Accepted Manuscript Title: Are we altering the natural history of primary myelofibrosis? Author: Michael R. Savona PII: DOI: Reference:

S0145-2126(14)00123-4 http://dx.doi.org/doi:10.1016/j.leukres.2014.04.012 LR 5162

To appear in:

Leukemia Research

Received date: Revised date: Accepted date:

10-3-2014 14-4-2014 24-4-2014

Please cite this article as: Savona MR, Are we altering the natural history of primary myelofibrosis?, Leukemia Research (2014), http://dx.doi.org/10.1016/j.leukres.2014.04.012 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Are we altering the natural history of primary myelofibrosis?

Michael R. Savona, MD, FACP*

ip t

Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA

us

cr



*Corresponding author: Division of Hematology/Oncology, Vanderbilt University Medical

an

Center, 2220 Pierce Avenue, 777 PRB, Nashville, TN 37232, USA. Tel: +1 615-3434677; Fax: +1 615-343-7602.

Ac ce p

te

d

M

E-mail address: [email protected] (M.R. Savona)

1 Page 1 of 40

ABSTRACT Primary myelofibrosis (PMF) is a clonal hematologic malignancy with a variable disease

ip t

course; survival ranges from months to years. Historically, only allogeneic

hematopoietic stem cell transplantation (alloHSCT) has demonstrated an ability to alter

cr

the natural history of PMF, but high treatment-related mortality risks limit the utility of

us

alloHSCT to a minority of patients with PMF or myelofibrosis secondary to other myeloproliferative neoplasms. The recent development of therapies that regulate the

an

Janus kinase–signal transducer and activator of transcription signaling pathway has changed the treatment landscape from primarily palliative treatment to potential disease

JAK inhibitors

d

Ac ce p

Myelofibrosis

te

Keywords (max = 6):

M

modification.

Proinflammatory cytokines Spleen volume

Targeted therapy

2 Page 2 of 40

1. Introduction Primary myelofibrosis (PMF) is a Philadelphia chromosome–negative

ip t

myeloproliferative neoplasm (MPN) characterized by progressive bone marrow fibrosis resulting in increasingly ineffective hematopoiesis, extramedullary hematopoiesis, a

cr

variety of inflammatory and vascular complications, and shortened survival. Patients

us

with PMF have high circulating levels of proinflammatory cytokines believed to be responsible for constitutional symptoms and much of the morbidity associated with the

an

disease. Common clinical manifestations of PMF include progressive hepatosplenomegaly, abnormal blood counts, and debilitating symptoms such as

M

fatigue, weight loss, night sweats, fever, pruritus, bone pain, early satiety, abdominal

d

pain or discomfort, arthralgias, myalgias, parasthesias, and even thromboembolic

te

disease [1,2]. Both post-essential thrombocythemia (ET) myelofibrosis (PET-MF) and post-polycythemia vera (PV) myelofibrosis (PPV-MF) are phenotypically similar to PMF

Ac ce p

and all 3 diseases have a similar clinical course [3]. Consequently, most recent and ongoing clinical trials have mixed study populations of patients with PMF, PPV-MF, or PET-MF. In the absence of a clonal MPN, fibrosis in the bone marrow can occur in nonneoplastic states. However, for the sake of simplicity and for the purposes of this manuscript, PMF, PPV-MF, or PET-MF will be collectively described as myelofibrosis (MF). MF pathogenesis involves aberrant activation of the Janus kinase–signal transducer and activator of transcription (JAK-STAT) pathway, which may result from somatic mutations directly affecting JAK activity, excessive cytokine stimulation, and/or

3 Page 3 of 40

epigenetic modifications of chromatin structure interfering with normal regulation of gene expression [4-6]. The JAK2V617F gain-of-function mutation was identified several years ago and its association with myeloproliferative phenotypes of PMF, ET, and PV is

ip t

well established [7-10]. Additional mutations within the JAK-STAT pathway have been identified since, and recent evidence suggests that mutations outside of the JAK-STAT

cr

pathway such as TET2 and ASXL1 also have important roles in pathogenesis [4,5].

us

Dysregulation within the JAK-STAT pathway and epigenetic modifiers that influence the JAK-STAT pathway are commonly seen in MPNs, but no single dominant, sufficient,

an

genetic mutation has been identified.

The discovery of the JAK2V617F mutation in patients with MPNs prompted

M

further investigation in preclinical models and eventually led to the clinical development

d

of JAK-targeted therapy for PMF. Several JAK inhibitors are currently in development

te

(Table 1), and one agent, the oral JAK1/JAK2 inhibitor ruxolitinib, has been approved for patients with intermediate-1, intermediate-2, or high-risk PMF, PPV-MF, or PET-MF.

Ac ce p

In 2 phase III clinical trials in patients with intermediate-2 or high-risk MF, ruxolitinib has been shown to significantly reduce spleen volume and to alleviate MF-related symptoms compared with placebo or best available therapy (BAT) [11,12]. These advantages translated into clinically important improvements in quality of life (QOL) measures [1214], and long-term follow-up data from the 2 studies suggest that ruxolitinib prolongs overall survival versus both placebo and BAT in patients with MF [15,16]. Aside from allogeneic hematopoietic stem cell transplantation (alloHSCT), previously used therapies for PMF have been palliative, but with limited benefits in QOL and symptom management. Until recently, the only treatment with a clearly

4 Page 4 of 40

demonstrated impact on disease progression has been alloHSCT, but treatment-related mortality is high and only a minority of patients qualify for this intensive therapy [17]. New agents have the potential to alter the natural history and make a long-term impact

ip t

on the disease. The aim of this article is to explore current evidence that new rationally

cr

designed therapy may alter the natural history of MF.

us

2. Markers of disease progression and incompleteness of prognostic scoring systems

an

The prognosis of PMF is variable. Risk stratification with the International Prognostic Scoring System (IPSS) [18], the Dynamic IPSS (DIPSS) [19], or the DIPSS-

M

Plus [20] has helped clinicians to assign risk and determine appropriate candidates for

d

alloHSCT. Each scoring system incorporates assorted patient risk factors for survival in

te

predictive models based on multiparametric regression analyses. Important disease characteristics predictive of poor survival that are included in all of these scoring

Ac ce p

systems are age of >65 years, presence of constitutional symptoms (weight loss >10% of the baseline value in the year preceding diagnosis and/or unexplained fever or excessive sweats persisting for >1 month), hemoglobin 25 × 109/L, and circulating blasts ≥1% [18-20]. In addition, DIPSS-Plus includes unfavorable karyotype, platelet count 95% of patients with PV [42]. It results in

ip t

constitutive activity of JAK2, which leads to excessive production of proinflammatory cytokines and transcription of cell survival–promoting molecules and anti-apoptotic

cr

molecules. The JAK-STAT pathway also may be overactive in patients who do not

us

harbor the JAK2V617F mutation. For example, mutations in JAK2 exon 12 and the myeloproliferative leukemia virus proto-oncogene (MPL) encoding the thrombopoietin

an

receptor, as well as in KIT, CBL, and TET2, have been consistently identified in patients with MPNs [1]. Recent analyses showed that most patients without a JAK2 or MPL

M

mutation harbor mutations in the calreticulin gene (CALR), and cells expressing these

d

mutations in CALR were sensitive to JAK2 inhibition [38,43]. In addition, patients without

te

the JAK2V617F mutation have shown clinical responses to JAK inhibitor therapy [11], including individual patients with CALR mutations [44]. Altered gene expression as a

Ac ce p

result of epigenetic dysregulation in MPNs also may affect genes that play a role in cell survival, differentiation, and/or proliferation, including those encoding calcitonin A, ABL1, SFRP2, WIF-1, SOCS1, SOCS3, CXCR4, and RARβ2 [4]. For example, ASXL1 mutations have been associated with epigenetic dysregulation resulting in the loss of transcriptional repression of leukemogenic genes and in the silencing of tumor suppressor genes [45]. Thus, aberrant expression of numerous genes may contribute to the pathogenesis of PMF, regardless of the presence or absence of the JAK2V617F mutation.

