Abstract
Introduction
The discovery of the activating JAK2V617F mutation in patients with myelofibrosis (MF) led to the development of JAK2 inhibitors. The first such inhibitor to enter clinical trials was ruxolitinib. This review summarizes pre-clinical and clinical data of ruxolitinib in MF.
Areas covered
A literature search through Medline employing the terms “ruxolitinib”, “INCB018424” and “myelofibrosis” was undertaken. The results from phase I/II studies in patients with MF showed that ruxolitinib led to durable improvements in splenomegaly, and symptoms associated with MF. Two phase III trials have compared ruxolitinib against placebo and best available therapy and in both studies ruxolitinib demonstrated superior rates of spleen control and symptom improvement, and additional analysis demonstrated a survival benefit with ruxolitinib treatment. The main toxicities seen with ruxolitinib are cytopenias, which are managed with dose adjustments. Recent reports documented sporadic cases of immunosuppression-related infections. Ruxolitinib is the first drug ever approved for the therapy of patients with MF.
Expert Opinion
Understanding the factors that predict the rate and duration of response to ruxolitinib would improve our ability to manage patients treated with this medication. Clinical trials combining ruxolitinib with novel compounds that are also active in MF will further improve therapy for this disease.
Keywords: Myelofibrosis, Ruxolitinib, JAK2 V617F
1 Introduction
Myelofibrosis (MF) is a Philadelphia-negative (Ph-negative) myeloproliferative neoplasm (MPN) characterized by bone marrow fibrosis, abnormalities in peripheral blood cell counts, extramedullary hematopoiesis (particularly in the spleen) and an increased risk of transformation to acute myeloid leukemia (AML)1. It is an uncommon disease, with an incidence estimated at 1.46 cases/100,000 persons per year, and median age at diagnosis is 67 years2. MF can be primary MF (PMF) or secondary to other Ph-negative MPNs, such as polycythemia vera (post-PV MF) and essential thrombocythemia (post-ET MF)1. Patients with MF usually refer a myriad of symptoms, including extreme fatigue, fever, weight loss, night sweats, pruritus, abdominal and bone pain and have very poor quality of life3,4.
MF is a disease that has few treatment options. For patients who develop massive splenomegaly, treatment options included hydroxyurea, other chemotherapeutic agents (e.g cladribine), splenic radiation and splenectomy5. For patients who have transfusion dependent anemia, drugs such as erythropoietin, androgens (e.g. danazol), and immunomodulatory compounds (e.g. lenalidomide) can be used6. No curative drug option exists, and median survival of newly-diagnosed patients is in the range of 5–6 years7. Allogeneic stem cell transplantation is a potential curative treatment approach; however it is applicable only in a minority of patients due to risks associated with procedure8.
The scenario for MF started to change with the report in 2005 of activating mutation in the JAK2 tyrosine kinase (TK) in 60% of patients with MF and ET, and 90% of patients with PV9–12. The JAK2 TK is part of the JAK family of TKs13. JAK2 plays an essential role in hematopoiesis, as evidenced by knock-out mice studies13–15. The V617F mutation leads to increased activity of the JAK2 TK, and consequently causes increased proliferation and resistance to apoptosis in the transformed hematopoietic cell10. Subsequently, mutations in other genes that also lead to increased JAK2 TK activity, and general dysregulation of JAK-signal transducer and activator of transcription (STAT) intracellular signaling pathway were described in patients with JAK2V617F-negative MF and ET (e.g. MPL, CALR)16,17. These findings demonstrate that JAK-STAT plays an essential role in the pathogenesis of MF and other classic Ph-negative MPNs.
The discovery of JAK2V617F spurred the development of JAK2 TK inhibitors (TKIs). The first such inhibitor to be developed and tested in clinical trials was ruxolitinib (formerly known as INCB028424; Jakafi®, Incyte, Wilmington, DE; Jakavi®, Novartis AG, Basel, Switzerland [outside the US]). Ruxolitinib is an orally available, potent JAK1 and JAK2 inhibitor18. Initial clinical trials demonstrated that ruxolitinib could lead to dramatic reductions in spleen size and symptom burdens in patients with MF18. Subsequently, the results of two phase III trials confirmed the efficacy of ruxolitinib for patients with MF, and it was the first drug to be approved for this indication by the FDA in November, 201119,20. This article will review the current data on the efficacy of ruxolitinib for patients with MF.
2 Pre-clinical studies
2.1 Chemistry
Ruxolitinib phosphate is a compound of the class of pyrrolopyrimidines that has activity as tyrosine kinase inhibitor. The chemical name of ruxolitinib is (R)-3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile phosphate.21 The molecular weight of ruxolitinib phosphate is 404.36 g/mole and its molecular formula is C17H21N6O4P. Ruxolitinib phosphate is a white to off-white to light pink powder; it is soluble in aqueous buffers across a pH of 1–8.21 Tablets of ruxolitinib phosphate21 should be stored at room temperatures of 20°C to 25°C (68°F to 77°F). The synthesis of ruxolitinib has been summarized before.22
2.2 Pharmacodynamics
The pre-clinical characterization of ruxolitinib was published in 2010 by Quintás-Cardama et al23. The authors demonstrated that ruxolitinib inhibited both JAK1 and JAK2 TKs with a half-maximal inhibitory concentration of 3.3 and 2.8 nM, respectively, while maintaining modest activity against TYK2 and minimal activity against JAK323. Whole blood stimulated with cytokines (Interleukin-6 [IL-6] and thrombopoietin [TPO]) confirmed the efficacy of ruxolitinib in decreasing phosphorylation of STAT3, a substrate of both JAK1 and JAK2 after stimulation with the aforementioned cytokines.
