Managing patients with myelofibrosis (MF), either those with primary MF or those whose MF has evolved from antecedent polycythemia vera or essential thrombocythemia, presents many challenges to the hematologist. MF patients have a range of debilitating disease manifestations (eg, massive splenomegaly, cytopenias, constitutional symptoms, and transformation to a treatment-refractory blast phase). Cure is potentially achievable through allogeneic stem cell transplantation; however, this therapy is either inappropriate or not feasible for the majority of patients. Therefore, remaining therapies are palliative but can be of significant value to some MF patients. In particular, management of symptomatic splenomegaly remains one of the most perplexing aspects of MF clinical care. Using medications is the simplest approach for reducing splenomegaly, yet achieving symptomatic response without undue myelosuppression is challenging. Splenectomy or radiotherapy offers benefit, but careful patient selection and close monitoring are required because both have the potential for dangerous adverse effects. Experimental medical therapies, such as JAK2 inhibitors, show promise and may soon play an important role in the management of symptomatic splenomegaly in MF patients. Future care of MF patients, including splenomegaly management, will continue to require the hematologist to select therapeutic options carefully in the context of realistic, achievable goals.

Myelofibrosis (MF) includes primary MF and MF that has arisen from an antecedent polycythemia vera or essential thrombocythemia.1  MF can affect patients in various ways, including progressive splenomegaly, cytopenias (particularly anemia), and troublesome constitutional symptoms (ie, night sweats, fevers, unintentional weight loss, debilitating fatigue).2  MF can also result in blastic transformation3  and death.4 

Splenomegaly can be a major independent source of morbidity and detriment to quality of life when it develops in MF patients.5,6  First, splenomegaly (sometimes massive, > 10 kg) can lead to pain, early satiety, bloating, or even portal hypertension.7  Second, the bulk of the spleen can result in areas of ischemia and painful episodes of splenic infarction. Finally, splenomegaly can result in development (or exacerbation) of cytopenias from splenic sequestration.

Management of MF-associated splenomegaly can be difficult, and the questions that arise represent a microcosm of the management issues for the entire condition. What are the treatment goals for an individual patient: cure or palliation? Who should receive an allogeneic stem cell transplantation? What are the benefits of current medical therapies? How do I manage the myelosuppression brought about by medical therapy? Which patients should be considered for splenectomy or splenic radiation? What is the impact of experimental drugs (particularly the JAK2 inhibitors) on splenomegaly, and would my patient benefit from treatment with one of these agents? These are the key challenges for hematologists involved in treating patients with MF (they also comprise a large proportion of the patient-related inquiries in my e-mail inbox), and each of these questions is addressed herein.

Deciding on a treatment plan for patients with MF requires both an assessment of the disease impact at diagnosis and an accurate assessment of prognosis. The International Working Group for Myelofibrosis Research and Treatment recently published an International Prognostic Scoring System (IPSS-MF; Table 1) to aid in assessing MF prognosis. The IPSS-MF defines 4 risk groups with projected survival medians for each group, ranging from 27 months for high-risk disease to 135 months for low-risk disease.4 

Table 1

The International Working Group for Myelofibrosis Research and Treatment (IWG-MRT) International Prognostic Scoring System for Primary Myelofibrosis (IPSS-PMF)4 

Risk groupNo. of risk factors*Median survival, mo (95% CI)
Low 135 (117-181) 
Intermediate 1 95 (79-114) 
Intermediate 2 48 (43-59) 
High ≥ 3 27 (23-31) 
Risk groupNo. of risk factors*Median survival, mo (95% CI)
Low 135 (117-181) 
Intermediate 1 95 (79-114) 
Intermediate 2 48 (43-59) 
High ≥ 3 27 (23-31) 
*

Risk factors for primary myelofibrosis (present at diagnosis): (1) age > 65 years, (2) hemoglobin < 10 g/dL, (3) leukocytes > 25 × 109/L, (4) peripheral blood blasts ≥ 1%, and (5) constitutional symptoms (weight loss > 10%, night sweats, or fevers).