9 Page 9 of 40

4. JAK inhibition: treatment responses and effect on survival Studies of JAK inhibition have shown benefits with regard to spleen volume, MFrelated symptoms, QOL, body weight, cholesterol levels, biomarkers and

ip t

histopathology, and overall survival.

cr

4.1. Spleen volume

us

The durability of spleen volume reductions with ruxolitinib treatment in patients with intermediate-2 or high-risk MF has now been demonstrated by long-term follow-up

an

data from both COMFORT-I and COMFORT-II. In COMFORT-I, almost two-thirds of the patients who achieved a ≥35% reduction in spleen volume from baseline maintained

M

this level of reduction for at least 2 years [15], and the probability of maintaining this

d

response for >132 weeks was 53% [46]. Durable reductions in spleen volume also were

te

observed in long-term analyses of data from COMFORT-II [16]. Treatment of patients with intermediate- or high-risk MF with the JAK1/2 inhibitor momelotinib (CYT387) for up

Ac ce p

to 2.5 years in a phase I/II trial resulted in reductions in palpable spleen size of ≥50% in more than half of the eligible patients, with a maximal response duration of almost 2 years [47]. Data from studies of other JAK inhibitors have thus far shorter periods of follow-up. Nonetheless, they also demonstrate the efficacy of JAK inhibition in improving splenomegaly. In a phase II trial, treatment with the JAK2 inhibitor pacritinib provided a ≥35% reduction in spleen volume at week 24 from baseline in 32% of patients with MF, and at the time of the report, the median duration of response had not been reached [48]. Similarly, considerable reductions in splenomegaly have been seen in early phase clinical trials with the JAK2 inhibitors fedratinib (SAR302503/TG101348) [49,50] and

10 Page 10 of 40

lestaurtinib [51], and with LY2784544 [52,53], a selective JAK2V617F inhibitor. In a phase II study of fedratinib, 30% to 64% of patients with MF achieved a ≥35% reduction in spleen volume after three 28-day cycles of therapy, illustrating the speed of

ip t

considerable spleen size reduction [49]. (Unfortunately, clinical development of the drug was discontinued because of safety concerns prompted by reports of cases of

us

cr

Wernicke’s encephalopathy [54]).

4.2. Myelofibrosis-related symptoms and quality of life

an

JAK inhibition improves MF-related symptoms, which seems to improve QOL. Ruxolitinib treatment has been shown to improve MF symptoms and QOL measures

M

compared with both placebo and BAT [11,12]. In addition, spleen volume reductions

d

were correlated with improvements in MF symptoms and QOL in COMFORT-I [13]. In

te

the phase I trial of momelotinib, most treated patients experienced an improvement in constitutional symptoms [47]. Pacritinib therapy was associated with significant

Ac ce p

reductions in MF symptoms at 6 months, which included abdominal pain, bone pain, early satiety, worst fatigue, inactivity, night sweats, and pruritus [48]. The clinical experience with fedratinib also is marked with improvement or resolution of early satiety, fatigue, night sweats, itching, abdominal pain, and abdominal discomfort [49,50]. Reductions of symptom burden with LY2784544 were seen within the first 2 treatment cycles and included improvements in key MPN-related symptoms, fatigue/enjoyment of life, fatigue/walking ability, itching, early satiety, inactivity, bone pain, and night sweats [52]. At 12 weeks, a ≥50% reduction in total symptom burden was observed in 56% of patients [53].

11 Page 11 of 40

4.3. Body weight and metabolic status Cachexia is a common manifestation of disease progression, and unwanted

ip t

weight loss has been established as a negative prognostic factor [18,23]. Cachexia in patients with MF likely is related to the excessive production of cytokines such as

cr

interleukin 6 (IL-6) and tumor necrosis factor alpha (TNF-α), which are elevated in these

us

patients [55]. Elevated blood levels of IL-6, which signals through the JAK-STAT pathway, have been associated with cancer-related cachexia in clinical studies [56,57],

an

and TNF-α infusion recently has been shown to promote muscle protein loss and increased IL-6 release in healthy individuals [58]. In COMFORT-I, ruxolitinib treatment

M

was associated with increases in body weight and total cholesterol from baseline,

d

whereas both parameters decreased over time in patients who received placebo [59].

te

Specific effects of JAK inhibition on cachexia are not well illustrated, but improvement of appetite and metabolic status are strongly correlated with spleen size reduction.

Ac ce p

Changes in body weight or total cholesterol with JAK inhibitors other than ruxolitinib have not been reported to date.

4.4. Biomarkers of inflammation

Chronically elevated levels of proinflammatory cytokines have been implicated in the pathogenesis of bone marrow fibrosis [60] as well as in the mediation of noxious effects underlying the compromised metabolic/nutritional status in affected patients [6,11,55]. In addition, the circulating levels of some cytokines, including IL-8, IL-2R, IL12, and IL-15, have been shown to be independent prognostic factors of survival in

12 Page 12 of 40

patients with MF, which suggests a possible role in disease progression [6]. Because these cytokines may support the tumor microenvironment through autocrine signaling that promotes tumor cell survival and proliferation, it is reasonable to hypothesize that

ip t

the normalization of circulating cytokine levels may alter the trajectory of disease

evolution. Rapid reductions in plasma levels of proinflammatory cytokines have been

cr

observed in response to ruxolitinib (IL-8, IL-6, and TNF-α) [11,55] and fedratinib (TNF-α,

us

IL-1RA, and IL-18) [49]. Given the association between elevated cytokine expression

an

and survival [6], this decrease in inflammatory biomarkers is encouraging.