The activity of ruxolitinib against cells harboring JAK2V617F was demonstrated against both Ba/F3 cells transformed by JAK2V617F and erythropoietin receptor (Ba/F3-EPOR-JAK2V617F cells) and HEL cells (human erythroleukemia cell line carrying the JAK2V617F mutation)23. Treatment of both cell lines with ruxolitinib reduced cell proliferation (Ba/F3 IC50=126 nM; HEL IC50=186 nM) and blocked phosphorylation of both STAT3 and STAT523. In Ba/F3-EPOR-JAK2V617F cells, treatment with ruxolitinib led to an increase in the number of apoptotic cells23.
Ruxolitinib led to a dose-dependent decrease in proliferation of erythroid progenitors from patients with PV compared to healthy controls (IC50 223 nM vs. 407 nM)23. Further evaluation demonstrated that ruxolitinib also inhibited endogenous EPO-independent endogenous erythroid colony formation from PV samples (IC50=67 nM). These results suggested that ruxolitinib inhibited hematopoietic cell proliferation, with greater activity against JAK2-mutated cells versus normal cells23.
In a mouse model of MPN, Ba/F3-EPOR-JAK2V617F cells were injected into the tail vein of Balb/c mice, and mice were randomized to treatment with vehicle or ruxolitinib23. Vehicle-treated mice developed massive splenomegaly and became moribund in 2–3 weeks. In contrast, most mice treated with ruxolitinib survived (90% survival at 22 days, versus 10% in the control group)23. At 15 days post-inoculation, vehicle-treated mice displayed splenomegaly (mean splenic weight 471 mg), while the spleen of mice treated with ruxolitinib were more similar to normal controls (mean weight 110 mg). Treatment with ruxolitinib also led to a marked reduction in the plasma levels of IL-6 and tumor necrosis factor alpha (TNF-α) compared to vehicle-treated mice, similarly to what has been observed in humans23.
2.3 Pharmacokinetics and metabolism
In healthy human subjects receiving a single dose of 25 mg of [14C]ruxolitinib in oral solution, results of pharmacokinetics analysis showed rapid absorption (mean time to peak concentrations 0.63h), with a mean peak concentration (Cmax) of 1093 nM and AUC0-∞ of 3200 nM.h24. Ruxolitinib concentration declined in a monophasic or biphasic fashion (t1/2 of 2.32h and 5.81h, respectively)24. The excretion of ruxolitinib-derived radioactivity was rapid, with 96% of administered radioactivity being recovered within 24 hours (74% in urine and 22% in feces)24. Metabolic profiling in urine and feces demonstrated hydroxyl and oxo metabolites as well as glucorinide conjugates of ruxolitinib, with the parent compound accounting for <1% of the excreted dose, suggesting that after an oral dose >95% of ruxolitinib is absorbed24. There were no major differences in concentrations of the parent compound or metabolites after single or multiple dosing, suggesting a lack of drug accumulation. In another pharmacokinetic study, healthy human subjects received either single, ascending doses of ruxolitinib (5 to 200 mg) or multiple ascending doses both once and twice daily25. Results of this study showed a good oral bioavailability of ruxolitinib and dose-proportional systemic exposures25. A high-fat meal reduced Cmax by 24%, but had no effect on ruxolitinib AUC. The maximum tolerated doses (MTD) in healthy volunteers were determined to be 25 mg twice daily and 100 mg once daily25.
3 Clinical studies
3.1 Phase I/II Study
Ruxolitinib was first evaluated in a phase I/II study (NCT00509899) that recruited 153 patients with MF18. Doses of ruxolitinib ranged from 10–50 mg twice daily to 25–200 mg once daily. The MTD was determined to be 25 mg twice daily, and the dose limiting toxicity (DLT) was thrombocytopenia18. However, an individualized dose schedule whereupon patients started with 15 mg twice daily and dose was escalated monthly in case there was no response and no significant toxicity proved to be the best strategy. Ruxolitinib led to significant improvements in spleen size and constitutional symptoms. By the International Working Group on Myelofibrosis Research and Treatment (IWG-MRT) response criteria, a clinical improvement (CI) in splenomegaly (≥50% in spleen reduction by palpation) was seen in 44% of patients across all dose levels18. Interestingly, responses were observed in both JAK2V6171F-positive (51%) and –negative (45%) patients, further evidence that deregulated JAK signaling is important to the pathogenesis of both mutated and wild type JAK218. There were significant improvements in the symptomatic burden of patients, including reductions in pruritus, night sweats, weight loss, fatigue, bone and abdominal pain, and patients had an improvement in their exercise ability18. A decrease in pro-inflammatory cytokines mirrored the improvement in systemic symptoms18.
Long term follow-up data for the cohort of patients participating in the aforementioned phase I/II trial who were treated at the M.D. Anderson Cancer Center has been published26. After a median follow-up of 2 years, the median overall survival (OS) of 107 patients with MF who received ruxolitinib was 69%26. A CI in splenomegaly by the IWG-MRT was reached at some point in 61 patients (57%)26. Responses were durable, as median duration of response was estimated around 3.2 years26. MF-related symptoms were assessed by serial completion of the Myelofibrosis Symptom Assessment Form (MFSAF). The total symptom score (TSS; composite score of night sweats, itching, abdominal pain/discomfort, bone pain) decreased a median of 60% during 2 years of therapy. Overall, 58 (54%) patients were still being treated with ruxolitinib at the time of report. The 2-years OS for these patients was 92% (International Prognostic Scoring System [IPSS] Intermediate-2) and 88% (IPSS High)26. A total of 49 patients (46%) discontinued therapy with ruxolitinib. The 2-years OS for these patients was 32% (IPSS Intermediate-2) and 21% (IPSS high 21%). Most frequent reasons for therapy discontinuation were death (12%) and progressive disease (11%). Patients who had a ≥50% reduction in spleen size had an improved survival compared to patients who had lesser degrees of spleen reduction (HR=0.223; 95% CI 0.09–0.51; p=0.0001)26.
