of these somewhat disappointing phase 3 results, momelotinib is no longer in development and the therapeutic void for patients with MF and anemia remains unfilled.
Several additional JAK inhibitors are under investigation in earlier clinical phases. NS018 is a selective inhibitor of JAK2 and Src that showed a favorable toxicity profile and promising efficacy signals in phase 1; the phase 2 portion of this study is ongoing (30). A phase 2 study of JAK1 inhibitor itacitinib demonstrated the ability of selective JAK1 inhibition to improve splenomegaly and symptoms related to MF while preserving hemoglobin levels (31). A multicenter phase 2 study evaluating itacitibine alone or in combination with low-dose ruxolitinib after ruxolitinib failure is planned (NCT03144687).
Beyond JAK Inhibitors
DNA Hypomethylating Agents
Epigenetic alterations, such as CpG island hypermethylation causing inactivation of tumor suppressor genes, have been implicated in the pathogenesis of many malignancies including MF (32-34). DNA hypomethylating agents (HMAs) such as azacitidine and decitabine are postulated to exert their effects, in part, through reactivation of hypomethylated genes. Both are FDA approved for the treatment of myelodysplastic syndrome (MDS), and are also frequently used for the treatment of acute myeloid leukemia (AML). Modest clinical responses have been reported with HMAs in patients with MF. A phase 2 study evaluated the effects of azacitidine in 34 patients with MF; 76% had received previous treatment (35). Clinical improvement (CI) was seen in 21%, and a partial response (PR) was achieved in 1 patient (3%). No complete responses (CR) were seen, and no improvement in bone marrow fibrosis was identified. Myelosuppression was common with this standard, 7-day azacitidine regimen. Another study administered azacitidine on a shortened 5-day schedule to 10 patients with MF, and no improvement was reported (36).
Low-dose subcutaneous decitabine (0.3 mg/kg/day on days 1-5 and days 8-12) has shown some evidence of efficacy. In this Phase II trial in MF, of 19 evaluable patients, a 37% overall response rate was reported (37). Myelosuppression was significant, though reversible. In a retrospective report, standard dose decitabine (20 mg/m2 intravenously daily on days 1-5) resulted in clinical benefit in 9 (82%) of patients with high risk MF, but no partial or complete responses, and benefits were maintained for a median of 9 months (38). Other case reports have described efficacious use of decitabine in controlling symptoms, improving splenomegaly, and decreasing transfusion requirements. (39).
Combination studies of JAK inhibitors and HMAs for chronic phase MF are underway. Clinical responses have been reported with either azacitidine or decitabine in combination with ruxolitinib in intermediate-2 or high risk MF (40, 41). A phase II study combining low-dose azacitidine with ruxolitinib for patients with chronic phase MF or myelodysplastic syndrome/myeloproliferative neoplasms (MDS/MPN) is ongoing (NCT01787487).
HMAs may be particularly useful in cases of accelerated or blast phase disease, where ORR as high as 52% has been reported with azacitidine (42) and encouraging activity has been reported in retrospective series with decitabine (38, 43). Two phase 1 studies of decitabine plus ruxolitinib in accelerated or blast phase MF demonstrated that the combination was tolerable and promising (44, 45), and a phase 2 portion through the Myeloproliferative Disorders Research Consortium is ongoing (NCT02076191).
Histone Deacetylace Inhibitors
Histone deacetylace inhibitors (HDACi) represent another epigenetic-targeted therapy under investigation for MF. As single agents, vorinostat, panobinostat, givinostat, and pracinostat have all demonstrated modest clinical activity (46-51). The most common class toxicities of HDACi include cytopenias, fatigue, and diarrhea. Two clinical trials investigating the combination of HDACi and JAK inhibitors are ongoing (NCT01693601 and NCT01433445). Preliminary results of the Phase 1b trial of the combination of panobinostat and ruxolitinib have been reported (52). Among 61 patients with MF, 57% and 39% achieved SVRs ≥35% at 24 weeks and 48 weeks, respectively. Improvement in bone marrow fibrosis occurred in 4 of 12 evaluable patients, and ≥20% decrease in JAK2 mutant allele burden was seen in 5 out of 17 tested patients.
PI3K/AKT/mTOR Pathway Inhibitors
The PI3K/AKT/mTOR signaling pathway and the JAK/STAT signaling pathways are intricately connected, and both are both aberrantly activated in MPN (53). Small molecule inhibitors of PI3K, AKT, and mTOR have all been subjects of preclinical investigation in MF, with encouraging results (54-56). In a phase I/II clinical trial, mTOR inhibitor everolimus induced responses in 23% (1 PR, 6 CI) (57). Pre-clinical synergy has been demonstrated with combinations of PI3K/AKT/mTOR inhibitors and JAK inhibitors, prompting several ongoing combination studies including PI3K inhibitor buparlisib with ruxolitinib (NCT01730248), PI3K inhibitor INCB050465 and ruxolitinib (NCT02718300), and selective PI3Kδ inhibitor TGR-1202 and ruxolitinib (NCT02493530). Early results from the buparlisib and ruxolitinib phase 1b study have been reported, and the combination was reasonably well tolerated (58). Clinical responses were noted, with palpable spleen length reduction of ≥50% in 82% of JAK inhibitor naïve and 55% of JAK inhibitor pre-treated patients.
RAF/MEK/ERK Pathway Inhibitors
In parallel to the PI3K/AKT/mTOR pathway, the RAF/MEK/ERK signaling pathway is also activated by increased JAK/STAT signaling and contributes to impaired cellular differentiation and increased proliferation (59-61). In a CALR deleted murine model with a MPN phenotype, treatment with the MEK inhibitor trametinib alone significantly reduced bone marrow fibrosis (62). Combining MEK inhibitor selumetinib with ruxolitinib has been shown to significantly inhibit malignant cell growth and rescue hematopoietic stem cell function, as well as prolong survival in a NRAS mutant murine model with a MDS/MPN phenotype (63). While clinical experience with MEK inhibitors in MF is limited, modest single-agent activity in AML has been demonstrated (64, 65). Further investigation of RAF/MEK/ERK pathway inhibitors in MF, in rationally designed combinations, is warranted and a trial combining the MEK inhibitor selumetinib (table 2) with the DNA hypomethylating agent azacitidine will soon be underway.
Table 2.