Currently, there is only one curative therapy for MF: allogeneic stem cell transplantation, which offers a 58% 3-year survival rate with a 32% relapse-free mortality rate.8  More favorable outcomes have been reported with reduced-intensity transplantation (85% survival rate at a median of 31 months), but long-term outcomes, including the burden of chronic graft-versus-host disease, need to be assessed.9  Therefore, the key question after MF diagnosis is whether the patient is a suitable candidate for allogeneic transplantation (Figure 1). Given that no other therapy has been found curative or has been proved to confer a survival advantage in MF, all other nontransplantation forms of therapy must be considered palliative in nature and intent.

Figure 1

Management algorithm for symptomatic splenomegaly in MF. Illustration by Debra T. Dartez.

Figure 1

Management algorithm for symptomatic splenomegaly in MF. Illustration by Debra T. Dartez.

Close modal

The first decision made in managing splenomegaly in MF patients is whether the patient has sufficient symptoms (ie, pain, mechanical symptoms, splenic infarcts, or refractory cytopenias) to merit therapy (Figure 1). Splenomegaly and cytopenias are sometimes present concurrently (ie, splenic sequestration exacerbates ineffective hematopoiesis from the marrow). Frequently, however, splenomegaly may exist with adequate peripheral blood counts in those patients whose marrow remains very proliferative. The presence or absence of concurrent cytopenias is essential in the decision-making process because many therapies have a detrimental impact on any cytopenias present. Nontransplantation (ie, palliative) therapeutic options are judged by their efficacy in achieving symptom relief balanced against toxicity. Assessing the patient's performance status and peripheral blood counts to determine the ability to tolerate the potential myelosuppression of medical therapy is central in therapy selection (Figure 1).

Medical therapy is the cornerstone therapeutic option for symptomatic splenomegaly in patients with MF (Table 2). In MF, sequestration of immature circulating myeloid progenitors leads to accumulation and proliferation in the splenic cords.19  Current medical therapies can ameliorate splenomegaly in MF mainly through nonspecific myelosuppression, thereby decreasing the immature circulating myeloid pool accumulating in the spleen.

Table 2

Medical therapy for symptomatic splenomegaly in myelofibrosis (available medications)

AgentAdministration routeDose/scheduleResponse rate*Additional efficacyToxicitiesReference
Tier 1       
    Hydroxyurea Oral 500-3000 mg daily 40%-50% None Myelosuppression, skin ulcers 10  
    Busulfan Oral 2-4 mg daily Variable None Myelosuppression, leukemia 11  
    Melphalan Oral 2.5 mg 3 times weekly 67% None Myelosuppression, leukemia 12  
    Interferon-2α Subcutaneous 0.5-1.0 × 106 units 3 times weekly Limited data; 75% with small spleens, less with larger None Myelosuppression, depression 13  
    Thalidomide with or without prednisone taper Oral 50 mg daily 19% Anemia, thrombocytopenia Neuropathy, sedation, myeloproliferation 14  
    Lenalidomide with or without prednisone taper Oral 5-10 mg daily 33% Anemia, thrombocytopenia Myelosuppression, rash, diarrhea 15  
Tier 2       
    Cladribine (2-CdA) Intravenous 5 mg/m2 per day × 5 days per month 56% None Myelosuppression 16  
    Daunorubicin Intravenous 60 mg/m2 on days 1-3 NA None Myelosuppression, cardiotoxicity None 
    5-Azacytidine Subcutaneous 75 mg/m2 on days 1-7 per month 21% Anemia Myelosuppression, gastrointestinal 17,18  
    Decitabine Subcutaneous 20 mg/m2 on days 1-5 per month NA  Myelosuppression None 
AgentAdministration routeDose/scheduleResponse rate*Additional efficacyToxicitiesReference
Tier 1       
    Hydroxyurea Oral 500-3000 mg daily 40%-50% None Myelosuppression, skin ulcers 10  
    Busulfan Oral 2-4 mg daily Variable None Myelosuppression, leukemia 11  
    Melphalan Oral 2.5 mg 3 times weekly 67% None Myelosuppression, leukemia 12  
    Interferon-2α Subcutaneous 0.5-1.0 × 106 units 3 times weekly Limited data; 75% with small spleens, less with larger None Myelosuppression, depression 13  
    Thalidomide with or without prednisone taper Oral 50 mg daily 19% Anemia, thrombocytopenia Neuropathy, sedation, myeloproliferation 14  
    Lenalidomide with or without prednisone taper Oral 5-10 mg daily 33% Anemia, thrombocytopenia Myelosuppression, rash, diarrhea 15  
Tier 2       
    Cladribine (2-CdA) Intravenous 5 mg/m2 per day × 5 days per month 56% None Myelosuppression 16  
    Daunorubicin Intravenous 60 mg/m2 on days 1-3 NA None Myelosuppression, cardiotoxicity None 
    5-Azacytidine Subcutaneous 75 mg/m2 on days 1-7 per month 21% Anemia Myelosuppression, gastrointestinal 17,18  
    Decitabine Subcutaneous 20 mg/m2 on days 1-5 per month NA  Myelosuppression None 