The survival of the 107 patients treated with ruxolitinib at M.D. Anderson Cancer Center was compared to the outcomes of 310 historical matched controls that would have fulfilled enrollment criteria for the phase I/II trial26. Therapy with ruxolitinib significantly improved survival compared to the historical control group (HR=0.58; 95% CI 0.39–0.85; p=0.005)26. The benefit was more evident in patients in the high IPSS risk group (HR=0.50; 95% CI 0.31–0.81; p=0.006). The OS at 2 years for high-risk IPSS was 83% in the ruxolitinib-treated patients versus 58% in the historical control group.26 The Mayo Clinic also published a report on the long-term outcome of 51 patients with MF treated with ruxolitinib in that center during the phase I/II study27. Contrary to the study based on the larger M.D. Anderson cohort, no improvement in OS was detected when their outcome was compared to that of 410 historical control patients with MF (all IPSS risk categories) treated at the same center (unadjusted p=0.43; p=0.58 after adjusting for the Dynamic International Prognostic Scoring System-Plus Score [DIPSS-Plus])27. This difference in results obtained may have stemmed from the very high treatment discontinuation rate in the Mayo cohort (1 year-51%; 3 years-89%)27, which were higher than the discontinuation rates at M.D. Anderson Cancer Center (1 year- 24%; 3 years-46%)26. The most common reasons for discontinuation in the Mayo cohort were patient withdrawal of consent (29.4%), physician decision (23.5%) and disease progression (19.6%)27. Additionally, the mean dose of ruxolitinib over time was much lower in the Mayo cohort compared to the M.D. Anderson cohort26.
Initial reports from the phase I/II study did not show any appreciable reduction in JAK2V617F allele burden or decrease in BM fibrosis18. However, longer term follow-up has demonstrated that this may not always be true. A recent report compared the reduction in the grade of BM fibrosis among patients receiving BAT (best available therapy; historical control) and ruxolitinib28. After 2 years (74% [ruxolitinib] vs. 60% [BAT]; odds ratio [OR] 2.62), 4 years (79% vs. 46%; OR 9.40) and 5 years (76% vs. 32%; OR 15.39) of therapy, more patients in the ruxolitinib-treated group had an improvement or stabilization in the grade of BM fibrosis compared to patients in the BAT group. Conversely, at 4 years (25% vs. 76%) and 5 years (26% vs. 75%), more patients in the BAT group had worsening of BM fibrosis. There was a strong correlation between reduction in splenomegaly and BM fibrosis reduction at 4 years in patients receiving ruxolitinib. One case report also described a patient with post-PV MF who participated in the COMFORT-II clinical trial (see below) and had complete resolution of BM fibrosis after 168 weeks of therapy with ruxolitinib29. This patient also had an improvement in hematopoiesis and a dramatic reduction in JAK2V671F allele burden. Both studies suggest that in selected cases, long-term therapy with a JAK2 inhibitor may reverse BM fibrosis.
3.2 Phase III Studies
There were two phase III clinical trials conducted with ruxolitinib in patients with MF. In the phase III COMFORT-I clinical trial (that was conducted in North America; NCT00952289), a total of 309 patients with intermediate-2 and high-risk IPSS MF were randomized (1:1) between ruxolitinib (15–20 mg twice daily, depending on the platelet count) and placebo19. Spleen response rate (RR) at 24 weeks was the primary endpoint, and was defined as a ≥35% reduction in spleen volume by magnetic resonance imaging (MRI), that corresponded to a decrease ≥50% of spleen size by palpation. After a median follow-up of 32 weeks, the RR was 41.9% in ruxolitinib-treated patients vs. 0.7% in placebo-treated patients (p<0.0001)19. Patients receiving ruxolitinib had a mean reduction in spleen volume of 31.6% at 24 weeks, compared to a mean increase of 8.1% in spleen volume at 24 weeks in placebo-treated patients19. There was a ≥50% reduction in the MFSAF-TSS in 46% of ruxolitinib-treated patients versus 5.3% of placebo-treated patients (p<0.0001)19. Most patients who were treated with ruxolitinib had an improvement in symptoms, whereas most patients treated with placebo had worsening of symptoms. No subgroup of patients (subtype of MF [primary vs. secondary], age, JAK2 mutational status, spleen size, hemoglobin, IPSS risk score) was identified that had a differential response to ruxolitinib19. After a median follow-up of 51 weeks, therapy with ruxolitinib led to an improvement in overall survival of patients with Intermediate-2/High risk MF (HR 0.5; p=0.04 versus placebo)19.
In the 3-years update of the COMFORT-I trial, after a median follow-up of 2.86 years, 77 of 155 patients randomized to ruxolitinib were still receiving therapy and 111 of 154 patients randomized to placebo were crossed-over to ruxolitinib30. The mean spleen volume reduction after 144 weeks was 34.1%30. Among all patients randomized to ruxolitinib, 59% achieved a ≥35% reduction in spleen volume at any point during study; the probability of maintaining this response after 132 weeks was 53%30. Patients who crossed over from placebo to ruxolitinib also had significant improvements in spleen volume (mean 30.0% reduction). Improvements in symptoms and quality of life were maintained in the majority of patients after 2 years of therapy31. After 3 years, a survival benefit with ruxolitinib was still present (HR=0.69, 95% CI 0.46–1.03, p=0.067), despite the fact that by 80 weeks all patients in the placebo group had discontinued or crossed over to ruxolitinib30.