Novel Agents for Myelofibrosis in Clinical Development
Class | Agent | Target | Phase | NCT identifier |
---|---|---|---|---|
JAK inhibitors | Itacitinib, alone or with ruxolitinib (+RUX) | JAK1 | 2 | NCT03144687 |
Pacritinib | JAK2/FLT3 | 2 | NCT03165734 | |
NS-018 | JAK2/Src | 2 | NCT01423851 | |
Epigenetic Agents | Pracinostat (+RUX) | HDAC | 2 | NCT02267278 |
Panobinostat | HDAC | 1/2 | NCT01693601 | |
IMG-7289 | LSD-1 | 1 | NCT03136185 | |
Azacitidine (+RUX) | DNA methylation | 2 | NCT01787487 | |
SGI-110 | DNA methylation | 2 | NCT03075826 | |
PI3K/AKT/mTOR Pathway Inhibitors |
INCB050465 (+RUX) | PI3K | 2 | NCT02718300 |
Buparlisib (+RUX) | PI3K | 1 | NCT01730248 | |
TGR-1202 (+RUX) | PI3Kδ | 1 | NCT02493530 | |
Hedgehog Pathway Inhibitors |
Vismodegib (+ RUX) | SMO | 1/2 | NCT02593760 |
Sonidegib (+ RUX) | SMO | 1/2 | NCT01787552 | |
Glasdegib | SMO | 2 | NCT02226172 | |
Other Small Molecule Inhibitors |
CPI-0610 | BET | 1 | NCT02158858 |
PIM447 (+ RUX) | pan-PIM kinases | 1b | NCT02370706 | |
Ribocicilib (+ RUX) | CDK4/6 | 1b | NCT02370706 | |
Alisertib | Aurora kinase A | 1 | NCT02530619 | |
Selumetinib | MEK kinase | 1 | *pending | |
Checkpoint Inhibitors |
Durvalumab | PD-L1 | 1 | NCT02871323 |
Pembrolizumab | PD-1 | 2 | NCT03065400 | |
Nivolumab | PD-1 | 2 | NCT02421354 | |
Nivolumab | PD-1 | 1/1b | NCT01822509 | |
Ipilimumab | CTLA4 | 1/1b | NCT01822509 | |
Other Agents | Imetelstat | Telomerase | 2 | NCT02426086 |
PRM-151 | Pentraxin 2 | 2 | NCT01981850 | |
Sotatercept | TGFβ | 2 | NCT01712308 | |
SL-401 | IL3 receptor (CD123) | 1/2 | NCT02268253 | |
P1101 | Peg-Interferon α | 2 | NCT02370329 | |
LCL-161 | SMAC mimetic | 2 | NCT02098161 |
Clinical trials.gov listing is pending at time of manuscript submission
Hedgehog Inhibitors
The hedgehog signaling pathway contributes to normal hematopoiesis, and overactive hedgehog signaling has been implicated in the pathogenesis of both malignant and fibrotic diseases (66, 67). Small molecule inhibitors of several hedgehog signaling proteins have shown clinical activity in MF. Early results of a phase 1/2 study of glasdegib as a single agent in patients with MF after JAK inhibitor therapy showed a favorable toxicity profile of the drug, with modest single-agent responses (68). The main toxicities noted were dysguesia, muscle spasms, alopecia, decreased appetite, and fatigue. No patient achieved SVR ≥35%, but 5 (24%) did have some degree of improvement in splenomegaly. Favorable symptom responses were seen, as 8 (38%) had ≥20% decrease in TSS. This study is ongoing (NCT02226172). Combinations of hedgehog pathway inhibitors and JAK inhibitors are also underway. Preliminary results from a phase 1b/2 study of sonidegib in combination with ruxolitinib in 27 patients with MF showed that 56% of patients achieved a SVR of ≥35% at any time during treatment (NCT01787552)(69).
Telomerase Inhibitors
Telomeres are repetitive DNA sequences that cap chromosomes, protect coding DNA, and shorten with each cycle of cell division (70). Many malignant cells express telomerase, a holoenzyme responsible for maintaining telomere length. Imetelstat is an oligonucleotide that binds the RNA template of telomerase and competitively inhibits enzymatic activity telomerase activity (71, 72). Imetelstat was investigated in a pilot study of 33 patients with intermediate-2 or high-risk MF, about half of whom were previously treated with ruxolitinib (73). Complete or partial remissions were seen in 7 patients (21%), with median response durations of 18 months and 10 months for those who achieved complete and partial remissions, respectively. Among the 4 patients who achieved a CR, bone marrow fibrosis was reversed in all 4 and molecular responses occurred in 3. Imetelstat was relatively well tolerated in this population, with the most common toxicities being cytopenias and transaminitis. A phase II study of 2 doses of imetelstat in patients with intermediate-2 or high-risk MF previously treated with a JAK inhibitor is ongoing (NCT02426086). Favorable responses to imetelstat have also noted in patients with essential thrombocythemia, however this indication has not been pursued further, likely due to the relatively indolent course associated with ET (74).
Anti-Fibrosing Agents
Targeting the complex pathogenic mechanisms that result in bone marrow fibrosis remains challenging. One novel therapeutic target is pentraxin 2, an endogenous protein that regulates differentiation of monocytes into fibrocytes and pro-fibrotic macrophages at sites of tissue damage (75-77). PRM-151 is a recombinant form of pentraxin 2 that was initially developed as an agent for pulmonary fibrosis, but has since been studied in MF. In a phase 2 study of PRM-151 in combination with ruxolitinib in patients with intermediate-1 or higher risk disease, 35% experienced an objective response, defined as CI (15%) and/or reduction in bone marrow fibrosis (23%) (78). Improvements in anemia (40%), spleen size (26%), and symptoms (38%) were also noted. A second stage of this study evaluating 3 dose levels is ongoing (NCT01981850).
Several other potential targets involved in fibrotic processes have been identified, however clinical results have been somewhat disappointing to date. Lysyl oxidase like (LOXL) is an amine oxidase enzyme that catalyzes a key step in the formation of crosslinks between collagen and elastin. In preclinical models, LOXL levels were found to be elevated, and inhibition of LOXL led to improvement in marrow fibrosis (79, 80). In a phase II study, a humanized antibody against LOXL2, simtuzumab, was well tolerated but failed to reduce marrow fibrosis or achieve clinical improvement (81).
Cytokine transforming growth factor-β (TGF-β) has been implicated in both the fibrotic and proliferative aspects of myelofibrosis (82). Sotatercept, a first-in-class activin receptor type IIA (ActRIIA) ligand trap, causes sequestration of TGF-β ligands and improvement in erythroid differentiation. A phase 2 study is ongoing in patients with MF-associated anemia, and interim results showed an anemia response in 5/14 (36%) evaluable patients, but effects on bone marrow fibrosis are not yet known (NCT01712308) (83). A phase I study of fresolimumab, a TGF-β-neutralizing monoclonal antibody, was initiated but the drug was withdrawn after 3 subjects were treated at the lowest planned dose level due to management decisions on the part of the pharmaceutical company. At that low dose, the agent was well tolerated and one patient experienced hematologic improvement, achieving transfusion independence (84). While the clinical experience with TGF-β targeted agents is limited, this remains an interesting avenue for future investigation, particularly in anemic patients.
Immunotherapy
Immune dysregulation is a central feature of MPNs, and immune based approaches to treatment are therefore appealing. Allogeneic stem cell transplantation (alloHSCT) remains the only potentially curative therapy for MF, and the only immunotherapeutic strategy known to be effective for MPN. However, only a minority of MF patients will be eligible due to older age at diagnosis, comorbid disease burden, or poor functional status (often caused by underlying MPN). Even among those well enough to undergo alloHSCT, the long-term outlook remains disappointing due to toxicity and refractory/relapsed disease, with expected 5 year OS of less than 50% in most studies (85-89).
Enhancement of anti-tumor immunity represents one of the most exciting recent advances in oncology. Many solid tumor and hematologic malignancies have evolved mechanisms by which they avoid immune recognition. Under normal circumstances, T cell surface receptors such as cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and programmed cell death protein 1 (PD-1) interact with their associated ligands (i.e. PD-L1) and act as checkpoints to recognize “self” and prevent activation of effector T cells (90). Malignant cells often aberrantly express these ligands, thereby selectively evading immune recognition.
Clinical experience with immune checkpoint inhibitors in several solid tumor malignancies and Hodgkin lymphoma has been encouraging and has led to FDA approval of several agents. In AML and MDS, modest clinical responses have been reported with anti-CTLA4 and anti-PD-L1/anti-PD-1 agents alone and in combination with hypomethylating agents (91, 92). The role of immune checkpoint inhibitors in MF remains unproven, however several clinical studies are ongoing. These include a single-center phase 1 study of anti-PD-L1 monoclonal antibody durvalumab (NCT02871323), which is now closed to accrual and a phase 2 study of anti-PD-1 antibody nivolumab (NCT02421354), both in patients with MF after JAK inhibitor failure, intolerance, or ineligibility. A phase 2, multi-center study of anti-PD-1 agent pembrolizumab in advanced MF is also planned (NCT03065400).
Immune checkpoint inhibition may hold some promise in the post-transplant relapse setting. A phase 1 study of anti-CTLA4 agent ipilimumab in patients with a variety of hematologic malignancies who experienced disease relapse after alloHSCT reported several complete responses (5/22 treated at the effective dose level), in addition to several partial responses (2/22) and several decreases in tumor burden in patients who did not qualify as responders (6/22) (93). Of note, only one patient with an MPN was included in this study, and that patient did not experience an objective response. A phase 1 study of either ipilimumab or nivolumab in patients with relapse of a hematologic malignancy (including MPN) after alloHSCT is ongoing (NCT01822509).