NA indicates not applicable.

When considering current medical therapies for splenomegaly in MF patients, I categorize them into 2 distinct tiers. Tier 1 consists predominantly of oral agents with (usually) modest toxicity; tier 2 includes more aggressive approaches suitable for patients with severe splenomegaly or involvement of other organs by extramedullary hematopoiesis (EMH; eg, lungs, abdominal cavity, pericardium). In those patients, a more intensive, frequently parenteral (intravenous or subcutaneous), myelosuppressive approach may be warranted. The choice of both initial and subsequent therapies should be based on toxicity profile and specifically the ability to withstand myelosuppression (Figure 2).

Figure 2

Medical therapy algorithm for symptomatic splenomegaly in MF. Illustration by Debra T. Dartez.

Figure 2

Medical therapy algorithm for symptomatic splenomegaly in MF. Illustration by Debra T. Dartez.

Close modal

Tier 1

Hydroxyurea.

Hydroxyurea, the most common initial medical therapy used to decrease splenomegaly in MF patients (estimated response rate of < 50% for splenomegaly, although very little prospective clinical trial data exist),10  is an oral myelosuppressive agent. There are, however, several limitations to the use of this agent in MF. First, hydroxyurea rarely induces a complete resolution of splenomegaly or even an International Working Group for Myelofibrosis Research and Treatment clinical improvement for splenomegaly (ie, > 50% improvement sustained for at least 2 months).20  Nevertheless, more modest reductions in splenomegaly may benefit some patients with MF. A second limitation is that splenomegaly, compared with thrombocytosis, is less responsive to hydroxyurea and might require a higher dose (ie, 2-3 g/day). Third, hydroxyurea therapy may potentially exacerbate cytopenias, particularly at higher doses. Responses to hydroxyurea for splenomegaly usually occur within 2 to 3 months, and the dose must be titrated to achieve the benefit for splenomegaly by lowering the leukocyte count as best tolerated or to the lower limits of the normal range.

Oral alkylators.

Alkylators, such as melphalan and busulfan, can alleviate splenomegaly in some MF patients. However, myelosuppression and the increased risk of blastic transformation are potential adverse effects for both medications. In one study,12  melphalan (2.5 mg orally 3 times per week) reduced spleen size in 66% of patients; however, 26% of the study cohort developed acute leukemia. Busulfan can be used to treat splenomegaly,21  although it was used more classically in the treatment of chronic myeloid leukemia in the pre-imatinib era.22 

Immunomodulatory drugs.

The immunomodulatory drugs known collectively as IMiDs (ie, thalidomide, lenalidomide, pomalidomide) inhibit cytokines and have antiangiogenic properties. IMiDs have been used primarily for palliation in MF patients with cytopenias (anemia and thrombocytopenia) but may also be used to reduce splenomegaly. This combination of potential effects should lead to consideration of IMiD therapy for patients with concurrent cytopenias and splenomegaly.