In the companion trial COMFORT-II (conducted in Europe; NCT00934544), 219 patients with Intermediate-2/High risk MF were randomized (2:1) among ruxolitinib and best available therapy (BAT)20. The primary endpoint was the spleen RR at 48 weeks as assessed by a MRI. The most commonly used agent in the BAT arm was hydroxyurea (47%). At 48 weeks, the majority of patients in the ruxolitinib arm had a reduction in spleen volume, and 28% achieved a ≥35% reduction in spleen volume20. Median time to response was 12.3 weeks. In the BAT arm, 0% of patients achieved a spleen volume response (p<0.0001)20. Mean relative change in spleen volume at 48 weeks was −30.1% (ruxolitinib) and +7.3% (BAT; p<0.001)20. Ruxolitinib also led to improvements in patient-reported quality of life measurements (assessed by the EORTC QLQ-C30 and FACT-Lym subscales) compared to patients receiving BAT, who consistently worsened during study period. Correlative studies showed that levels of proinflammatory cytokines (IL-6, TNF-α, C-reactive protein) decreased in patients receiving ruxolitinib and there was an increase in erythropoietin and leptin20.
A 3-years update of COMFORT-II was published recently32. After a median follow-up of 151 weeks, 97% of patients receiving ruxolitinib had some degree of spleen reduction, and 51% achieved a response (≥35% reduction) in spleen volume at some point32. The probability of maintaining this response at 144 weeks was 50%. Among patients with MF who were treated with ruxolitinib and were positive for the JAK2V617F mutation, a ≥20% reduction in JAK2V617F allele burden at 48 weeks was seen in 22% of patients32. This was associated with a higher spleen volume RR (73% vs. 19%; odds ratio [OR] 4.6). The probability of survival at 144 weeks was 81% in the ruxolitinib arm and 61% in the BAT arm (HR=0.48; p=0.009), consistent with a survival benefit for ruxolitinib, similarly to the results seen with COMFORT-I32.
One study reported on a pooled survival analysis of both COMFORT studies, and the outcomes of 301 patients with MF who received therapy with ruxolitinib were compared to 227 patients who received control therapy33. Results of this analysis demonstrated that therapy with ruxolitinib reduced the risk of death (HR =0.65; 95% CI 0.46–0.90)33. In another post-hoc analysis, the survival of MF patients who were treated with ruxolitinib in the COMFORT-II study was compared to the survival of a matched cohort of patients in the dynamic IPSS (DIPSS) database34. Survival was measured from the date of diagnosis, not from the date of start of ruxolitinib therapy. Results from this study demonstrated that patients who received ruxolitinib at some point during the course of their disease had improved survival (median OS 5 years vs. 3.5 years; HR 0.61; 95% CI 0.41–0.91; p=0.0148)34. Another report analyzed the impact of high-risk mutations (i.e. mutations in ASXL1, EZH2, IDH1/2, SRSF2) on RR to ruxolitinib in patients treated in the COMFORT-II trial35. There was no difference in the RR in patients in the high-risk molecular group, and there was a trend for a reduced risk of death in patients with high risk mutations treated with ruxolitinib compared to BAT (HR=0.57; 95% CI 0.30–1.80)35.
Recently, two papers reported on novel calreticulin gene (CALR) mutations that occur in patients with Ph-negative MF and ET who are negative for both JAK2 and MPL mutations16,36. These mutations are found in 67% of JAK2/MPL-negative ET patients and up to 88% of JAK2/MPL-negative MF patients36. CALR mutations are always frameshift insertions and deletions that occur in exon 9, and the most common mutations are a 52-bp deletion (c.1092_1043del; type 1 mutation) and a 5-bp insertion (c.1154_1155insTTGTC).36 Mutated calreticulin appears to induce STAT5 activation through an unknown mechanism36, and immunohistochemistry directed against mutated calreticulin have revealed that the protein is mostly expressed in megakaryocytes, with a faint staining in erythroid and granulocytic cells37. Patients with CALR mutations have higher platelet counts, decreased thrombotic risk and improved survival compared to patients harboring either JAK or MPL mutations and triple-negative (JAK/MPL/CALR-negative) patients36,38. One paper reported recently on two patients with CALR-mutated MF who were treated with the JAK2 inhibitor fedratinib39. Both patients had a clinical response, as evidenced by a decrease in spleen size39. This report suggests that ruxolitinib and other JAK2 inhibitors may be effective in patients harboring CALR mutation, and may explain the observed fact that the JAK2V617F mutational status did not predict response to ruxolitinib in the aforementioned clinical trials. However, there is still need for a formal evaluation of the clinical response to ruxolitinib in patients with CALR-mutated MF.
The results of clinical trials with ruxolitinib are summarized in table 1.
Table 1.
Summary of clinical studies conducted with ruxolitinib
| Study | Number of patients | Phase | Therapy | Spleen Response Rate* | Other response endpoints |
|---|---|---|---|---|---|
| Verstovsek et al.18 | 153 | I/II | Ruxolitinib (all patients) | 44% | Reduction in pruritus, night sweats, weight loss, fatigue, bone pain, abdominal pain; improvement in exercise ability |
| Verstovsek et al.19 (COMFORT-I) | 309 | III | Ruxolitinib (155 patients) vs. placebo (154 patients) | 41.9% (ruxolitinib) vs. 0.7% (placebo)** | Symptom improvement: 46% (ruxolitinib) vs. 5.3% (placebo)** Overall Survival: HR 0.50 (vs. placebo)† |
| Harrison et al.20 (COMFORT-II) | 219 | III | Ruxolitinib (146 patients) vs. best available therapy (73 patients) | 28% (ruxolitinib) vs. 0% (best available therapy)** | Symptom and quality of life improvement with ruxolitinib Overall survival: HR 0.48 (vs. best available therapy)‡ |
-spleen response was defined as ≥50% reduction in spleen size by palpation in the phase I/II trial; spleen response was defined as ≥35% reduction in spleen volume by magnetic resonance imaging at 24 weeks (COMFORT-I) and 48 weeks (COMFORT-II).