Outside of immune checkpoint inhibitors, other antigen-specific immunotherapies including monoclonal antibodies, antibody-drug conjugates, cancer vaccines, and chimeric antigen receptor T-cell (CAR-T) therapies have proven effective for various solid tumor and lymphoid malignancies. However, such strategies have been problematic in myeloid malignancies. The antigenic heterogeneity and antigen shift over time that is characteristic of myeloid disorders presents a significant challenge to the development of antigen-targeted therapies (94). In addition, immune-mediated toxicities may be particularly limiting in patients with myeloid malignancies who are often older and less fit at diagnosis. Despite these limitations, several antibody-drug conjugates have shown promise in AML, and Natural Killer Group 2D (NKG2D) CAR-T cell therapy is under investigation for AML and MDS (NCT02203825) (95). It has yet to be seen whether similar therapies may have a role in the treatment of MF.
Other Novel Agents
Proviral integrations of Moloney virus (PIM) kinases are a family of serine/threonine kinases that regulate JAK/STAT signaling (96). In addition to affecting the JAK/STAT pathway, the PIM kinases also contribute to oncogenesis through phosphorylation of cell cycle regulators, activation of anti-apoptotic proteins, and enhancement of MYC expression (97-99). PIM kinases appear to be important in MPN pathogenesis, and may represent a therapeutic target. Two family members, PIM1 and PIM2, have been found to be upregulated in MPN (100). PIM inhibitors have shown preclinical synergy with JAK inhibitors, as well as the ability to overcome JAK inhibitor resistance in MPN cell lines (101, 102). A phase 1b study of ruxolitinib plus PIM inhibitor PIM447, or ruxolitinib plus CDK4/6 inhibitor ribocicilib (LEE011), or the combination of all three is underway in several non-U.S. countries (NCT02370706). Other kinase inhibitors under investigation in MF include the aurora kinase inhibitor-alisertib (Table2), based on its potential role in megakaryocytic differentiation in MF.
Anti-apoptotic proteins represent another potential target for MF therapy. Members of the B-cell lymphoma 2 (BCL-2) family of proteins inhibit the mitochondrial apoptosis pathway and promote erythropoietin-independent erythropoiesis in MPN (103). Activation of the JAK/STAT pathway mediates the transcription of BCL-2 family proteins and therefore contributes to anti-apoptotic signaling (104). The BCL-2 inhibitor venetoclax has shown activity in AML as a single agent and in combinations, and has received an FDA breakthrough therapy designation for this indication (105). Obatoclax, a pan-BCL-2 inhibitor, was studied in 22 patients with MF (106). Clinical activity was minimal, with no complete or partial responses though 1 patient (4%) experienced hematologic improvement. In mouse models of JAK2 mutant MPNs, combined targeting of JAK and BCL-2 family proteins led to disease regression, and was able to overcome resistance to single-agent JAK inhibition (107). Combination studies with JAK inhibitors or HMAs may be useful for the future and are in development, however the myelosuppressive potential of BCL-2 inhibitors may be limiting in patients with MF.
Novel Nonpharmacologic Approaches
Adjuvant nonpharmacologic psychosocial and lifestyle interventions have anecdotally shown promise in decreasing symptom burden in patients with MPNs, including MF. Interest in formally studying these interventions has piqued in recent years.
Physical activity during cancer treatment has been shown to improve various quality of life measures (108, 109). These benefits are likely generalizable to hematologic malignancies including MPNs (110). In a feasibility study of an online-streaming yoga program, 244 patients with MPNs were asked to perform 60 minutes of yoga per week over 12 weeks, following instructional yoga videos designed either specifically for MPN patients or with splenomegaly in mind (111). Actual yoga participation averaged about 51 minutes per week, and was associated with significant improvements in total symptom burden, fatigue, depression, anxiety, and sleep. A subsequent randomized study utilizing an at-home yoga program is planned, with endpoints including symptom measures, activity levels as measured by Fitbit tracking, and cytokine assessments.
The role of diet in MPNs remains largely unexplored. Certain dietary patterns have been associated with lower levels of proinflammatory cytokines, however it remains unclear whether these findings can be exploited for clinical benefit (112-114).
Mood disturbances such as anxiety and depression are common among patients with MPNs (115). No prospective studies have evaluated pharmacologic or non-pharmacologic methods of addressing mood disturbances in this population. Acceptance and commitment therapy (ACT), a multi-pronged psychosocial intervention, has demonstrated utility in several cancers and chronic medical and psychiatric conditions (116-118). A feasibility and health-related quality of life study of ACT in patients with MPNs is planned.
Conclusion
Discoveries of molecular mechanisms of MPN pathogenesis have led to the development of the first targeted therapy for MF, ruxolitinib. While ruxolitinib improves symptoms and splenomegaly with modest effects on survival, significant areas of unmet therapeutic need remain within this heterogeneous disease and future research should be aimed at filling these gaps. First, JAK pathway inhibitors should be developed and utilized to maximize clinical benefit. Cytopenias prevent many patients from receiving ruxolitinib, and therefore a second generation of JAK pathway inhibitors with less myelosuppressive potential is needed. Even in those who do receive ruxolitinib and achieve clinical benefit, the MF clone persists in nearly all and drug resistance eventually develops in most. Prevention and management of this resistance with novel agents or combinations is needed. Efforts to develop more potent and specific inhibitors of mutant JAK2 are ongoing, however promising clinical candidates have yet to emerge (119).
Second, methods to selectively target and eradicate the underlying malignant clone in MF must be prioritized. Select molecular, epigenetic, and immunologic targets under clinical investigation and their associated pharmacotherapies are depicted in figure 1, but thus far all of these approaches fall short of inducing deep molecular responses across subgroups of patients. A better understanding of the roles of immune dysregulation and the stem cell microenvironment in MF is needed to guide further therapeutic development. Gene editing may be a future direction for MPN research, however biological, technical, and ethical issues limit clinical applications at the present time (120, 121).
Figure 1.
Molecular targets for myelofibrosis and their associated agents that have shown promise or are under investigation. Multiple signaling cascades have been implicated in the pathogenesis of MF, including JAK/STAT, PI3K/AKT/mTOR, RAF/MEK/ERK, and Hedgehog (through smoothened receptor SMO). Small molecule inhibitors of various steps in these pathways have either shown a signal of clinical efficacy for MF, or are in clinical development. Targets include JAK (ruxolitinib, pacritinib, momelotinib, NS-018, itacitinib), PI3K (buparlisib, INCB050465, TGR-1202), mTOR (everolimus), or SMO (vismodegib, sonidegib, glasdegib). Epigenetic modulators such as hypomethylating agents (HMAs; azacitidine, decitabine, SGI-110) and histone deacetylase inhibitors (HDAC-i; panobinostat, pracinostat, vorinostat, givinostat) have shown activity in MF, and research continues into optimal dose and combinations of these agents with JAK inhibitors. Inhibitors of immune checkpoint receptors CTLA4 (ipilimumab) and PD-1 (pembrolizumab and nivolumab) or ligand PD-L1 (durvalumab) are also under investigation in various myeloid malignancies including MF. Other ongoing combination studies include small molecule cell cycle inhibitors of PIM1/2 (PIM447) and CDK4/6 (ribocicilib) with JAK inhibitors. Other novel targets in MF include telomerase (imetelstat), pentraxin-2 (PTX; PRM-151), and TGFβ (sotatercept).
Despite these challenges, the current pace of drug development for MF provides cause for excitement. A search of open, interventional studies for MF returned a list of 131 current ongoing clinical trials (122). Table 2 illustrates the breadth of ongoing trials of novel agents for MF therapy. In addition to pharmacotherapy, psychosocial and lifestyle interventions will likely prove integral to MF management. The phenotypic heterogeneity of MF necessitates a heterogeneous set of treatment options, and a deeper understanding of disease biology will be key to individualizing these treatment plans and improving outcomes for patients with MF.
Acknowledgments
KP was supported by the National Institutes of Health/National Institute of General Medical Sciences Clinical Therapeutics grant (T32 GM007019).
Contributor Information
Kristen Pettit, Clinical Instructor, Section of Hematology/Oncology, Department of Medicine, The University of Chicago, 5841 S. Maryland Ave, MC2115, Chicago, IL 60637, Phone (773) 702-8623, Fax (773) 702-3163.