Thalidomide produced responses for cytopenias in MF patients but was not well tolerated.23  Low-dose (50 mg/day) thalidomide combined with a prednisone taper (thalidomide-prednisone regimen)14  achieves significant responses for anemia (67%), thrombocytopenia (75%), and splenomegaly (33%) in MF patients and was better tolerated than single-agent thalidomide. Subsequently, lenalidomide (a second-generation IMiD) was evaluated in 68 patients with symptomatic MF, achieving overall response rates of 22% for anemia, 33% for splenomegaly, and 50% for thrombocytopenia.15  Lenalidomide was effective in patients where prior treatment with thalidomide had failed and, given its more significant myelosuppressive properties, was better tolerated if patients' pretreatment peripheral blood neutrophil and platelet counts were adequate. In addition, as in del(5q) myelodysplastic syndromes,24  MF patients with an abnormality of chromosome 5 seem to respond best to lenalinomide.25 

Currently, I recommend thalidomide-prednisone as first-line treatment for MF patients with cytopenias and splenomegaly, with lenalidomide as a second-line treatment (first-line treatment if a del5[q] is present in the marrow karyotype).

Interferon-α.

Therapy with interferon-α (IFN-α) has been used in MF patients based on its cytoreductive properties and has been effective in patients with polycythemia vera.26  However, clinical trials of IFN-α in MF with both standard preparations27  and pegylated IFN-α (PEG-IFN-α)28  have demonstrated poor patient tolerance and negligible response rates. In one study,28  subcutaneous PEG-IFN-α-2b (PEG-intron) was administered weekly to 36 patients with BCR-ABL-negative myeloproliferative neoplasms, and none of the patients with MF responded. Therefore, I do not recommend use of IFN-α in advanced MF at present.

Tier 2

Cladribine.

Palliative benefit from the purine nucleoside analog 2-chlorodeoxyadenosine (2-CdA) has been reported in MF patients.16  2-CdA has been administered as 4 to 6 once-monthly cycles of treatment with either 0.1 mg/kg per day intravenously by continuous infusion for 7 days or 5 mg/m2 intravenously over 2 hours for 5 consecutive days. In the Mayo Clinic experience, we observed responses in 55%, 50%, 55%, and 40% of patients for splenomegaly, thrombocytosis, leukocytosis, and anemia, respectively.16  Responses were frequently durable and lasted for a median of 6 months after discontinuation of treatment. Myelosuppression is the primary toxicity of this regimen, with occasional additional adverse effects of gastrointestinal disturbances. I think that 2-CdA is the most helpful of the tier 2 agents and should be given on monthly cycles (up to 4 cycles) to achieve maximal response for refractory splenomegaly.

Hypomethylating agents.

The 2 hypomethylating agents approved for myelodysplastic syndromes, azacytidine and decitabine, have been tested in MF patients with a goal of improving cytopenias and splenomegaly or delaying blastic transformation. Recent trials of azacytidine (75 mg/m2 per day for 5 or 7 days)17,18  showed a 21% response rate for splenomegaly in MF using the 7-day regimen only.17  The lack of success for the 5-day regimen in the trial leads me to suggest the use of only the 7-day regimen at this time. Decitabine use in MF is in the early stages of testing, and trials are under way. The frequent visits required for administration and myelosuppression, the most common adverse effect, limit the use of these therapies in MF patients.

Low-intensity therapy for acute leukemia.

A range of myelosuppressive therapies most typically used for acute leukemia or higher-risk myelodysplastic syndromes have been used anecdotally to treat MF patients with refractory splenomegaly. Low-dose subcutaneous cytarabine and daunorubicin administered as a single agent (60 mg/m2 per day for 3 days) are included in this group. These strategies, however, have not been vetted through structured clinical trials, and I use them only in difficult cases in which clinical trial participation is not an option.

Therapeutic splenectomy in MF

The experience with splenectomy in MF patients dates back to the beginning of the 20th century (Table 3).29  Progressive surgical series demonstrated a significant but slowly decreasing (because of improvements in surgical technique, antimicrobials, and patient selection) perioperative mortality rate. Although splenectomy can be helpful for improving symptoms in MF patients, it seems to have no clear effect on patient survival, disease course, or intramedullary manifestations of the disease.