- p<0.0001.
- p=0.04.
- p=0.009
3.4 Safety and tolerability
The most common side effects observed with ruxolitinib are dose-dependent anemia and thrombocytopenia (table 2). Data from the COMFORT-I trial demonstrate that ruxolitinib leads to grade 3–4 anemia in 45.2% of patients compared to 19.2% of placebo-treated patients (34% increase) and causes grade 3–4 thrombocytopenia in 12.9% of patients compared to 1.3% in patients treated with placebo (11.6% increase) when these rates of hematological side effects were assessed at any time during the study (initial follow-up period of 51 weeks)19. Grade3–4 neutropenia is uncommon (7.1% in COMFORT-I). Anemia and thrombocytopenia are more common in the early phases of therapy, and tend to improve with time19. The most common pattern is for patients to present with a fall of 1.5 g/dL in hemoglobin during the first 8–12 weeks of therapy, followed by an increase to a new steady state with 24 weeks of therapy19. In COMFORT-I, the majority of novel cases of grade 3–4 anemia/thrombocytopenia occurred within the first 6 months of therapy, and after 12 months the incidence of new cases was lower (i.e. 12% [anemia] and 2.8% [thrombocytopenia])30. Despite the relatively high incidence of cytopenias in the first 12 months of therapy, few patients (only one in each phase III trial) had to discontinue therapy due to cytopenias. Ruxolitinib dose adjustments and transient red blood cell transfusions can be implemented in case of drug-induced cytopenias. Data from COMFORT-I suggests that patients who were dose-reduced to 10 mg twice daily after nadir hemoglobin had faster recovery to pre-treatment levels, and had similar benefits regarding spleen volume reduction, symptom control and survival30,31.
Table 2.
Incidence of anemia and thrombocytopenia in clinical studies with ruxolitinib
| Study | Therapy | Anemia Grade 1–2 |
Anemia Grade 3–4 | Thrombocytopenia Grade 1–2 |
Thrombocytopenia Grade 3–4 |
|---|---|---|---|---|---|
| Verstovsek et al.18 | Ruxolitinib | NR | 23% | NR | 20% |
| Verstovsek et al.19 (COMFORT-I) | Ruxolitinib | 51% | 45% | 57% | 13% |
| Verstovsek et al.19 (COMFORT-I) | Placebo | 68% | 19% | 29% | 1.5% |
| Harrison et al.20 (COMFORT-II) | Ruxolitinib | 54% | 42% | 60% | 8% |
| Harrison et al.20 (COMFORT-II) | Best available therapy | 63% | 31% | 22% | 7% |
NR means not reported;
Non-hematological side effects are usually grade 1–2 in severity, and rarely lead to treatment discontinuation (8% and 11% in both phase III trials, discontinuation rate similar to that seen in control arms)19,20. Most common non-hematological side effects compared to placebo were included ecchymosis (18.7% vs. 9.3%), headache (14.8% vs. 5.3%) and dizziness (14.8% vs. 6.6%)19.
Recently, there were several cases reports of systemic, opportunistic infections in patients being treated with ruxolitinib, including hepatitis B virus reactivation40, herpes simplex reactivation41, Cryptococcus neoformans pneumonia42, toxoplasmosis retinitis43, tuberculosis44 and progressive multifocal leukoencephalopathy (the latter being only a single case)45. There is some in vitro evidence to suggest that ruxolitinib impairs dendritic cell functions, which may result in impaired CD4+ and CD8+ T-lymphocyte activation46. On the other hand, one study reported on a single patient with MF who had leukopenia and recurrent infections and had an improvement in white blood cell count and reduced risk of infections after receiving therapy with low-dose ruxolitinib47. While there is no conclusive data on the immunosuppressive potential of ruxolitinib, the physician should be mindful of possible atypical infectious complications in patients being treated with this drug48.
After discontinuing therapy, patients experience a rapid relapse of symptoms after a median of 7 days post drug discontinuation19. One study reported on the development of a cytokine rebound “syndrome”, similar to an inflammatory shock state, after ruxolitinib abrupt discontinuation49. In the COMFORT-I trial, however, there was no increased rate of severe adverse events after treatment discontinuation in patients receiving ruxolitinib versus patients receiving placebo (6.1% vs. 5.6%, respectively)19. Despite this, the prescribing information for ruxolitinib in the United States recommends dose tapering (5 mg BIDper week) to decrease the risk of withdrawal return of symptoms.
4 Regulatory Affairs
Ruxolitinib was discovered and developed within Incyte. A new drug application (NDA) was submitted to the U.S. Food and Drug Administration (FDA) in June 8, 2011. The drug was approved by the FDA for treatment of patients with intermediate and high-risk MF in November 16, 2011. Ruxolitinib was also approved in the European Union in August 29, 2012 for therapy of disease-related splenomegaly or symptoms in adults with PMF, post-PV MF and post-ET MF.
5 Conclusion
The approval of ruxolitinib represented a milestone in the history of therapy fof MF, since it is the first drug that was ever approved for the treatment of this disease. Ruxolitinib is an effective drug for treating splenomegaly and disease-related symptoms in patients with MF. Its effects on splenomegaly are durable, and it improves patients’ quality of life and functioning (patients walk more and gain weight). There is now strong data from both pivotal phase III trials demonstrating that therapy with ruxolitinib improves survival of patients with MF. Furthermore, the drug is active in all subsets of MF, including patients who do not harbor the JAK2V617F mutation. The main toxicity of ruxolitinib is drug-related cytopenias, which can be treated with dose adjustments and rarely lead to treatment interruption.
6 Expert Opinion
While it is clear that ruxolitinib can improve splenomegaly, MF-associated signs and symptoms, as well as survival, there is more that needs to be learned. There are several areas of research that we believe of importance to further consolidate the role, and improve the use of ruxolitinib as therapy for patients with MF.