Olatoyosi Odenike, Associate Professor of Medicine, Section of Hematology/Oncology, Department of Medicine, The University of Chicago, 5841 S. Maryland Ave, MC2115, Chicago, IL 60637.
References
- 1.Mesa RA, Kiladjian JJ, Verstovsek S, Al-Ali HK, Gotlib J, Gisslinger H, et al. Comparison of placebo and best available therapy for the treatment of myelofibrosis in the phase 3 COMFORT studies. Haematologica. 2014 Feb;99(2):292–8. doi: 10.3324/haematol.2013.087650. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Mesa R, Miller CB, Thyne M, Mangan J, Goldberger S, Fazal S, et al. Myeloproliferative neoplasms (MPNs) have a significant impact on patients’ overall health and productivity: the MPN Landmark survey. BMC cancer. 2016 Feb 27;16:167. doi: 10.1186/s12885-016-2208-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Geyer HL, Andreasson B, Kosiorek HE, Dueck AC, Scherber RM, Martin KA, et al. The role of sexuality symptoms in myeloproliferative neoplasm symptom burden and quality of life: An analysis by the MPN QOL International Study Group. Cancer. 2016 Jun 15;122(12):1888–96. doi: 10.1002/cncr.30013. [DOI] [PubMed] [Google Scholar]
- 4•.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 Jul;28(7):1472–7. doi: 10.1038/leu.2014.3. This paper identifies high risk molecular mutations in MF. [DOI] [PubMed] [Google Scholar]
- 5•.Rumi E, Pietra D, Pascutto C, Guglielmelli P, Martinez-Trillos A, Casetti I, et al. Clinical effect of driver mutations of JAK2, CALR, or MPL in primary myelofibrosis. Blood. 2014 Aug 14;124(7):1062–9. doi: 10.1182/blood-2014-05-578435. This paper identifies es prognostic implications of driver mutations in PMF. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Vainchenker W, Kralovics R. Genetic basis and molecular pathophysiology of classical myeloproliferative neoplasms. Blood. 2017 Feb 09;129(6):667–79. doi: 10.1182/blood-2016-10-695940. [DOI] [PubMed] [Google Scholar]
- 7.Mesa RA, Silverstein MN, Jacobsen SJ, Wollan PC, Tefferi A. Population-based incidence and survival figures in essential thrombocythemia and agnogenic myeloid metaplasia: an Olmsted County Study, 1976-1995. American journal of hematology. 1999 May;61(1):10–5. doi: 10.1002/(sici)1096-8652(199905)61:1<10::aid-ajh3>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
- 8.Harrison CN, Vannucchi AM, Kiladjian JJ, Al-Ali HK, Gisslinger H, Knoops L, et al. Long-term findings from COMFORT-II, a phase 3 study of ruxolitinib vs best available therapy for myelofibrosis. Leukemia. 2016 Aug;30(8):1701–7. doi: 10.1038/leu.2016.148. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9•.Verstovsek S, Mesa RA, Gotlib J, Gupta V, DiPersio JF, Catalano JV, et al. Long-term treatment with ruxolitinib for patients with myelofibrosis: 5-year update from the randomized, double-blind, placebo-controlled, phase 3 COMFORT-I trial. Journal of hematology & oncology. 2017 Feb 22;10(1):55. doi: 10.1186/s13045-017-0417-z. This paper provides a 5 year follow up of experience with ruxolitinib on the COMFORT-1 trial in MF underscoring both ongoing benefitin some patients as well as the fact that majority of patients are off therapy at 5 years. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.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 Feb 01;29(4):392–7. doi: 10.1200/JCO.2010.32.2446. [DOI] [PubMed] [Google Scholar]
- 11.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 Mar 04;115(9):1703–8. doi: 10.1182/blood-2009-09-245837. [DOI] [PubMed] [Google Scholar]
- 12•.Mesa R, Jamieson C, Bhatia R, Deininger MW, Gerds AT, Gojo I, et al. Myeloproliferative Neoplasms, Version 2.2017 NCCN Clinical Practice Guidelines in Oncology. Journal of the National Comprehensive Cancer Network : JNCCN. 2016 Dec;14(12):1572–611. doi: 10.6004/jnccn.2016.0169. This paper provides inaugural guideline recommendations by the NCCN for the diagnostic work up and management of patients with MPNs. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Kroger 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 May 21;125(21):3347–50. doi: 10.1182/blood-2014-10-608315. quiz 64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. The New England journal of medicine. 2012 Mar 01;366(9):799–807. doi: 10.1056/NEJMoa1110557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Harrison C, Kiladjian JJ, Al-Ali HK, Gisslinger H, Waltzman R, Stalbovskaya V, et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. The New England journal of medicine. 2012 Mar 01;366(9):787–98. doi: 10.1056/NEJMoa1110556. [DOI] [PubMed] [Google Scholar]
- 16.Cervantes F, Vannucchi AM, Kiladjian JJ, 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 Dec 12;122(25):4047–53. doi: 10.1182/blood-2013-02-485888. [DOI] [PubMed] [Google Scholar]
- 17.Deininger M, Radich J, Burn TC, Huber R, Paranagama D, Verstovsek S. The effect of long-term ruxolitinib treatment on JAK2p.V617F allele burden in patients with myelofibrosis. Blood. 2015 Sep 24;126(13):1551–4. doi: 10.1182/blood-2015-03-635235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Talpaz M, Paquette R, Afrin L, Hamburg SI, Prchal JT, Jamieson K, et al. Interim analysis of safety and efficacy of ruxolitinib in patients with myelofibrosis and low platelet counts. Journal of hematology & oncology. 2013 Oct 29;6(1):81. doi: 10.1186/1756-8722-6-81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bhagwat N, Levine RL, Koppikar P. Sensitivity and resistance of JAK2 inhibitors to myeloproliferative neoplasms. International journal of hematology. 2013 Jun;97(6):695–702. doi: 10.1007/s12185-013-1353-5. [DOI] [PubMed] [Google Scholar]
- 20•.Winter PS, Sarosiek KA, Lin KH, Meggendorfer M, Schnittger S, Letai A, et al. RAS signaling promotes resistance to JAK inhibitors by suppressing BAD-mediated apoptosis. Science signaling. 2014 Dec 23;7(357):ra122. doi: 10.1126/scisignal.2005301. This paper underscores the role of the RAS signaling pathway in mediating resistance to JAK inhibition. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Deshpande A, Reddy MM, Schade GO, Ray A, Chowdary TK, Griffin JD, et al. Kinase domain mutations confer resistance to novel inhibitors targeting JAK2V617F in myeloproliferative neoplasms. Leukemia. 2012 Apr;26(4):708–15. doi: 10.1038/leu.2011.255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Marit MR, Chohan M, Matthew N, Huang K, Kuntz DA, Rose DR, et al. Random mutagenesis reveals residues of JAK2 critical in evading inhibition by a tyrosine kinase inhibitor. PloS one. 2012;7(8):e43437. doi: 10.1371/journal.pone.0043437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Weigert O, Lane AA, Bird L, Kopp N, Chapuy B, van Bodegom D, et al. Genetic resistance to JAK2 enzymatic inhibitors is overcome by HSP90 inhibition. The Journal of experimental medicine. 2012 Feb 13;209(2):259–73. doi: 10.1084/jem.20111694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Komrokji RS, Seymour JF, Roberts AW, Wadleigh M, To LB, Scherber R, et al. Results of a phase 2 study of pacritinib (SB1518), a JAK2/JAK2(V617F) inhibitor, in patients with myelofibrosis. Blood. 2015 Apr 23;125(17):2649–55. doi: 10.1182/blood-2013-02-484832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25•.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. The Lancet Haematology. 2017 May;4(5):e225–e36. doi: 10.1016/S2352-3026(17)30027-3. This paper underscores activity of pacritinib in some patients with MF irrespective of baseline cytopenias. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Mascarenhas J, Hoffman R, Talpaz M. Results of the Persist-2 Phase 3 Study of Pacritinib (PAC) Versus Best Available Therapy (BAT), Including Ruxolitinib (RUX), in Patients with Myelofibrosis (MF) and Platelet Counts <100,000/uL. Blood. 2016;128:LBA-5. [Google Scholar]