Table 3

Indications and outcomes for elective splenectomy in myelofibrosis: Mayo Clinic experience29 

VariableResult
Preoperative  
    Surgical indications  
        Anemia 78 (25%) 
        Symptomatic splenomegaly 156 (49%) 
        Portal hypertension 47 (15%) 
        Severe thrombocytopenia 33 (11%) 
    Median intervals, mo (range)  
        Diagnosis to splenectomy (n = 314) 26.3 (0-389) 
        Follow-up period from diagnosis (n = 314) 54.6 (0.6-487) 
        Follow-up period from splenectomy (n = 314) 18.6 (0.2-151) 
Perioperative  
    Median spleen mass, g (range) 2700 (380-11 750) 
        Complications, no. (%) 87 (27.7) 
            Bleeding 44 (14.0) 
            Thrombosis 31 (9.9) 
            Infection 31 (9.9) 
        Fatal complications, no. (%) 21 (6.7) 
            Infection 8 (2.6) 
            Hemorrhage 7 (2.2) 
            Thrombosis 2 (0.6) 
            Other 5 (1.6) 
        Median hospital stay, d (range) 9 (1-63) 
        Laboratory values at discharge, median (range)  
            Hemoglobin, g/dL 10 (7-16.3) 
            White blood cell count, ×109/L 21.2 (1.2-217) 
            Platelet count, ×109/L 256 (10-1968) 
            Patients (n, %) with > 450 × 109/L 90 (28.7%) 
            Patients (n, %) with > 1000 × 109/L 17 (5.4%) 
Postsplenectomy outcomes  
    Leukemic transformation 45 (14.3%) 
    Accelerated hepatomegaly 32 (10.2%) 
    Long-term improvements  
        Symptomatic splenomegaly 121/156 (48.8%) 
        Anemia 39/78 (50.0%) 
        Portal hypertension 19/47 (40.4%) 
    Severe thrombocytopenia 10/33 (30.3%) 
Survival after splenectomy (by indication)  
    Overall 19 mo (95% CI, 14-22) 
        Anemia 70.1% (at 1 y) 
        Symptomatic splenomegaly 66% (at 1 y) 
        Portal hypertension 59.5% (at 1 y) 
        Severe thrombocytopenia 66.7% (at 1 y) 
VariableResult
Preoperative  
    Surgical indications  
        Anemia 78 (25%) 
        Symptomatic splenomegaly 156 (49%) 
        Portal hypertension 47 (15%) 
        Severe thrombocytopenia 33 (11%) 
    Median intervals, mo (range)  
        Diagnosis to splenectomy (n = 314) 26.3 (0-389) 
        Follow-up period from diagnosis (n = 314) 54.6 (0.6-487) 
        Follow-up period from splenectomy (n = 314) 18.6 (0.2-151) 
Perioperative  
    Median spleen mass, g (range) 2700 (380-11 750) 
        Complications, no. (%) 87 (27.7) 
            Bleeding 44 (14.0) 
            Thrombosis 31 (9.9) 
            Infection 31 (9.9) 
        Fatal complications, no. (%) 21 (6.7) 
            Infection 8 (2.6) 
            Hemorrhage 7 (2.2) 
            Thrombosis 2 (0.6) 
            Other 5 (1.6) 
        Median hospital stay, d (range) 9 (1-63) 
        Laboratory values at discharge, median (range)  
            Hemoglobin, g/dL 10 (7-16.3) 
            White blood cell count, ×109/L 21.2 (1.2-217) 
            Platelet count, ×109/L 256 (10-1968) 
            Patients (n, %) with > 450 × 109/L 90 (28.7%) 
            Patients (n, %) with > 1000 × 109/L 17 (5.4%) 
Postsplenectomy outcomes  
    Leukemic transformation 45 (14.3%) 
    Accelerated hepatomegaly 32 (10.2%) 
    Long-term improvements  
        Symptomatic splenomegaly 121/156 (48.8%) 
        Anemia 39/78 (50.0%) 
        Portal hypertension 19/47 (40.4%) 
    Severe thrombocytopenia 10/33 (30.3%) 
Survival after splenectomy (by indication)  
    Overall 19 mo (95% CI, 14-22) 
        Anemia 70.1% (at 1 y) 
        Symptomatic splenomegaly 66% (at 1 y) 
        Portal hypertension 59.5% (at 1 y) 
        Severe thrombocytopenia 66.7% (at 1 y) 

CI indicates confidence interval.