4.5. Bone marrow fibrosis

M

Bone marrow fibrosis is variable and typically graded by the density of reticulin

d

and collagen fibers in marrow replacement [61]. In PMF, marrow fibrosis is usually

te

progressive and there is increasing evidence that bone marrow fibrosis grade has prognostic significance [21,22]. Marrow fibrosis may resolve spontaneously in rare

Ac ce p

cases [62,63] or with alloHSCT [64] and may respond to treatment with interferon-alpha (IFN-α) in select patients [65]. Otherwise, reversal of marrow fibrosis has not been seen with traditional therapies. However, a post hoc analysis from the ruxolitinib phase I/II study suggests that JAK inhibition may be able to slow the progression of bone marrow fibrosis in some patients [66,67]. Bone marrow biopsies from patients treated with ruxolitinib in a single institution phase I/II study were matched to historical control bone marrow biopsies from patients who received BAT in Europe at corresponding time points. At 5 years of follow-up, 35% of ruxolitinib-treated compared with 3% of BATtreated patients showed improvement in bone marrow fibrosis grade. Furthermore, the

13 Page 13 of 40

odds for worsening bone fibrosis was significantly greater with BAT (76%; n=29) than ruxolitinib (26%; n=23) [67]. Consistent with the notion that JAK inhibition may have a positive impact on bone marrow fibrosis [68], data from the phase I study of LY2784544

ip t

showed that after 5 cycles of therapy, bone marrow fibrosis grade was reduced in 3 of 5 MF patients analyzed [52].

cr

Although encouraging, the improvement in bone marrow fibrosis observed in

us

patients receiving JAK inhibitors generally occurred slowly and overall was modest. For many patients lasting improvement of bone marrow function may require a multifaceted

an

approach beyond the use of JAK inhibitors. In the future, this approach may include the use of epigenetic modifiers such as the histone deacetylase (HDAC) inhibitor

M

panobinostat [69,70], or antifibrotic agents such as PRM-151 (recombinant human

d

pentraxin-2) [71] and a monoclonal antibody against LOXL-2 [72]. These agents will be

Ac ce p

4.6. Overall survival

te

discussed in more detail in Section 5.

Follow-up data from COMFORT-I and COMFORT-II suggest that ruxolitinib therapy was associated with a survival advantage compared with both placebo and BAT [15,16]. These analyses were based on randomized treatment allocation at the beginning of the studies, and the control arms included patients who crossed over to ruxolitinib treatment after meeting prespecified conditions of disease progression. In COMFORT-I, all patients randomized to placebo had discontinued or were receiving ruxolitinib within 3 months after the primary analysis [15]. The short exposure to placebo taken together with the growing exposure to ruxolitinib treatment in the placebo group

14 Page 14 of 40

has made interpretation of survival benefits difficult [46]. Exploratory statistical analyses suggest that the diminished survival disadvantage of the placebo arm (compared with the ruxolitinib arm) at 3 versus 2 years follow-up may be a direct result of the crossover

ip t

to ruxolitinib in this treatment arm [46]. A corresponding effect was not observed in COMFORT-II [16], likely because of delayed crossover from BAT to ruxolitinib

cr

consequent to differences in crossover rules between the COMFORT studies.

us

Nonetheless, depending on the time of analysis, patients randomized to ruxolitinib generally had a survival advantage of 30% to 50% compared with those randomized to

an

placebo [11,15] or BAT [16]. Consistent with these findings, a comparison of survival data from patients with PMF from the COMFORT-II study with those from a matched

M

control population of the DIPSS study [73] revealed a 36% reduction in the risk of death

d

with ruxolitinib versus conventional therapy (HR = 0.64; 95% confidence interval:

te

0.4‒0.96; P = 0.034). Additional analyses showed that the use of ruxolitinib doubled the 8-year survival probability at diagnosis from 15.9% to 32.2%. Together, these data

Ac ce p

suggest that ruxolitinib may affect disease progression and alter the natural history of the disease.

5. Rational approaches to modification of disease beyond JAK inhibition In addition to the investigational JAK inhibitors, agents targeted to other signaling pathways or to epigenetic modifiers are in various stages of clinical development for the treatment of MPN-associated MF. They include HDAC inhibitors and immunomodulatory drugs. In addition, the antifibrotic monoclonal antibodies fresolimumab and GS-6624, recombinant PTX-2, and the telomerase inhibitor imetelstat

15 Page 15 of 40

(GRN163L) may have potential roles in treatment as single agents or in combination therapy. Recently, encouraging results were obtained in a single-center study of the telomerase inhibitor imetelstat in patients with intermediate-2 or high-risk MF. Of 18

ip t

patients followed for at least 3 months, 8 (44%) had an overall response, including 4 patients with complete remission, as evidenced by the reversal of BM fibrosis and

cr

recovery of normal megakaryocyte morphology, and 1 patient with partial remission

us

[74].

In addition to imetelstat, approaches complementary to JAK inhibitor therapy to

an

reverse bone marrow fibrosis or retard its progression are being explored. The human monoclonal antibody fresolimumab antagonizes transforming growth factor beta

M

(TGFβ), a mediator of bone marrow fibrosis in thrombopoietin-induced MF [75,76], and

d

it has been safely administered to patients with focal segmental glomerulosclerosis [77].

te

Likewise, a monoclonal antibody (GS6624, AB0023) targeting lysyl oxidase-like 2 (LOXL-2), a protein active in the remodeling of extracellular matrix architecture in

Ac ce p

pathologic fibrosis, has been shown to reduce TGFβ pathway signaling and fibroblast activation in murine models of fibrosis [72]. A phase II study of the anti-LOXL-2 antibody GS6624 as monotherapy or in combination with ruxolitinib is underway in patients with MF. PRM-151 (recombinant human serum amyloid P/pentraxin 2), an antifibrotic protein, has recently been shown in an initial phase I study to be well tolerated and to reduce fibrocyte numbers by 30% to 50% in patients with pulmonary fibrosis [78]. Investigation in patients with MF is underway. Panobinostat (LBH589) is a pan-HDAC inhibitor that was shown in a phase I trial to achieve a 100% reduction in palpable splenomegaly in 3 of 5 patients who received

16 Page 16 of 40

at least 6 cycles of treatment and were evaluable for response; the other 2 patients had stable disease [79]. One of the 3 patients who had 100% reduction in splenomegaly demonstrated bone marrow fibrosis resolution after 16 cycles [79]. In a phase II trial of

ip t

22 patients with intermediate- or high-risk MF, the HDAC inhibitor pracinostat achieved a median 3-cm reduction in splenomegaly in 6 (27%) patients [80]. These and other

cr

targeted agents may be most effective when given in combination with JAK inhibitors to

us

maximize blockade of disease mechanisms. Studies of combination therapy with rational combinations of these targeted agents are in progress. In a recent phase I

an

study, prolonged therapy with panobinostat elicited dramatic reversal of bone marrow histopathologic abnormalities in 2 patients [79]. A combination study of panobinostat

M

and ruxolitinib in patients with MF is ongoing.

d

The use of IFN-α may lead to spleen size and symptom improvement as well as

te

reduction in marrow fibrosis in some patients, particularly those with early-stage disease [65]. However, as a recent analysis of studies of IFN-α shows, widely variable

Ac ce p

responses among different groups of MF patients have been reported, and the use of IFN-α in more advanced disease is not recommended [81]. The value of IFN-α for certain subgroups of patients with MF or in combination with JAK inhibitors has yet to be demonstrated.