It is important to determine biomarkers of response to ruxolitinib to best select patients for this therapy. It is already known that the JAK2V617F mutational status does not predict for response to ruxolitinib, as JAK2V617F negative patients have the same response as those with mutation. The concomitant presence of additional epigenetic and chromatin-modifying mutations, such as has been reported recently for therapy with pegylated interferon-α2a in PV and ET50, should be explored. It is also clear that some patients present with significant responses to ruxolitinib, manifested by major decreases in spleen size, JAK2V617F allele burden and BM fibrosis reduction. If we could unravel the molecular mechanisms behind those patients with outstanding response this could help us to better understand the mechanism of action of ruxolitinib in MF and possibly identify biomarkers of response and duration of response.
Additionally, the use of ruxolitinib should be explored in patients with lower-risk disease. Patients with low risk MF have relatively preserved blood cell count, no significant constitutional symptoms, and very rarely symptomatic splenomegaly. The use of ruxolitinib in these patients, possibly at lower doses to decrease the incidence of drug-related cytopenias, would mark a shift from treating patients with established advanced, symptomatic disease, to treating patients to prevent the development of signs and symptoms.
Allogeneic stem cell transplantation is the only known curative treatment for patients with MF. Improvements in conditioning regimen-related toxicities and HLA typing have made transplantation a possible therapeutic option for a greater percentage of patients than in the past.8 While clinical trials with ruxolitinib have focused solely on patients deemed ineligible for transplantation, the use of ruxolitinib as a pre-transplant therapy in transplant eligible patients should be explored. Ruxolitinib therapy may decrease spleen size, which may possibly improve engraftment time and graft function after transplant. Patients may improve performance status and gain weight, and be in better physical condition for the procedure. Preliminary experience of such use of ruxolitinib have already been reported.51,52 A question that is important is whether responding patients should be transplanted at the time of optimal response or when they start to lose response to the drug. Well conducted clinical trials will help us better define the role of ruxolitinib in transplant-eligible patients with MF.
While ruxolitinib represents an important advance for therapy of MF, it is clear that it is not a curative option, and over time patients will lose response to the drug. Loss of response may present as worsening splenomegaly, worsening symptoms, and/or worsening cytopenias, after prolonged stable response to therapy. Little is known about this process. There is in vitro data suggesting that resistance to ruxolitinib is related to stabilization and increase of JAK2 protein levels followed by heterodimerization with JAK1 and TYK2 that leads to JAK2 transactivation and resumption of signaling53. Stopping exposure of cells to ruxolitinib leads to a regain of sensitivity. While these results are intriguing, this strategy still needs to be confirmed in the clinical setting, and will be explored in an upcoming clinical trial (ReTreatment trial; NCT02091752)53. Combinatorial trials evaluating ruxolitinib with additional agents known to be active in this disease, or shown active in preclinical models, are of utmost importance. It is known that inhibition of Heat Shock Protein 90 (Hsp90) leads to JAK2 degradation, and compounds such as Hsp90 Inhibitors54 and histone deacetylase 6 inhibitors55 have pre-clinical activity in animal models of JAK2-mediated MPN. Clinical trials of these compounds in combination with ruxolitinib are already underway (PRIME trial; NCT01693601), and hopefully will add other therapeutic options for the treatment of patients with MF.
Drug Summary Box.

Footnotes
Conflicts of interest: FPSS has received research funding from Novartis; SV has received research support from Incyte
Declaration of interest:
The authors have received research funding from Novartis and Incyte
References
- 1.Thiele J, Kvaniscka H, Tefferi A, et al. Primary Myelofibrosis. In: Swerdlow S, Campo E, Harris N, et al., editors. WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. 4th. Lyon: IARC Press; 2008. pp. 44–47. [Google Scholar]
- 2.Mesa RA, Silverstein MN, Jacobsen SJ, et al. Population-based incidence and survival figures in essential thrombocythemia and agnogenic myeloid metaplasia: an Olmsted County Study, 1976–1995. Am J Hematol. 1999;61:10–5. doi: 10.1002/(sici)1096-8652(199905)61:1<10::aid-ajh3>3.0.co;2-i. [DOI] [PubMed] [Google Scholar]
- 3.Mesa RA, Shields A, Hare T, et al. Progressive burden of myelofibrosis in untreated patients: assessment of patient-reported outcomes in patients randomized to placebo in the COMFORT-I study. Leuk Res. 2013;37:911–6. doi: 10.1016/j.leukres.2013.04.017. [DOI] [PubMed] [Google Scholar]