- 27.Asshoff M, Petzer V, Warr MR, Haschka D, Tymoszuk P, Demetz E, et al. Momelotinib inhibits ACVR1/ALK2, decreases hepcidin production, and ameliorates anemia of chronic disease in rodents. Blood. 2017 Mar 30;129(13):1823–30. doi: 10.1182/blood-2016-09-740092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Mesa RA, Kiladjian JJ, Catalano JV. Phase 3 trial of momelotinib (MMB) vs ruxolitinib (RUX) in JAK inhibitor (JAKi) naive patients with myelofibrosis (MF) J Clin Oncol. 2017;35:7000. doi: 10.1200/JCO.2017.73.4418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Harrison C. Phase 3 randomized trial of momelotinib (MMB) versus best available therapy (BAT) in patients with myelofibrosis (MF) previously treated with ruxolitinib (RUX) J Clin Oncol. 2017;35:7001. [Google Scholar]
- 30.Verstovsek S, Talpaz M, Ritchie EK. Phase 1/2 Study of NS-018, an Oral JAK2 Inhibitor, in Patients with Primary Myelofibrosis (PMF), Post-Polycythemia Vera Myelofibrosis (postPV MF), or Post-Essential Thrombocythemia Myelofibrosis (postET MF) Blood. 2016;634:1936. [Google Scholar]
- 31.Mascarenhas JO, Talpaz M, Gupta V, Foltz LM, Savona MR, Paquette R, et al. Primary analysis of a phase II open-label trial of INCB039110, a selective JAK1 inhibitor, in patients with myelofibrosis. Haematologica. 2017 Feb;102(2):327–35. doi: 10.3324/haematol.2016.151126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Jones PA, Baylin SB. The epigenomics of cancer. Cell. 2007 Feb 23;128(4):683–92. doi: 10.1016/j.cell.2007.01.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Wang JC, Chen W, Nallusamy S, Chen C, Novetsky AD. Hypermethylation of the P15INK4b and P16INK4a in agnogenic myeloid metaplasia (AMM) and AMM in leukaemic transformation. British journal of haematology. 2002 Mar;116(3):582–6. doi: 10.1046/j.0007-1048.2001.03319.x. [DOI] [PubMed] [Google Scholar]
- 34.Kumagai T, Tefferi A, Jones L, Koeffler HP. Methylation analysis of the cell cycle control genes in myelofibrosis with myeloid metaplasia. Leukemia research. 2005 May;29(5):511–5. doi: 10.1016/j.leukres.2004.11.002. [DOI] [PubMed] [Google Scholar]
- 35.Quintas-Cardama A, Tong W, Kantarjian H, Thomas D, Ravandi F, Kornblau S, et al. A phase II study of 5-azacitidine for patients with primary and post-essential thrombocythemia/polycythemia vera myelofibrosis. Leukemia. 2008 May;22(5):965–70. doi: 10.1038/leu.2008.91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Mesa RA, Verstovsek S, Rivera C, Pardanani A, Hussein K, Lasho T, et al. 5-Azacitidine has limited therapeutic activity in myelofibrosis. Leukemia. 2009 Jan;23(1):180–2. doi: 10.1038/leu.2008.136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Odenike O, Godwin J, van Besien KM. Phase II Trial of Low Dose, Subcutaneous Decitabine in Myelofibrosis. Blood. 2008;112:2809. doi: 10.1182/bloodadvances.2024013215. [DOI] [PubMed] [Google Scholar]
- 38.Badar T, Kantarjian HM, Ravandi F, Jabbour E, Borthakur G, Cortes JE, et al. Therapeutic benefit of decitabine, a hypomethylating agent, in patients with high-risk primary myelofibrosis and myeloproliferative neoplasm in accelerated or blastic/acute myeloid leukemia phase. Leukemia research. 2015 Sep;39(9):950–6. doi: 10.1016/j.leukres.2015.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Danilov AV, Relias V, Feeney DM, Miller KB. Decitabine is an effective treatment of idiopathic myelofibrosis. British journal of haematology. 2009 Apr;145(1):131–2. doi: 10.1111/j.1365-2141.2008.07541.x. [DOI] [PubMed] [Google Scholar]
- 40.Tabarroki A, Saunthararajah Y, Visconte V, Cinalli T, Colaluca K, Rogers HJ, et al. Ruxolitinib in combination with DNA methyltransferase inhibitors: clinical responses in patients with symptomatic myelofibrosis with cytopenias and elevated blast(s) counts. Leukemia & lymphoma. 2015 Feb;56(2):497–9. doi: 10.3109/10428194.2014.916805. [DOI] [PubMed] [Google Scholar]
- 41.Daver N, Cortes J, Pemmaraju N. Ruxolitinib (RUX) in Combination with 5-Azacytidine (AZA) As Therapy for Patients (pts) with Myelofibrosis (MF) Blood. 2016;128:1127. [Google Scholar]
- 42.Thepot S, Itzykson R, Seegers V, Raffoux E, Quesnel B, Chait Y, et al. Treatment of progression of Philadelphia-negative myeloproliferative neoplasms to myelodysplastic syndrome or acute myeloid leukemia by azacitidine: a report on 54 cases on the behalf of the Groupe Francophone des Myelodysplasies (GFM) Blood. 2010 Nov 11;116(19):3735–42. doi: 10.1182/blood-2010-03-274811. [DOI] [PubMed] [Google Scholar]
- 43.Mascarenhas J, Navada S, Malone A, Rodriguez A, Najfeld V, Hoffman R. Therapeutic options for patients with myelofibrosis in blast phase. Leukemia research. 2010 Sep;34(9):1246–9. doi: 10.1016/j.leukres.2010.05.008. [DOI] [PubMed] [Google Scholar]
- 44.Bose P, Verstovsek S, Gasior Y. Phase I/II Study of Ruxolitinib (RUX) with Decitabine (DAC) in Patients with Post-Myeloproliferative Neoplasm Acute Myeloid Leukemia (post-MPN AML): Phase I Results. Blood. 2016;128:4262. [Google Scholar]
- 45.Rampal R, Mascarenhas J, Kosiorek HE. Safety and Efficacy of Combined Ruxolitinib and Decitabine in Patients with Blast-Phase MPN and Post-MPN AML: Results of a Phase I Study (Myeloproliferative Disorders Research Consortium 109 trial) Blood. 2016;128:1124. [Google Scholar]
- 46.Andersen CL, Mortensen NB, Klausen TW, Vestergaard H, Bjerrum OW, Hasselbalch HC. A phase II study of vorinostat (MK-0683) in patients with primary myelofibrosis and post-polycythemia vera myelofibrosis. Haematologica. 2014 Jan;99(1):5–7. doi: 10.3324/haematol.2013.096669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.DeAngelo DJ, Mesa RA, Fiskus W, Tefferi A, Paley C, Wadleigh M, et al. Phase II trial of panobinostat, an oral pan-deacetylase inhibitor in patients with primary myelofibrosis, post-essential thrombocythaemia, and post-polycythaemia vera myelofibrosis. British journal of haematology. 2013 Aug;162(3):326–35. doi: 10.1111/bjh.12384. [DOI] [PubMed] [Google Scholar]
- 48.Mascarenhas J, Lu M, Li T, Petersen B, Hochman T, Najfeld V, et al. A phase I study of panobinostat (LBH589) in patients with primary myelofibrosis (PMF) and post-polycythaemia vera/essential thrombocythaemia myelofibrosis (post-PV/ET MF) British journal of haematology. 2013 Apr;161(1):68–75. doi: 10.1111/bjh.12220. [DOI] [PubMed] [Google Scholar]