6. JAK inhibition and changing the natural history of myelofibrosis Development of therapy that regulates the JAK-STAT pathway represents an important step forward towards therapy that alters the natural history of the disease. Although the most salient benefits are the alleviation of splenomegaly and non-spleen–

17 Page 17 of 40

related symptoms, concomitant with an improvement in QOL measures [11,12], there is compelling evidence that these therapies may improve the prognosis of these patients [15,16,82]. The mitigation of cachexia-associated risk factors, including weight loss

ip t

[18,59], hypocholesterolemia [23,24,59], and elevated inflammatory cytokines [6,11,55] likely contributes to the apparent improved survival seen with the use of ruxolitinib.

cr

Moreover, at least in some patients treated with these new agents, progression of

us

fibrosis may be retarded or relent [67,68,74]. With our current understanding, worsening marrow fibrosis, while clearly associated with cytopenias and ineffective hematopoiesis,

an

is not definitively linked with a decline in overall survival. Moreover, although most patients with marrow fibrosis improvements had corresponding improvements in

M

symptoms and spleen size in observational studies, changes in marrow fibrosis do not

d

always parallel spleen size reduction or relief of disease-related symptoms [65,67]. In

te

addition, the rapid clinical improvements seen in the COMFORT-II study were not accompanied by or dependent on major changes in marrow histomorphology [12].

Ac ce p

This raises the intriguing possibility that new therapies delay disease progression, including symptom onset in asymptomatic patients. A thorough assessment of the benefits and risks of these agents for lower-risk patients may require further exploration in clinical trials. Intervention in early stages of PMF with targeted therapies capable of changing the disease trajectory (Fig. 2) may lead to better outcomes, including longer survival and increased QOL. Because of the inherent clinical and genetic heterogeneity of the disease, individualized risk assessment may be helpful in predicting the disease course of individual patients more accurately. It is conceivable that specific patterns of inflammatory cytokine expression or gene expression

18 Page 18 of 40

signatures may be used in the future to identify patients who are at risk of more rapid deterioration and therefore would be candidates for earlier intervention. Elevated expression of IL-8 in the serum, which appears to be secondary to increased expression

ip t

of nuclear factor eythroid-2 [83], has been identified as an independent risk factor for shortened survival in PMF [6], and serum levels may revert to normal values in both

cr

JAK2V617F-positive and -negative patients in response to JAK inhibitor therapy [55].

us

JAK inhibitors have shown to be effective in patients with and without the JAK2V617F mutation [11,84].

an

There are conflicting arguments as to the extent by which JAK2V617F allele burden correlates with disease severity in ET, PV and MF [85-87], but the general

M

consensus is that allele burden is higher in more severe disease, at least in the case of

d

ET. Furthermore, low allele burden at baseline was associated with improved treatment

te

response to IFN-α in patients with PV or ET [88], whereas allele burden >1% shortly after alloHSCT was associated with increased risk of relapse and poorer overall survival

Ac ce p

[89]. JAK2V617F allele burden, which can be measured reliably from either peripheral blood or bone marrow biopsies [90] may prove useful and convenient in some clinical settings, such as estimating the risks of myelofibrotic transformation or thrombosis in patients with ET or PV [91]. However, JAK2V617F allele burden has not yet proven useful as a prognostic marker or harbinger of clinical responses to JAK inhibitors [84,92].

7. Conclusions

19 Page 19 of 40

With the exception of alloHSCT, and perhaps IFN-α in a select group of patients, conventional therapies do not impact the natural history of PMF in any appreciable way. Considering the successes achieved with therapy targeting the JAK-STAT pathway and

ip t

the promising results achieved in early trials with other therapies targeted to pathogenic mechanisms of PMF, the treatment strategies and expectations are changing. Major

cr

advances in the treatment have been realized with the introduction of JAK inhibitors,

us

and now a more versatile armamentarium is needed, given both the heterogeneity of the disease and the diversity of risk factors. Antifibrotic agents, immunomodulators, and

an

epigenetic modifiers, which are currently in clinical trials, are plausible candidates as monotherapy or in combination with JAK inhibitors. It is hoped that earlier identification

M

of patients with poor prognosis, together with more options for disease-modifying

te

patients.

d

therapy, will eventually lead to greater improvements in survival in a greater number of

A growing sentiment that the new rational therapies may change the natural

Ac ce p

history of the disease is reflected in the new consensus recommendations by the International Working Group‒Myeloproliferative Neoplasms Research and Treatment and European LeukemiaNet [93]. In addition to specifying the clinical and hematologic criteria for clinical improvement, anemia response, spleen response, and symptom response, the consensus recommendations provide rigorous definitions for complete and partial remission based on cytogenetic and molecular response [93]. With the accumulation of evidence indicating the potential for targeted agents to modify the disease course, there is new optimism that the goal of obtaining a complete response with drug therapy in an increasing number of patients is within reach. Disease activity in

20 Page 20 of 40

these patients is manifested by a variety of symptoms and metabolic derangements that seem to improve with JAK inhibition. In a progressive catabolic state such as PMF, sustained mitigation of these clinical characteristics seems to lead to improved survival,

ip t

consistent with the notion that the trajectory of the natural history in these patients has

cr

changed.

us

Funding

an

Medical writing support was funded by Incyte Corporation.

Conflicts of interest

M

Dr. Savona is a consultant for Karyopharm, Gilead Corporation, and Celgene Corporation, and serves on advisory boards for Karyopharm, Celgene, Bristol-Myers

Ac ce p

Acknowledgments

te

d

Squibb, Novartis, and Incyte Corporation.

I thank Professor Jerry Spivak for key observations and thoughtful suggestions. Medical writing support, funded by Incyte Corporation, was provided by Stephanie Leinbach, PhD, and by Roland Tacke, PhD, of Evidence Scientific Solutions.

21 Page 21 of 40

References [1]

Mascarenhas J, Mughal TI, Verstovsek S. Biology and clinical management of

ip t

myeloproliferative neoplasms and development of the JAK inhibitor ruxolitinib. Curr Med Chem 2012;19:4399–413.

Vannucchi AM. Management of myelofibrosis. Hematology Am Soc Hematol Educ

cr

[2]

[3]

us

Program 2011;2011:222–30.

Abdel-Wahab O, Pardanani A, Bernard OA, Finazzi G, Crispino JD, Gisslinger H, et al.

an

Unraveling the genetic underpinnings of myeloproliferative neoplasms and understanding their effect on disease course and response to therapy: proceedings from

[4]

M

the 6th International Post-ASH Symposium. Am J Hematol 2012;87:562–8. Mascarenhas J, Roper N, Chaurasia P, Hoffman R. Epigenetic abnormalities in

d

myeloproliferative neoplasms: a target for novel therapeutic strategies. Clin Epigenetics

[5]

te

2011;2:197–212.

Cross NC. Genetic and epigenetic complexity in myeloproliferative neoplasms.

[6]

Ac ce p

Hematology Am Soc Hematol Educ Program 2011;2011:208–14. Tefferi A, Vaidya R, Caramazza D, Finke C, Lasho T, Pardanani A. Circulating interleukin (IL)-8, IL-2R, IL-12, and IL-15 levels are independently prognostic in primary myelofibrosis: a comprehensive cytokine profiling study. J Clin Oncol 2011;29:1356–63. [7]

Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 2005;365:1054–61.

[8]

James C, Ugo V, Le Couédic JP, Staerk J, Delhommeau F, Lacout C, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005;434:1144–8.

22 Page 22 of 40

[9]

Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg JR, et al. A gain-offunction mutation of JAK2 in myeloproliferative disorders. N Engl J Med 2005;352:1779– 90. Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntly BJ, et al. Activating

ip t

[10]

mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia,

[11]

cr

and myeloid metaplasia with myelofibrosis. Cancer Cell 2005;7:387–97.

Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, et al. A double-blind,

[12]

us

placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med 2012;366:799–807. Harrison C, Kiladjian JJ, Al-Ali HK, Gisslinger H, Waltzman R, Stalbovskaya V, et al. JAK

an

inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med 2012;366:787–98.

Mesa RA, Gotlib J, Gupta V, Catalano JV, Deininger MW, Shields AL, et al. Effect of

M

[13]

ruxolitinib therapy on myelofibrosis-related symptoms and other patient-reported

Harrison CN, Mesa RA, Kiladjian JJ, Al-Ali HK, Gisslinger H, Knoops L, et al. Health-

Ac ce p

[14]

te

Oncol 2013;31:1285–92.

d

outcomes in COMFORT-I: a randomized, double-blind, placebo-controlled trial. J Clin

related quality of life and symptoms in patients with myelofibrosis treated with ruxolitinib versus best available therapy. Br J Haematol 2013;162:229–39. [15]

Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, Dipersio JF, et al. Efficacy, safety and survival with ruxolitinib treatment in patients with myelofibrosis: results of a median 2-year follow-up of COMFORT-I. Haematologica 2013;98:1865–71.

[16]

Cervantes F, Vannucchi AM, Kiladjian JJ, Al-Ali HK, Sirulnik A, Stalbovskaya V, et al.; on behalf of the COMFORT-II investigators. 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.

23 Page 23 of 40

[17]

Gupta V, Hari P, Hoffman R. Allogeneic hematopoietic cell transplantation for myelofibrosis in the era of JAK inhibitors. Blood 2012;120:1367–79.

[18]

Cervantes F, Dupriez B, Pereira A, Passamonti F, Reilly JT, Morra E, et al. New

ip t

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. Passamonti F, Cervantes F, Vannucchi AM, Morra E, Rumi E, Pereira A, et al. A

cr

[19]

dynamic prognostic model to predict survival in primary myelofibrosis: a study by the

us

IWG-MRT (International Working Group for Myeloproliferative Neoplasms Research and Treatment). Blood 2010;115:1703–8.

Gangat N, Caramazza D, Vaidya R, George G, Begna K, Schwager S, et al. DIPSS plus:

an

[20]

a refined Dynamic International Prognostic Scoring System for primary myelofibrosis that

M

incorporates prognostic information from karyotype, platelet count, and transfusion status. J Clin Oncol 2011;29:392–7.

Gianelli U, Vener C, Bossi A, Cortinovis I, Iurlo A, Fracchiolla NS, et al. The European

d

[21]

te

Consensus on grading of bone marrow fibrosis allows a better prognostication of

[22]

Ac ce p

patients with primary myelofibrosis. Mod Pathol 2012;25:1193–202. Lekovic D, Gotic M, Perunicic-Jovanovic M, Vidovic A, Bogdanovic A, Jankovic G, et al. Contribution of comorbidities and grade of bone marrow fibrosis to the prognosis of survival in patients with primary myelofibrosis. Med Oncol 2014;31:869. [23]

Mesa RA, Schwager S, Huang J, Pardanani AD, Hussein K, Camoriano J, et al. Weight loss, splenomegaly, and hypocholesterolemia in myeloproliferative neoplasms: patterns and relevance from the pre JAK2 inhibitor era. Blood (ASH Annual Meeting Abstracts) 2009;114:3918.

[24]

Sulai N, Mengistu B, Gangat N, Hanson CA, Ketterling RP, Pardanani A, et al. Decreased levels of total or HDL cholesterol in primary myelofibrosis are associated with

24 Page 24 of 40

shortened survival: DIPSS-Plus independent prognostic value. Blood (ASH Annual Meeting Abstracts) 2012;120:2851. [25]

Vannucchi AM, Kantarjian H, Kiladjian J-J, Gotlib J, Cervantes F, Mesa RA, et al. A

ip t

pooled overall survival analysis of the COMFORT studies: 2 randomized phase 3 trials of ruxolitinib for the treatment of myelofibrosis. Blood (ASH Annual Meeting Abstracts)

[26]

cr

2013;122:2820.

Vannucchi AM, Lasho TL, Guglielmelli P, Biamonte F, Pardanani A, Pereira A, et al.

[27]

us

Mutations and prognosis in primary myelofibrosis. Leukemia 2013;27:1861–9. Mesa RA, Niblack J, Wadleigh M, Verstovsek S, Camoriano J, Barnes S, et al. The

an

burden of fatigue and quality of life in myeloproliferative disorders (MPDs): an international Internet-based survey of 1179 MPD patients. Cancer 2007;109:68–76. Mesa RA, Li CY, Ketterling RP, Schroeder GS, Knudson RA, Tefferi A. Leukemic

M

[28]

transformation in myelofibrosis with myeloid metaplasia: a single-institution experience

Firmbach-Kraft I, Byers M, Shows T, Dalla-Favera R, Krolewski JJ. tyk2, prototype of a

te

[29]

d

with 91 cases. Blood 2005;105:973–7.

[30]

Ac ce p

novel class of non-receptor tyrosine kinase genes. Oncogene 1990;5:1329–36. Wilks AF, Harpur AG, Kurban RR, Ralph SJ, Zürcher G, Ziemiecki A. Two novel proteintyrosine kinases, each with a second phosphotransferase-related catalytic domain, define a new class of protein kinase. Mol Cell Biol 1991;11:2057–65. [31]

Harpur AG, Andres AC, Ziemiecki A, Aston RR, Wilks AF. JAK2, a third member of the JAK family of protein tyrosine kinases. Oncogene 1992;7:1347–53.

[32]

Rane SG, Reddy EP. JAK3: a novel JAK kinase associated with terminal differentiation of hematopoietic cells. Oncogene 1994;9:2415–23.

[33]

Neubauer H, Cumano A, Müller M, Wu H, Huffstadt U, Pfeffer K. Jak2 deficiency defines an essential developmental checkpoint in definitive hematopoiesis. Cell 1998;93:397– 409. 25 Page 25 of 40

[34]

Parganas E, Wang D, Stravopodis D, Topham DJ, Marine JC, Teglund S, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell 1998;93:385–95.

[35]

Witthuhn BA, Quelle FW, Silvennoinen O, Yi T, Tang B, Miura O, et al. JAK2 associates

ip t

with the erythropoietin receptor and is tyrosine phosphorylated and activated following stimulation with erythropoietin. Cell 1993;74:227–36.

Shuai K, Ziemiecki A, Wilks AF, Harpur AG, Sadowski HB, Gilman MZ, et al. Polypeptide

cr

[36]

signalling to the nucleus through tyrosine phosphorylation of Jak and Stat proteins.

[37]

us

Nature 1993;366:580–3.

Penta K, Sawyer ST. Erythropoietin induces the tyrosine phosphorylation, nuclear

an

translocation, and DNA binding of STAT1 and STAT5 in erythroid cells. J Biol Chem 1995;270:31282–7.

Klampfl T, Gisslinger H, Harutyunyan AS, Nivarthi H, Rumi E, Milosevic JD, et al.

M

[38]

2013;369:2379–90.