- 4.Mesa RA, Kantarjian H, Tefferi A, et al. Evaluating the serial use of the Myelofibrosis Symptom Assessment Form for measuring symptomatic improvement: performance in 87 myelofibrosis patients on a JAK1 and JAK2 inhibitor (INCB018424) clinical trial. Cancer. 2011;117:4869–77. doi: 10.1002/cncr.26129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Tefferi A, Pardanani A, Lim KH, et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia. 2009;23:905–11. doi: 10.1038/leu.2009.47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Quintas-Cardama A, Kantarjian HM, Manshouri T, et al. Lenalidomide plus prednisone results in durable clinical, histopathologic, and molecular responses in patients with myelofibrosis. J Clin Oncol. 2009;27:4760–6. doi: 10.1200/JCO.2009.22.6548. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Cervantes F, Dupriez B, Pereira A, et al. New prognostic scoring system for primary myelofibrosis based on a study of the International Working Group for Myelofibrosis Research and Treatment. Blood. 2009;113:2895–901. doi: 10.1182/blood-2008-07-170449. [DOI] [PubMed] [Google Scholar]
- 8.Kroger N, Holler E, Kobbe G, et al. Allogeneic stem cell transplantation after reduced-intensity conditioning in patients with myelofibrosis: a prospective, multicenter study of the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Blood. 2009;114:5264–70. doi: 10.1182/blood-2009-07-234880. [DOI] [PubMed] [Google Scholar]
- 9.Baxter EJ, Scott LM, Campbell PJ, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet. 2005;365:1054–61. doi: 10.1016/S0140-6736(05)71142-9. [DOI] [PubMed] [Google Scholar]
- 10.James C, Ugo V, Le Couedic JP, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature. 2005;434:1144–8. doi: 10.1038/nature03546. [DOI] [PubMed] [Google Scholar]
- 11.Kralovics R, Passamonti F, Buser AS, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med. 2005;352:1779–90. doi: 10.1056/NEJMoa051113. [DOI] [PubMed] [Google Scholar]
- 12.Levine RL, Wadleigh M, Cools J, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell. 2005;7:387–97. doi: 10.1016/j.ccr.2005.03.023. [DOI] [PubMed] [Google Scholar]
- 13.Leonard WJ, O’Shea JJ. Jaks and STATs: biological implications. Annu Rev Immunol. 1998;16:293–322. doi: 10.1146/annurev.immunol.16.1.293. [DOI] [PubMed] [Google Scholar]
- 14.Parganas E, Wang D, Stravopodis D, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell. 1998;93:385–95. doi: 10.1016/s0092-8674(00)81167-8. [DOI] [PubMed] [Google Scholar]
- 15.Neubauer H, Cumano A, Muller M, et al. Jak2 deficiency defines an essential developmental checkpoint in definitive hematopoiesis. Cell. 1998;93:397–409. doi: 10.1016/s0092-8674(00)81168-x. [DOI] [PubMed] [Google Scholar]
- 16.Nangalia J, Massie CE, Baxter EJ, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med. 2013;369:2391–405. doi: 10.1056/NEJMoa1312542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Pikman Y, Lee BH, Mercher T, et al. MPLW515L is a novel somatic activating mutation in myelofibrosis with myeloid metaplasia. PLoS Med. 2006;3:e270. doi: 10.1371/journal.pmed.0030270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Verstovsek S, Kantarjian H, Mesa RA, et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med. 2010;363:1117–27. doi: 10.1056/NEJMoa1002028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med. 2012;366:799–807. doi: 10.1056/NEJMoa1110557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Harrison C, Kiladjian JJ, Al-Ali HK, et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med. 2012;366:787–98. doi: 10.1056/NEJMoa1110556. [DOI] [PubMed] [Google Scholar]
- 21.Incyte: Ruxolitinib Prescribing Information. 2013 [Google Scholar]
- 22.Campas-Moya C. Ruxolitinib. Drugs Future. 2010;35:457–465. [Google Scholar]
- 23.Quintas-Cardama A, Vaddi K, Liu P, et al. Preclinical characterization of the selective JAK1/2 inhibitor INCB018424: therapeutic implications for the treatment of myeloproliferative neoplasms. Blood. 2010;115:3109–17. doi: 10.1182/blood-2009-04-214957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Shilling AD, Nedza FM, Emm T, et al. Metabolism, excretion, and pharmacokinetics of [14C]INCB018424, a selective Janus tyrosine kinase 1/2 inhibitor, in humans. Drug Metab Dispos. 2010;38:2023–31. doi: 10.1124/dmd.110.033787. [DOI] [PubMed] [Google Scholar]
- 25.Shi JG, Chen X, McGee RF, et al. The pharmacokinetics, pharmacodynamics, and safety of orally dosed INCB018424 phosphate in healthy volunteers. J Clin Pharmacol. 2011;51:1644–54. doi: 10.1177/0091270010389469. [DOI] [PubMed] [Google Scholar]
- 26.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;120:1202–9. doi: 10.1182/blood-2012-02-414631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Tefferi A, Litzow MR, Pardanani A. Long-term outcome of treatment with ruxolitinib in myelofibrosis. N Engl J Med. 2011;365:1455–7. doi: 10.1056/NEJMc1109555. [DOI] [PubMed] [Google Scholar]
- 28.Kvaniscka H, Thiele J, Bueso-Ramos CE, et al. Effects Of Five-Years Of Ruxolitinib Therapy On Bone Marrow Morphology In Patients With Myelofibrosis and Comparison With Best Available Therapy [abstract] Blood. 2013;122 Abstract 4055. [Google Scholar]
- 29.Wilkins BS, Radia D, Woodley C, et al. Resolution of bone marrow fibrosis in a patient receiving JAK1/JAK2 inhibitor treatment with ruxolitinib. Haematologica. 2013;98:1872–6. doi: 10.3324/haematol.2013.095109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Verstovsek S, Gotlib J, Levy RS, et al. Long-Term Outcomes Of Ruxolitinib Therapy In Patients With Myelofibrosis: 3-Year Update From COMFORT-I [abstract] Blood. 2013;122 Abstract 396. [Google Scholar]