- 49.Mascarenhas J, Sandy L, Lu M, Yoon J, Petersen B, Zhang D, et al. A phase II study of panobinostat in patients with primary myelofibrosis (PMF) and post-polycythemia vera/essential thrombocythemia myelofibrosis (post-PV/ET MF) Leukemia research. 2017 Feb;53:13–9. doi: 10.1016/j.leukres.2016.11.015. [DOI] [PubMed] [Google Scholar]
- 50.Rambaldi A, Dellacasa CM, Finazzi G, Carobbio A, Ferrari ML, Guglielmelli P, et al. A pilot study of the Histone-Deacetylase inhibitor Givinostat in patients with JAK2V617F positive chronic myeloproliferative neoplasms. British journal of haematology. 2010 Aug;150(4):446–55. doi: 10.1111/j.1365-2141.2010.08266.x. [DOI] [PubMed] [Google Scholar]
- 51.Quintas-Cardama A, Kantarjian H, Estrov Z, Borthakur G, Cortes J, Verstovsek S. Therapy with the histone deacetylase inhibitor pracinostat for patients with myelofibrosis. Leukemia research. 2012 Sep;36(9):1124–7. doi: 10.1016/j.leukres.2012.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Harrison C, Kiladjian JJ, Heidel FH. Efficacy, Safety, and Confirmation of the Recommended Phase 2 Starting Dose of the Combination of Ruxolitinib (RUX) and Panobinostat (PAN) in Patients (Pts) with Myelofibrosis (MF) Blood. 2015;126:4060. [Google Scholar]
- 53.Grimwade LF, Happerfield L, Tristram C, McIntosh G, Rees M, Bench AJ, et al. Phospho-STAT5 and phospho-Akt expression in chronic myeloproliferative neoplasms. British journal of haematology. 2009 Nov;147(4):495–506. doi: 10.1111/j.1365-2141.2009.07870.x. [DOI] [PubMed] [Google Scholar]
- 54.Bartalucci N, Tozzi L, Bogani C, Martinelli S, Rotunno G, Villeval JL, et al. Co-targeting the PI3K/mTOR and JAK2 signalling pathways produces synergistic activity against myeloproliferative neoplasms. Journal of cellular and molecular medicine. 2013 Nov;17(11):1385–96. doi: 10.1111/jcmm.12162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Fiskus W, Verstovsek S, Manshouri T, Smith JE, Peth K, Abhyankar S, et al. Dual PI3K/AKT/mTOR inhibitor BEZ235 synergistically enhances the activity of JAK2 inhibitor against cultured and primary human myeloproliferative neoplasm cells. Molecular cancer therapeutics. 2013 May;12(5):577–88. doi: 10.1158/1535-7163.MCT-12-0862. [DOI] [PubMed] [Google Scholar]
- 56.Khan I, Huang Z, Wen Q, Stankiewicz MJ, Gilles L, Goldenson B, et al. AKT is a therapeutic target in myeloproliferative neoplasms. Leukemia. 2013 Sep;27(9):1882–90. doi: 10.1038/leu.2013.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Guglielmelli P, Barosi G, Rambaldi A, Marchioli R, Masciulli A, Tozzi L, et al. Safety and efficacy of everolimus, a mTOR inhibitor, as single agent in a phase 1/2 study in patients with myelofibrosis. Blood. 2011 Aug 25;118(8):2069–76. doi: 10.1182/blood-2011-01-330563. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Durrant ST, Nagler A, Vannucchi AM. An Open-Label, Multicenter, 2-Arm, Dose-Finding, Phase 1b Study of the Combination of Ruxolitinib and Buparlisib (BKM120) in Patients with Myelofibrosis: Results from HARMONY Study. Blood. 2015;126:827. doi: 10.3324/haematol.2018.209965. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.McCubrey JA, Steelman LS, Abrams SL, Bertrand FE, Ludwig DE, Basecke J, et al. Targeting survival cascades induced by activation of Ras/Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways for effective leukemia therapy. Leukemia. 2008 Apr;22(4):708–22. doi: 10.1038/leu.2008.27. [DOI] [PubMed] [Google Scholar]
- 60.Steelman LS, Abrams SL, Whelan J, Bertrand FE, Ludwig DE, Basecke J, et al. Contributions of the Raf/MEK/ERK, PI3K/PTEN/Akt/mTOR and Jak/STAT pathways to leukemia. Leukemia. 2008 Apr;22(4):686–707. doi: 10.1038/leu.2008.26. [DOI] [PubMed] [Google Scholar]
- 61.Oku S, Takenaka K, Kuriyama T, Shide K, Kumano T, Kikushige Y, et al. JAK2 V617F uses distinct signalling pathways to induce cell proliferation and neutrophil activation. British journal of haematology. 2010 Aug;150(3):334–44. doi: 10.1111/j.1365-2141.2010.08249.x. [DOI] [PubMed] [Google Scholar]
- 62.Nguyen TK, Tata P, Brooks S. The MEK/ERK Inhibitor Trametinib Reduces Fibrosis in a Transduction-Transplantation Model of Mutated Calreticulin. Blood. 2016;128:635. [Google Scholar]
- 63.Kong G, Wunderlich M, Yang D, Ranheim EA, Young KH, Wang J, et al. Combined MEK and JAK inhibition abrogates murine myeloproliferative neoplasm. The Journal of clinical investigation. 2014 Jun;124(6):2762–73. doi: 10.1172/JCI74182. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Jain N, Curran E, Iyengar NM, Diaz-Flores E, Kunnavakkam R, Popplewell L, et al. Phase II study of the oral MEK inhibitor selumetinib in advanced acute myelogenous leukemia: aUniversity of Chicago phase II consortium trial. Clinical cancer research : an official journal of the American Association for Cancer Research. 2014 Jan 15;20(2):490–8. doi: 10.1158/1078-0432.CCR-13-1311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Borthakur G, Popplewell L, Boyiadzis M, Foran J, Platzbecker U, Vey N, et al. Activity of the oral mitogen-activated protein kinase kinase inhibitor trametinib in RAS-mutant relapsed or refractory myeloid malignancies. Cancer. 2016 Jun 15;122(12):1871–9. doi: 10.1002/cncr.29986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.McGowan SE, McCoy DM. Platelet-derived growth factor-A and sonic hedgehog signaling direct lung fibroblast precursors during alveolar septal formation. American journal of physiology Lung cellular and molecular physiology. 2013 Aug 01;305(3):L229–39. doi: 10.1152/ajplung.00011.2013. [DOI] [PubMed] [Google Scholar]
- 67.Greenbaum LE. Hedgehog signaling in biliary fibrosis. The Journal of clinical investigation. 2008 Oct;118(10):3263–5. doi: 10.1172/JCI37189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Gerds A, Tauchi T, Ritchie E. Phase I/II trial of glasdegib in patients with primary or secondary myelofibrosis. J Clin Oncol. 2017;35(suppl) abstr 7061. [Google Scholar]
- 69.Gupta V, Harrison C, Hasselbalch H. Phase 1b/2 Study of the Efficacy and Safety of Sonidegib (LDE225) in Combination with Ruxolitinib (INC424) in Patients with Myelofibrosis. Blood. 2015;126:825. [Google Scholar]
- 70.Blackburn EH, Greider CW, Szostak JW. Telomeres and telomerase: the path from maize, Tetrahymena and yeast to human cancer and aging. Nature medicine. 2006 Oct;12(10):1133–8. doi: 10.1038/nm1006-1133. [DOI] [PubMed] [Google Scholar]
- 71.Asai A, Oshima Y, Yamamoto Y, Uochi TA, Kusaka H, Akinaga S, et al. A novel telomerase template antagonist (GRN163) as a potential anticancer agent. Cancer research. 2003 Jul 15;63(14):3931–9. [PubMed] [Google Scholar]
- 72.Herbert BS, Gellert GC, Hochreiter A, Pongracz K, Wright WE, Zielinska D, et al. Lipid modification of GRN163, an N3′–>P5′ thio-phosphoramidate oligonucleotide, enhances the potency of telomerase inhibition. Oncogene. 2005 Aug 04;24(33):5262–8. doi: 10.1038/sj.onc.1208760. [DOI] [PubMed] [Google Scholar]
- 73•.Tefferi A, Lasho TL, Begna KH, Patnaik MM, Zblewski DL, Finke CM, et al. A Pilot Study of the Telomerase Inhibitor Imetelstat for Myelofibrosis. The New England journal of medicine. 2015 Sep 03;373(10):908–19. doi: 10.1056/NEJMoa1310523. This paper documents the potential activity of the telomerase inhibitor inhibitor imetelstat in myelofibrosis and underscores the need for additional studies. [DOI] [PubMed] [Google Scholar]