Lundberg P, Karow A, Nienhold R, Looser R, Hao-Shen H, Nissen I, et al. Clonal

te

[39]

d

Somatic mutations of calreticulin in myeloproliferative neoplasms. N Engl J Med

Ac ce p

evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms. Blood 2014 Jan 29 [Epub ahead of print]. [40]

Ferrer-Marín F, Bellosillo B, Martinez-Avilés L, Soler G, Carbonell P, Luengo-Gil G, et al. Leukemic transformation driven by an ASXL1 mutation after a JAK2V617F-positive primary myelofibrosis: clonal evolution and hierarchy revealed by next-generation sequencing. J Hematol Oncol 2013;6:68.

[41]

Anand S, Stedham F, Beer P, Gudgin E, Ortmann CA, Bench A, et al. Effects of the JAK2 mutation on the hematopoietic stem and progenitor compartment in human myeloproliferative neoplasms. Blood 2011;118:177–81.

[42]

Vannucchi AM, Guglielmelli P, Tefferi A. Advances in understanding and management of myeloproliferative neoplasms. CA Cancer J Clin 2009;59:171–91. 26 Page 26 of 40

[43]

Nangalia J, Massie CE, Baxter EJ, Nice FL, Gundem G, Wedge DC, et al. Somatic CALR mutations in myeloproliferative neoplasms with nonmutated JAK2. N Engl J Med 2013;369:2391–405. Passamonti F, Caramazza D, Maffioli M. JAK inhibitor in CALR-mutant myelofibrosis. N

ip t

[44]

Engl J Med 2014;370:1168-9.

Abdel-Wahab O, Adli M, LaFave LM, Gao J, Hricik T, Shih AH, et al. ASXL1 mutations

cr

[45]

promote myeloid transformation through loss of PRC2-mediated gene repression.

[46]

us

Cancer Cell 2012;22:180–93.

Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, Dipersio JF, et al. Long-term

an

outcomes of ruxolitinib therapy in patients with myelofibrosis: 3-year update from COMFORT-I. Blood (ASH Annual Meeting Abstracts) 2013;122:396. Pardanani A, Laborde RR, Lasho TL, Finke C, Begna K, Al-Kali A, et al. Safety and

M

[47]

2013;27:1322–7.

Komrokji RS, Wadleigh M, Seymour JF, Roberts AW, To LB, Zhu HJ, et al. Results of a

te

[48]

d

efficacy of CYT387, a JAK1 and JAK2 inhibitor, in myelofibrosis. Leukemia

Ac ce p

Phase 2 study of pacritinib (SB1518), a novel oral JAK2 inhibitor, in patients with primary, post-polycythemia vera, and post-essential thrombocythemia myelofibrosis. Blood (ASH Annual Meeting Abstracts) 2011;118:282. [49]

Talpaz M, Jamieson C, Gabrail NY, Lebedinsky C, Gao G, Liu F, et al. A Phase II randomized dose-ranging study of the JAK2-selective inhibitor SAR302503 in patients with intermediate-2 or high-risk primary myelofibrosis (MF), post-polycythemia vera (PV) MF, or post-essential thrombocythemia (ET) MF. Blood (ASH Annual Meeting Abstracts) 2012;120:2837.

[50]

Pardanani A, Gotlib JR, Jamieson C, Cortes JE, Talpaz M, Stone RM, et al. Safety and efficacy of TG101348, a selective JAK2 inhibitor, in myelofibrosis. J Clin Oncol 2011;29:789–96. 27 Page 27 of 40

[51]

Santos FP, Kantarjian HM, Jain N, Manshouri T, Thomas DA, Garcia-Manero G, et al. Phase 2 study of CEP-701, an orally available JAK2 inhibitor, in patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis. Blood 2010;115:1131–

[52]

ip t

6. Verstovsek S, Mesa RA, Rhoades SK, Giles JLK, Pitou C, Jones E, et al. Phase I study

cr

of the JAK2 V617F inhibitor, LY2784544, in patients with myelofibrosis (MF),

polycythemia vera (PV), and essential thrombocythemia (ET). Blood (ASH Annual

[53]

us

Meeting Abstracts) 2011;118:2814.

Verstovsek S, Mesa RA, Salama ME, Giles JLK, Pitou C, Zimmermann AH, et al. Phase

an

I study of LY2784544, a JAK2 selective inhibitor, in patients with myelofibrosis (MF),

Meeting Abstracts) 2013;122:665. [54]

M

polycythemia vera (PV), and essential thrombocythemia (ET). Blood (ASH Annual

Sanofi discontinues clinical development of investigational JAK2 agent fedratinib

d

(SAR302503) [press release]. Paris, France: Sanofi; 8 Nov 2013. http://m-

[55]

Ac ce p

2014.

te

en.sanofi.com/Images/34935_20131118_JAK-2-FEDRATINIB_en.pdf. Accessed 27 Feb

Verstovsek S, Kantarjian H, Mesa RA, Pardanani AD, Cortes-Franco J, Thomas DA, et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med 2010;363:1117–27.

[56]

Scott HR, McMillan DC, Crilly A, McArdle CS, Milroy R. The relationship between weight loss and interleukin 6 in non-small-cell lung cancer. Br J Cancer 1996;73:1560–2.

[57]

Iwase S, Murakami T, Saito Y, Nakagawa K. Steep elevation of blood interleukin-6 (IL-6) associated only with late stages of cachexia in cancer patients. Eur Cytokine Netw 2004;15:312–6.

[58]

Bach E, Nielsen RR, Vendelbo MH, Møller AB, Jessen N, Buhl M, et al. Direct effects of TNF-α on local fuel metabolism and cytokine levels in the placebo-controlled, bilaterally 28 Page 28 of 40

infused human leg: increased insulin sensitivity, increased net protein breakdown, and increased IL-6 release. Diabetes 2013;62:4023–9. [59]

Mesa RA, Verstovsek S, Gupta V, Mascarenhas J, Atallah E, Sun W, et al. Improvement

ip t

in weight and total cholesterol and their association with survival in ruxolitinib-treated patients with myelofibrosis from COMFORT-I. Blood (ASH Annual Meeting Abstracts)

Hasselbalch HC. The role of cytokines in the initiation and progression of myelofibrosis. Cytokine Growth Factor Rev 2013;24:133–45.

[61]

us

[60]

cr

2012;120:1733.

Thiele J, Kvasnicka HM, Facchetti F, Franco V, van der Walt J, Orazi A. European

an

consensus on grading bone marrow fibrosis and assessment of cellularity. Haematologica 2005;90:1128–32.

Argano SA, Marasa L, Daniele E, Pitre V, Morello V, Lampasona G, et al. Experimental

M

[62]

myelofibrosis. Problems related to histogenesis and to spontaneous and therapeutic

Inoue S, Limsuwan A, McQueen R. Spontaneous resolution of myelofibrosis and

te

[63]

d

reversibility of the lesions. Haematologica 1977;62:590–602.

Ac ce p

pancytopenia followed by the development of acute myeloid leukemia with an extramedullary mass. J Pediatr Hematol Oncol 1998;20:268–70. [64]

Kröger N, Kvasnicka M, Thiele J. Replacement of hematopoietic system by allogeneic stem cell transplantation in myelofibrosis patients induces rapid regression of bone marrow fibrosis. Fibrogenesis Tissue Repair 2012;5 Suppl 1:S25.