- 31.Verstovsek S, Mesa RA, Gotlib J, et al. Efficacy, safety and survival with ruxolitinib in patients with myelofibrosis: results of a median 2-year follow-up of COMFORT-I. Haematologica. 2013;98:1865–71. doi: 10.3324/haematol.2013.092155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cervantes F, Vannucchi AM, Kiladjian JJ, et al. Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood. 2013;122:4047–53. doi: 10.1182/blood-2013-02-485888. [DOI] [PubMed] [Google Scholar]
- 33.Vannucchi A, Kantarjian H, Kiladjian J-J, et al. A Pooled Overall Survival Analysis of The COMFORT Studies: 2 Randomized Phase 3 Trials of Ruxolitinib For The Treatment of Myelofibrosis [abstract] Blood. 2013;122 Abstract 2820. [Google Scholar]
- 34.Passamonti F, Maffioli M, Cervantes F, et al. Impact of ruxolitinib on the natural history of primary myelofibrosis: a comparison of the DIPSS and the COMFORT-2 cohorts. Blood. 2014 doi: 10.1182/blood-2013-12-544411. [DOI] [PubMed] [Google Scholar]
- 35.Guglielmelli P, Biamonte F, Rotunno G, et al. Impact of mutational status on outcomes in myelofibrosis patients treated with ruxolitinib in the COMFORT-II Study. Blood. 2014 doi: 10.1182/blood-2013-11-536557. [DOI] [PubMed] [Google Scholar]
- 36.Klampfl T, Gisslinger H, Harutyunyan AS, et al. Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med. 2013;369:2379–90. doi: 10.1056/NEJMoa1311347. [DOI] [PubMed] [Google Scholar]
- 37.Vannucchi AM, Rotunno G, Bartalucci N, et al. Calreticulin mutation-specific immunostaining in myeloproliferative neoplasms: pathogenetic insight and diagnostic value. Leukemia. 2014 doi: 10.1038/leu.2014.100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.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 doi: 10.1038/leu.2014.3. [DOI] [PubMed] [Google Scholar]
- 39.Passamonti F, Caramazza D, Maffioli M. JAK inhibitor in CALR-mutant myelofibrosis. N Engl J Med. 2014;370:1168–9. doi: 10.1056/NEJMc1400499. [DOI] [PubMed] [Google Scholar]
- 40.Caocci G, Murgia F, Podda L, et al. Reactivation of hepatitis B virus infection following ruxolitinib treatment in a patient with myelofibrosis. Leukemia. 2014;28:225–7. doi: 10.1038/leu.2013.235. [DOI] [PubMed] [Google Scholar]
- 41.Tong LX, Jackson J, Kerstetter J, et al. Reactivation of herpes simplex virus infection in a patient undergoing ruxolitinib treatment. J Am Acad Dermatol. 2014;70:e59–60. doi: 10.1016/j.jaad.2013.09.035. [DOI] [PubMed] [Google Scholar]
- 42.Wysham NG, Sullivan DR, Allada G. An opportunistic infection associated with ruxolitinib, a novel janus kinase 1,2 inhibitor. Chest. 2013;143:1478–9. doi: 10.1378/chest.12-1604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Goldberg RA, Reichel E, Oshry LJ. Bilateral toxoplasmosis retinitis associated with ruxolitinib. N Engl J Med. 2013;369:681–3. doi: 10.1056/NEJMc1302895. [DOI] [PubMed] [Google Scholar]
- 44.Colomba C, Rubino R, Siracusa L, et al. Disseminated tuberculosis in a patient treated with a JAK2 selective inhibitor: a case report. BMC Res Notes. 2012;5:552. doi: 10.1186/1756-0500-5-552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wathes R, Moule S, Milojkovic D. Progressive multifocal leukoencephalopathy associated with ruxolitinib. N Engl J Med. 2013;369:197–8. doi: 10.1056/NEJMc1302135. [DOI] [PubMed] [Google Scholar]
- 46.Heine A, Held SA, Daecke SN, et al. The JAK-inhibitor ruxolitinib impairs dendritic cell function in vitro and in vivo. Blood. 2013;122:1192–202. doi: 10.1182/blood-2013-03-484642. [DOI] [PubMed] [Google Scholar]
- 47.Islam R, Yellu MR, Rafiullah, et al. Low-dose ruxolitinib for improving leukopaenia and reducing recurrent infections associated with myelofibrosis. BMJ Case Rep. 2013;2013 doi: 10.1136/bcr-2013-010068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Heine A, Brossart P, Wolf D. Ruxolitinib is a potent immunosuppressive compound: is it time for anti-infective prophylaxis? Blood. 2013;122:3843–4. doi: 10.1182/blood-2013-10-531103. [DOI] [PubMed] [Google Scholar]
- 49.Tefferi A, Pardanani A. Serious adverse events during ruxolitinib treatment discontinuation in patients with myelofibrosis. Mayo Clin Proc. 2011;86:1188–91. doi: 10.4065/mcp.2011.0518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Quintas-Cardama A, Abdel-Wahab O, Manshouri T, et al. Molecular analysis of patients with polycythemia vera or essential thrombocythemia receiving pegylated interferon alpha-2a. Blood. 2013;122:893–901. doi: 10.1182/blood-2012-07-442012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Stubig T, Alchalby H, Ditschkowski M, et al. JAK inhibition with ruxolitinib as pretreatment for allogeneic stem cell transplantation in primary or post-ET/PV myelofibrosis. Leukemia. 2014 doi: 10.1038/leu.2014.86. [DOI] [PubMed] [Google Scholar]
- 52.Jaekel N, Behre G, Behning A, et al. Allogeneic hematopoietic cell transplantation for myelofibrosis in patients pretreated with the JAK1 and JAK2 inhibitor ruxolitinib. Bone Marrow Transplant. 2014;49:179–84. doi: 10.1038/bmt.2013.173. [DOI] [PubMed] [Google Scholar]
- 53.Koppikar P, Bhagwat N, Kilpivaara O, et al. Heterodimeric JAK-STAT activation as a mechanism of persistence to JAK2 inhibitor therapy. Nature. 2012;489:155–9. doi: 10.1038/nature11303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Bhagwat N, Koppikar P, Keller M, et al. Improved targeting of JAK2 leads to increased therapeutic efficacy in myeloproliferative neoplasms. Blood. 2014 doi: 10.1182/blood-2014-01-547760. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.DeAngelo DJ, Mesa RA, Fiskus W, 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. Br J Haematol. 2013;162:326–35. doi: 10.1111/bjh.12384. [DOI] [PubMed] [Google Scholar]