- 74.Baerlocher GM, Burington B, Snyder DS. Telomerase Inhibitor Imetelstat in Essential Thrombocythemia and Myelofibrosis. The New England journal of medicine. 2015 Dec 24;373(26):2580. doi: 10.1056/NEJMc1512663. [DOI] [PubMed] [Google Scholar]
- 75.Dillingh MR, van den Blink B, Moerland M, van Dongen MG, Levi M, Kleinjan A, et al. Recombinant human serum amyloid P in healthy volunteers and patients with pulmonary fibrosis. Pulmonary pharmacology & therapeutics. 2013 Dec;26(6):672–6. doi: 10.1016/j.pupt.2013.01.008. [DOI] [PubMed] [Google Scholar]
- 76.Pilling D, Buckley CD, Salmon M, Gomer RH. Inhibition of fibrocyte differentiation by serum amyloid P. Journal of immunology. 2003 Nov 15;171(10):5537–46. doi: 10.4049/jimmunol.171.10.5537. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Verstovsek S, Manshouri T, Pilling D, Bueso-Ramos CE, Newberry KJ, Prijic S, et al. Role of neoplastic monocyte-derived fibrocytes in primary myelofibrosis. The Journal of experimental medicine. 2016 Aug 22;213(9):1723–40. doi: 10.1084/jem.20160283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Verstovsek S, Mesa R, Foltz L. Phase 2 Trial of PRM-151, an Anti-Fibrotic Agent, in Patients with Myelofibrosis: Stage 1 Results. Blood. 2014;124:713. [Google Scholar]
- 79.Tadmor T, Bejar J, Attias D, Mischenko E, Sabo E, Neufeld G, et al. The expression of lysyl-oxidase gene family members in myeloproliferative neoplasms. American journal of hematology. 2013 May;88(5):355–8. doi: 10.1002/ajh.23409. [DOI] [PubMed] [Google Scholar]
- 80.Eliades A, Papadantonakis N, Bhupatiraju A, Burridge KA, Johnston-Cox HA, Migliaccio AR, et al. Control of megakaryocyte expansion and bone marrow fibrosis by lysyl oxidase. The Journal of biological chemistry. 2011 Aug 05;286(31):27630–8. doi: 10.1074/jbc.M111.243113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Verstovsek S, Savona MR, Mesa RA, Dong H, Maltzman JD, Sharma S, et al. A phase 2 study of simtuzumab in patients with primary, post-polycythaemia vera or post-essential thrombocythaemia myelofibrosis. British journal of haematology. 2017 Mar;176(6):939–49. doi: 10.1111/bjh.14501. [DOI] [PubMed] [Google Scholar]
- 82.Le Bousse-Kerdiles MC, Martyre MC. Dual implication of fibrogenic cytokines in the pathogenesis of fibrosis and myeloproliferation in myeloid metaplasia with myelofibrosis. Annals of hematology. 1999 Oct;78(10):437–44. doi: 10.1007/s002770050595. [DOI] [PubMed] [Google Scholar]
- 83.Bose P, Daver N, Jabbour E. Phase-2 Study of Sotatercept (ACE-011) in Myeloproliferative Neoplasm-Associated Myelofibrosis and Anemia. Blood. 2016;128:478. [Google Scholar]
- 84.Mascarenhas J, Li T, Sandy L, Newsom C, Petersen B, Godbold J, et al. Anti-transforming growth factor-beta therapy in patients with myelofibrosis. Leukemia & lymphoma. 2014 Feb;55(2):450–2. doi: 10.3109/10428194.2013.805329. [DOI] [PubMed] [Google Scholar]
- 85.Ditschkowski M, Elmaagacli AH, Trenschel R, Gromke T, Steckel NK, Koldehoff M, et al. Dynamic International Prognostic Scoring System scores, pre-transplant therapy and chronic graft-versus-host disease determine outcome after allogeneic hematopoietic stem cell transplantation for myelofibrosis. Haematologica. 2012 Oct;97(10):1574–81. doi: 10.3324/haematol.2011.061168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Scott BL, Gooley TA, Sorror ML, Rezvani AR, Linenberger ML, Grim J, et al. The Dynamic International Prognostic Scoring System for myelofibrosis predicts outcomes after hematopoietic cell transplantation. Blood. 2012 Mar 15;119(11):2657–64. doi: 10.1182/blood-2011-08-372904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Alchalby H, Yunus DR, Zabelina T, Kobbe G, Holler E, Bornhauser M, et al. Risk models predicting survival after reduced-intensity transplantation for myelofibrosis. British journal of haematology. 2012 Apr;157(1):75–85. doi: 10.1111/j.1365-2141.2011.09009.x. [DOI] [PubMed] [Google Scholar]
- 88.Gupta V, Malone AK, Hari PN, Ahn KW, Hu ZH, Gale RP, et al. Reduced-intensity hematopoietic cell transplantation for patients with primary myelofibrosis: a cohort analysis from the center for international blood and marrow transplant research. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation. 2014 Jan;20(1):89–97. doi: 10.1016/j.bbmt.2013.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89•.Rondelli D, Goldberg JD, Isola L, Price LS, Shore TB, Boyer M, et al. MPD-RC 101 prospective study of reduced-intensity allogeneic hematopoietic stem cell transplantation in patients with myelofibrosis. Blood. 2014 Aug 14;124(7):1183–91. doi: 10.1182/blood-2014-04-572545. This paper documents the potential for reduced intensity conditioning to induce durable remssions post allogeneic stem cell transplantation for myelofibrosis. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Choi DC, Tremblay D, Iancu-Rubin C, Mascarenhas J. Programmed cell death-1 pathway inhibition in myeloid malignancies: implications for myeloproliferative neoplasms. Annals of hematology. 2017 Jun;96(6):919–27. doi: 10.1007/s00277-016-2915-4. [DOI] [PubMed] [Google Scholar]
- 91.Garcia-Manero G, Tallman M, Martinelli G. Pembrolizumab, a PD-1 Inhibitor, in Patients with Myelodysplastic Syndrome (MDS) after Failure of Hypomethylating Agent Treatment. Oral Abstract American Society of Hematology Annual Meeting; Dec 4, 2016. [Google Scholar]
- 92.Garcia-Manero G, Daver N, Montalban-Bravo G. A Phase II Study Evaluating the Combination of Nivolumab (Nivo) or Ipilimumab (Ipi) with Azacitidine in Pts with Previously Treated or Untreated Myelodysplastic Syndromes (MDS). Oral Abstract American Society of Hematology Annual Meeting; Dec 4, 2016. [Google Scholar]
- 93.Davids MS, Kim HT, Bachireddy P, Costello C, Liguori R, Savell A, et al. Ipilimumab for Patients with Relapse after Allogeneic Transplantation. The New England journal of medicine. 2016 Jul 14;375(2):143–53. doi: 10.1056/NEJMoa1601202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Rashidi A, Walter RB. Antigen-specific immunotherapy for acute myeloid leukemia: where are we now, and where do we go from here? Expert review of hematology. 2016;9(4):335–50. doi: 10.1586/17474086.2016.1142868. [DOI] [PubMed] [Google Scholar]
- 95.Nikiforow S, Werner L, Murad J. Safety Data from a First-in-Human Phase 1 Trial of NKG2D Chimeric Antigen Receptor-T Cells in AML/MDS and Multiple Myeloma. Poster Abstract American Society of Hematology Annual Meeting; Dec 5, 2016. [Google Scholar]
- 96.Brault L, Gasser C, Bracher F, Huber K, Knapp S, Schwaller J. PIM serine/threonine kinases in the pathogenesis and therapy of hematologic malignancies and solid cancers. Haematologica. 2010 Jun;95(6):1004–15. doi: 10.3324/haematol.2009.017079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Hammerman PS, Fox CJ, Birnbaum MJ, Thompson CB. Pim and Akt oncogenes are independent regulators of hematopoietic cell growth and survival. Blood. 2005 Jun 01;105(11):4477–83. doi: 10.1182/blood-2004-09-3706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Berns A, Mikkers H, Krimpenfort P, Allen J, Scheijen B, Jonkers J. Identification and characterization of collaborating oncogenes in compound mutant mice. Cancer research. 1999 Apr 01;59(7 Suppl):1773s–7s. [PubMed] [Google Scholar]