[65]

Silver RT, Vandris K, Goldman JJ. Recombinant interferon-alpha may retard progression of early primary myelofibrosis: a preliminary report. Blood 2011;117:6669-72.

[66]

Kvasnicka HM, Thiele J, Bueso-Ramos CE, Hou K, Cortes JE, Kantarjian HM, et al. Exploratory analysis of the effect of ruxolitinib on bone marrow morphology in patients with myelofibrosis. J Clin Oncol 2013;31 Suppl:7030.

29 Page 29 of 40

[67]

Kvasnicka HM, Thiele J, Bueso-Ramos CE, Sun W, Cortes J, Kantarjian HM, et al. Effects of five-years of ruxolitinib therapy on bone marrow morphology in patients with myelofibrosis and comparison with best available therapy. Blood (ASH Annual Meeting

[68]

ip t

Abstracts) 2013;122:4055. Wilkins BS, Radia D, Woodley C, El Farhi S, Keohane C, Harrison CN. Resolution of

cr

bone marrow fibrosis in a patient receiving JAK1/JAK2 inhibitor treatment with ruxolitinib. Haematologica 2013;98:1872–6.

Santos FP, Verstovsek S. What is next beyond janus kinase 2 inhibitors for primary myelofibrosis? Curr Opin Hematol 2013;20:123–9.

Evrot E, Ebel N, Romanet V, Roelli C, Andraos R, Qian Z, et al. JAK1/2 and pan-

an

[70]

us

[69]

deacetylase inhibitor combination therapy yields improved efficacy in preclinical mouse

[71]

M

models of JAK2V617F-driven disease. Clin Cancer Res 2013;19:6230–41. Duffield JS, Lupher ML, Jr. PRM-151 (recombinant human serum amyloid P/pentraxin 2)

Barry-Hamilton V, Spangler R, Marshall D, McCauley S, Rodriguez HM, Oyasu M, et al.

te

[72]

d

for the treatment of fibrosis. Drug News Perspect 2010;23:305–15.

Ac ce p

Allosteric inhibition of lysyl oxidase-like-2 impedes the development of a pathologic microenvironment. Nat Med 2010;16:1009–17. [73]

Passamonti F, Maffioli M, Cervantes F, Vannucchi AM, Morra E, Barbui T, et al. Impact of ruxolitinib on the natural history of primary myelofibrosis: a comparison of the DIPSS and the COMFORT-2 cohorts. Blood 2014;123:1833-5.

[74]

Tefferi A, Begna K, Laborde RR, Patnaik MM, Lasho TL, Zblewski D, et al. Imetelstat, a telomerase inhibitor, induces morphologic and molecular remissions in myelofibrosis and reversal of bone marrow fibrosis. Blood (ASH Annual Meeting Abstracts) 2013;122:662.

[75]

Chagraoui H, Komura E, Tulliez M, Giraudier S, Vainchenker W, Wendling F. Prominent role of TGF-β1 in thrombopoietin-induced myelofibrosis in mice. Blood 2002;100:3495– 503. 30 Page 30 of 40

[76]

Vannucchi AM, Bianchi L, Paoletti F, Pancrazzi A, Torre E, Nishikawa M, et al. A pathobiologic pathway linking thrombopoietin, GATA-1, and TGF-β1 in the development of myelofibrosis. Blood 2005;105:3493–501. Trachtman H, Fervenza FC, Gipson DS, Heering P, Jayne DR, Peters H, et al. A phase

ip t

[77]

1, single-dose study of fresolimumab, an anti-TGF-β antibody, in treatment-resistant

[78]

cr

primary focal segmental glomerulosclerosis. Kidney Int 2011;79:1236–43.

Dillingh MR, van den Blink B, Moerland M, van Dongen MG, Levi M, Kleinjan A, et al.

fibrosis. Pulm Pharmacol Ther 2013;26:672–6.

Mascarenhas J, Lu M, Li T, Petersen B, Hochman T, Najfeld V, et al. A phase I study of

an

[79]

us

Recombinant human serum amyloid P in healthy volunteers and patients with pulmonary

panobinostat (LBH589) in patients with primary myelofibrosis (PMF) and post-

M

polycythaemia vera/essential thrombocythaemia myelofibrosis (post-PV/ET MF). Br J Haematol 2013;161:68–75.

Quintás-Cardama A, Kantarjian H, Estrov Z, Borthakur G, Cortes J, Verstovsek S.

d

[80]

te

Therapy with the histone deacetylase inhibitor pracinostat for patients with myelofibrosis.

[81]

Ac ce p

Leuk Res 2012;36:1124–7.

Nguyen HM, Kiladjian JJ. Is there a role for the use of IFN-alpha in primary myelofibrosis? Hematology Am Soc Hematol Educ Program 2012;2012:567-70.

[82]

Verstovsek S, Kantarjian HM, Estrov Z, Cortes JE, Thomas DA, Kadia T, et al. Longterm outcomes of 107 patients with myelofibrosis receiving JAK1/JAK2 inhibitor ruxolitinib: survival advantage in comparison to matched historical controls. Blood 2012;120:1202–9.

[83]

Wehrle J, Seeger TS, Schwemmers S, Pfeifer D, Bulashevska A, Pahl HL. Transcription factor nuclear factor erythroid-2 mediates expression of the cytokine interleukin 8, a known predictor of inferior outcome in patients with myeloproliferative neoplasms. Haematologica 2013;98:1073–80. 31 Page 31 of 40

[84]

Verstovsek S, Mesa RA, Gotlib J, Levy RS, Gupta V, DiPersio JF, et al. The clinical benefit of ruxolitinib across patient subgroups: analysis of a placebo-controlled, Phase III study in patients with myelofibrosis. Br J Haematol 2013;161:508–16. Ha JS, Kim YK, Jung SI, Jung HR, Chung IS. Correlations between Janus kinase 2

ip t

[85]

V617F allele burdens and clinicohematologic parameters in myeloproliferative

[86]

cr

neoplasms. Ann Lab Med 2012;32:385-91.

Pich A, Riera L, Beggiato E, Nicolino B, Godio L, Campisi P, et al. JAK2V617F mutation

us

and allele burden are associated with distinct clinical and morphological subtypes in patients with essential thrombocythaemia. J Clin Pathol 2012;65:953-5. Gebauer N, Bernard V, Gebauer W, Feller AC, Merz H. MicroRNA expression and JAK2

an

[87]

allele burden in bone marrow trephine biopsies of polycythemia vera, essential

[88]

M

thrombocythemia and early primary myelofibrosis. Acta Haematol 2013;129:251–6. Stauffer Larsen T, Iversen KF, Hansen E, Mathiasen AB, Marcher C, Frederiksen M, et

d

al. Long term molecular responses in a cohort of Danish patients with essential

te

thrombocythemia, polycythemia vera and myelofibrosis treated with recombinant

[89]

Ac ce p

interferon alpha. Leuk Res 2013;37:1041-5. Lange T, Edelmann A, Siebolts U, Krahl R, Nehring C, Jakel N, et al. JAK2 p.V617F allele burden in myeloproliferative neoplasms one month after allogeneic stem cell transplantation significantly predicts outcome and risk of relapse. Haematologica 2013;98:722-8. [90]

Takahashi K, Patel KP, Kantarjian H, Luthra R, Pierce S, Cortes J, et al. JAK2 p.V617F detection and allele burden measurement in peripheral blood and bone marrow aspirates in patients with myeloproliferative neoplasms. Blood 2013;122:3784-6.