- 99.Nawijn MC, Alendar A, Berns A. For better or for worse: the role of Pim oncogenes in tumorigenesis. Nature reviews Cancer. 2011 Jan;11(1):23–34. doi: 10.1038/nrc2986. [DOI] [PubMed] [Google Scholar]
- 100.Wernig G, Gonneville JR, Crowley BJ, Rodrigues MS, Reddy MM, Hudon HE, et al. The Jak2V617F oncogene associated with myeloproliferative diseases requires a functional FERM domain for transformation and for expression of the Myc and Pim proto-oncogenes. Blood. 2008 Apr 01;111(7):3751–9. doi: 10.1182/blood-2007-07-102186. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Mazzacurati L, Lambert QT, Pradhan A, Griner LN, Huszar D, Reuther GW. The PIM inhibitor AZD1208 synergizes with ruxolitinib to induce apoptosis of ruxolitinib sensitive and resistant JAK2-V617F-driven cells and inhibit colony formation of primary MPN cells. Oncotarget. 2015 Nov 24;6(37):40141–57. doi: 10.18632/oncotarget.5653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Huang SM, Wang A, Greco R, Li Z, Barberis C, Tabart M, et al. Combination of PIM and JAK2 inhibitors synergistically suppresses MPN cell proliferation and overcomes drug resistance. Oncotarget. 2014 May 30;5(10):3362–74. doi: 10.18632/oncotarget.1951. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Silva M, Richard C, Benito A, Sanz C, Olalla I, Fernandez-Luna JL. Expression of Bcl-x in erythroid precursors from patients with polycythemia vera. The New England journal of medicine. 1998 Feb 26;338(9):564–71. doi: 10.1056/NEJM199802263380902. [DOI] [PubMed] [Google Scholar]
- 104.Socolovsky M, Fallon AE, Wang S, Brugnara C, Lodish HF. Fetal anemia and apoptosis of red cell progenitors in Stat5a−/−5b−/− mice: a direct role for Stat5 in Bcl-X(L) induction. Cell. 1999 Jul 23;98(2):181–91. doi: 10.1016/s0092-8674(00)81013-2. [DOI] [PubMed] [Google Scholar]
- 105.Konopleva M, Pollyea DA, Potluri J, Chyla B, Hogdal L, Busman T, et al. Efficacy and Biological Correlates of Response in a Phase II Study of Venetoclax Monotherapy in Patients with Acute Myelogenous Leukemia. Cancer discovery. 2016 Oct;6(10):1106–17. doi: 10.1158/2159-8290.CD-16-0313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Parikh SA, Kantarjian H, Schimmer A, Walsh W, Asatiani E, El-Shami K, et al. Phase II study of obatoclax mesylate (GX15070), a small-molecule BCL-2 family antagonist, for patients with myelofibrosis. Clinical lymphoma, myeloma & leukemia. 2010 Aug;10(4):285–9. doi: 10.3816/CLML.2010.n.059. [DOI] [PubMed] [Google Scholar]
- 107.Waibel M, Solomon VS, Knight DA, Ralli RA, Kim SK, Banks KM, 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 reports. 2013 Nov 27;5(4):1047–59. doi: 10.1016/j.celrep.2013.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Brown JC, Winters-Stone K, Lee A, Schmitz KH. Cancer, physical activity, and exercise. Comprehensive Physiology. 2012 Oct;2(4):2775–809. doi: 10.1002/cphy.c120005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Bade BC, Thomas DD, Scott JB, Silvestri GA. Increasing physical activity and exercise in lung cancer: reviewing safety, benefits, and application. Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer. 2015 Jun;10(6):861–71. doi: 10.1097/JTO.0000000000000536. [DOI] [PubMed] [Google Scholar]
- 110.Eckert R, Huberty J, Gowin K, Mesa R, Marks L. Physical Activity as a Nonpharmacological Symptom Management Approach in Myeloproliferative Neoplasms: Recommendations for Future Research. Integrative cancer therapies. 2016 Jul 24; doi: 10.1177/1534735416661417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Huberty J, Eckert R, Gowin K. Online-Streamed Yoga As a Non-Pharmacologic Symptom Management Approach in Myeloproliferative Neoplasms. Blood. 2016;128:5478. [Google Scholar]
- 112.Arthur AE, Peterson KE, Shen J, Djuric Z, Taylor JM, Hebert JR, et al. Diet and proinflammatory cytokine levels in head and neck squamous cell carcinoma. Cancer. 2014 Sep 01;120(17):2704–12. doi: 10.1002/cncr.28778. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Lucas R, Parikh SJ, Sridhar S, Guo DH, Bhagatwala J, Dong Y, et al. Cytokine profiling of young overweight and obese female African American adults with prediabetes. Cytokine. 2013 Oct;64(1):310–5. doi: 10.1016/j.cyto.2013.05.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Olendzki BC, Silverstein TD, Persuitte GM, Ma Y, Baldwin KR, Cave D. An anti-inflammatory diet as treatment for inflammatory bowel disease: a case series report. Nutrition journal. 2014 Jan 16;13:5. doi: 10.1186/1475-2891-13-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Scherber RM, Kosiorek HE, Senyak Z, Dueck AC, Clark MM, Boxer MA, et al. Comprehensively understanding fatigue in patients with myeloproliferative neoplasms. Cancer. 2016 Feb 01;122(3):477–85. doi: 10.1002/cncr.29753. [DOI] [PubMed] [Google Scholar]
- 116.Hulbert-Williams NJ, Storey L, Wilson KG. Psychological interventions for patients with cancer: psychological flexibility and the potential utility of Acceptance and Commitment Therapy. European journal of cancer care. 2015;24(1):15–27. doi: 10.1111/ecc.12223. [DOI] [PubMed] [Google Scholar]
- 117.Feros DL, Lane L, Ciarrochi J, Blackledge JT. Acceptance and Commitment Therapy (ACT) for improving the lives of cancer patients: a preliminary study. Psycho-oncology. 2013 Feb;22(2):459–64. doi: 10.1002/pon.2083. [DOI] [PubMed] [Google Scholar]
- 118.Kangas M, McDonald S, Williams JR, Smee RI. Acceptance and commitment therapy program for distressed adults with a primary brain tumor: a case series study. Supportive care in cancer : official journal of the Multinational Association of Supportive Care in Cancer. 2015 Oct;23(10):2855–9. doi: 10.1007/s00520-015-2804-8. [DOI] [PubMed] [Google Scholar]
- 119•.Leroy E, Constantinescu SN. Rethinking JAK2 inhibition: towards novel strategies of more specific and versatile janus kinase inhibition. Leukemia. 2017 May;31(5):1023–38. doi: 10.1038/leu.2017.43. This paper provides a comprehensive review of strategies under development for more specific JAK inhibitors. [DOI] [PubMed] [Google Scholar]
- 120.Smith C, Abalde-Atristain L, He C, Brodsky BR, Braunstein EM, Chaudhari P, et al. Efficient and allele-specific genome editing of disease loci in human iPSCs. Molecular therapy : the journal of the American Society of Gene Therapy. 2015 Mar;23(3):570–7. doi: 10.1038/mt.2014.226. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121•.Mead AJ, Mullally A. Myeloproliferative neoplasm stem cells. Blood. 2017 Mar 23;129(12):1607–16. doi: 10.1182/blood-2016-10-696005. This paper provides a contemporary review of driver mutations in MPN pathogenesis and clonal expansion and novel strategies and approaches under consideration for targeting MPN stem cells. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Search terms myelofibrosis and interventional and open. [cited Accessed 5/24/2017]. Available from: http://www.clinicaltrials.gov.