[91]

Alvarez-Larran A, Bellosillo B, Pereira A, Kerguelen A, Hernandez-Boluda JC, MartinezAviles L, et al. JAK2V617F monitoring in polycythemia vera and essential

32 Page 32 of 40

thrombocythemia: clinical usefulness for predicting myelofibrotic transformation and thrombotic events. Am J Hematol 2014. [92]

Constantinescu SN, Vainchenker W. Small-molecule inhibitors in myeloproliferative

ip t

neoplasms: are we aiming for the right targets? Hematology Am Soc Hematol Educ Program 2012;2012:553–60.

cr

Tefferi A, Cervantes F, Mesa R, Passamonti F, Verstovsek S, Vannucchi AM, et al. Revised response criteria for myelofibrosis: International Working Group-

us

Myeloproliferative Neoplasms Research and Treatment (IWG-MRT) and European

te

d

M

an

LeukemiaNet (ELN) consensus report. Blood 2013;122:1395–8.

Ac ce p

[93]

33 Page 33 of 40

Table 1 JAK inhibitors in development for the treatment of PMF, PPV-MF, and PET-MF. Phase of

inhibited

development

Pacritinib (SB1518)

JAK2

III

Momelotinib (CYT387)

JAK1, JAK2

III

Lestaurtinib (CEP-701)

JAK2

II

LY2784544

JAK2V617F

II

AZD1480

JAK2

I/II

BMS-911543

JAK2

NS-018

JAK2

Company

ip t

JAK(s)

cr

Cell Therapeutics, Inc. Gilead Sciences, Inc.

an

us

Teva Pharmaceuticals

Eli Lilly and Company AstraZeneca

I/II

Bristol-Myers Squibb

I/II

NS Pharma, Inc.

M

Agent

JAK, Janus Kinase; PET-MF, post-essential thrombocythemia myelofibrosis; PMF,

Ac ce p

te

d

primary myelofibrosis; PPV-MF, post-polycythemia vera myelofibrosis.

34 Page 34 of 40

ip t cr

Table 2

us

DIPSS-Plus risk factors and major clinical manifestations of disease progression not captured by DIPSS-Plus. Validated laboratory risk factors not

Clinical risk factors not included in

included in DIPSS-Plus

DIPSS-Plus

Age >65

Elevated cytokines (eg, IL-8, IL-2R,

Marked splenomegaly (>10 cm below

Constitutional symptomsa

IL-12, and Il-15) [6]

left costal margin) [23] or increased

Somatic mutations (eg, ASXL1) [26]

spleen volume [25]

Hypocholesterolemia [23,24]

ce pt

(anemia)

ed

Hemoglobin 25 × 109/L (leukocytosis)

MF-related symptom burden aside from constitutional symptomsa Worsening bone marrow fibrosis [21,22]

9

Ac

Platelet count 10% of the baseline value in the year preceding diagnosis and/or unexplained fever or excessive sweats

cr

a

b

us

persisting for more than 1 month.

Complex karyotype or sole or 2 abnormalities that include +8, ‒7/7q-, i(17q), ‒5/5q-, 12p-, inv(3), or 11q23

M an

rearrangement.

Ac

ce pt

ed

DIPSS, Dynamic International Prognostic Scoring System; IL, interleukin; RBC, red blood cell; WBC, white blood cell.

36 Page 36 of 40

Figure Legends Fig. 1. Primary myelofibrosis (PMF) pathogenesis. Common overexpression of Janus Kinase 2 (JAK2) may result in additive activation of the Janus kinase–signal transducer

ip t

and activator of transcription (JAK2-STAT) pathway by facilitating increased cell surface expression of hematopoietic growth factor receptors. Somatic mutations in the

cr

thrombopoietin (TPO) receptor gene MPL or in JAK2 (ie, JAK2V617F) result in

us

constitutive activation of the JAK-STAT pathway in hematopoietic stem cells. JAK2V617F may cause epigenetic dysregulation by preventing histone H2A and H4

an

methylation through inactivation of protein arginine methyl transferase 5 or by decreasing chromatin-binding of HP1α, a repressor of heterochromatin activity. Other

M

epigenetic mutations associated with PMF, which may occur antecedent or precedent to

d

the acquisition of the JAK2V617 mutation [39], may affect the status of DNA methylation

te

(TET2), histone H3 methylation (EZH2), or chemical histone modification (ASXL1), resulting in dysregulated transcriptional activity [4,5]. CALR mutations, which are

Ac ce p

mutually exclusive with JAK2 and MPL mutations, appear to be early events in PMF pathogenesis, and, on occasion, have been associated with cytokine-independent STAT5 activation [38].

Fig. 2. Theoretical changes in hematopoietic potential with treatment of primary myelofibrosis (PMF) with rationally designed therapy. Hematopoietic function declines normally with age (gray dotted line), but under normal circumstances this doesn’t not contribute to mortality. Patients with PMF have shortened overall survival associated with progressive decline in normal hematopoiesis (red line), but therapeutic intervention

37 Page 37 of 40

may diminish progression of disease (green lines) to normal physiologic conditions or even lead to the resumption of hematopoiesis expected in age-matched controls (green dotted line). It is hoped that future efforts tailored to earlier intervention with rational

ip t

therapies will delay the accumulation of high-risk genetic lesions and consequent

Ac ce p

te

d

M

an

us

cr

clinical progression.

38 Page 38 of 40

TPO

an

us

cr

ip t

Figure 1

M

TPO TPO

TPO

JAK2

JAK K2 JAK2

JAK2

Ac ce p

JAK2 JAK2

te

d

MPL mutations

JAK2

Regulated activation of JAK-STAT

Overactivation of JAK-STAT

Additive STAT activation

STAT

STAT

STAT

STAT

?

JAK2 JAK2 V617F

JAK1/ JAK2

JAK2V617F activity results in chromatin demethylation and decreased binding of HP1α

STAT STAT

CALR mutations

Myeloproliferation Cell Survival Inflammatory cytokine expression

Epigenetic mutations (eg, TET2, ASXL1, EZH2)

Page 39 of 40

Hypothetical changes in the natural history of PMF with new therapies

Ac ce p

Expected natural history of PMF

te

d

M

an

us

cr

ip t

Figure 2

Hematopoiesis

Asymptomatic Symptomatic

Increasing BM impairment, clonal expansion, and declining performance status

Earlier vs later intervention may improve prognosis

Potential reversal of BM fibrosis and normalization of hematopoiesis?

Page 40 of 40

Increasing Age

Are we altering the natural history of primary myelofibrosis?

Primary myelofibrosis (PMF) is a clonal hematologic malignancy with a variable disease course; survival ranges from months to years. Historically, onl...
5MB Sizes 3 Downloads 5 Views