Current outcomes.

We recently analyzed 3 decades of experience with palliative splenectomy in MF at the Mayo Clinic30,31  to determine whether better control of postsplenectomy thrombocytosis and modern operative techniques and supportive care have diminished morbidity and mortality. Patients and outcomes are summarized in Table 2.32  Although meaningful improvement in symptoms was observed in 30% to 50% of patients, complication rates (27.7% overall, 6.7% fatal) are sobering and indicate that candidates for surgery should be chosen carefully and monitored closely in the perioperative period.

Candidates for splenectomy.

Given the potential dangers of splenectomy, the procedure should be considered only for patients who meet several criteria: substantial and refractory splenic symptoms (ie, requiring narcotics or significantly interfering with quality of life); status as adequate surgical candidates without decompensated coagulopathy or significant comorbidities; treated unsuccessfully with at least one medical therapy for splenomegaly (if eligible and able to enroll into a JAK2 inhibitor trial, “JAK2 inhibitors,” this should be considered); and have an adequate performance status and life expectancy (ie, > 1 year) to consider this surgical procedure. I would consider patients meeting these criteria with refractory cytopenias candidates for splenectomy, but they should be aware that the cytopenias may not improve. In addition, many of the constitutional symptoms of the disease (ie, fever, night sweats, and fatigue) are not linked to splenomegaly; therefore, improvement in these symptoms should not be expected. Splenectomy preceding allogeneic stem cell transplant speeds engraftment but without a significant improvement in outcomes; given the risks outlined in Table 3, routine pretransplantation splenectomy is not recommended.33 

Perioperative management.

The major risk with splenectomy remains perioperative thrombosis and hemorrhage; given that fact, our practice at Mayo Clinic has evolved to aggressively control postoperative platelet counts. Indeed, in our analysis, perioperative thrombohemorrhagic complications decreased in the last decade because we use platelet apheresis as necessary and promptly administer cytoreductive agents to counteract postsplenectomy thrombocytosis.32  I now use hydroxyurea to treat patients with postoperative platelet counts rising above 300 × 109/L and plateletpheresis to treat those with counts exceeding 450 × 109/L, within postoperative weeks 1 and 2, although these guidelines have not been vetted by a randomized trial.

Radiotherapy for MF

Radiotherapy has several uses in MF (Table 4). EMH is exquisitely sensitive to external beam radiotherapy in patients with MF, regardless of location. In patients with MF, the lungs41  (where EMH can contribute to pulmonary hypertension), paraspinal masses,6  and spleen are frequently irradiated sites. Splenomegaly can be treated with external beam radiotherapy for palliation, but the benefit is typically transient and myelosuppression can be severe.

Table 4

Published experience with splenic radiotherapy for myelofibrosis

ReferencePatients, nDose, cGyFractionsResponse/comments
Silverstein34  39 NR NR 63% response rate 
Greenberger et al35  14 600 95% response rate; significant cytopenias in 57% 
Slanina et al36  25 NR NR 88% with symptomatic decrease in splenomegaly 
Wagner et al37  10 150-300 6-10 Responses in 90%; cytopenias common 
Elliott et al38  23 277 7.5 93.9% decrease in spleen size; median duration of response 6 months; toxicities: cytopenias, complicated subsequent splenectomy 
Bouabdallah et al39  15 980 22 59% response rate; toxicities: cytopenias 
McFarland et al40  450 All patients with a palliative response; toxicities: cytopenias 
ReferencePatients, nDose, cGyFractionsResponse/comments
Silverstein34  39 NR NR 63% response rate 
Greenberger et al35  14 600 95% response rate; significant cytopenias in 57% 
Slanina et al36  25 NR NR 88% with symptomatic decrease in splenomegaly 
Wagner et al37  10 150-300 6-10 Responses in 90%; cytopenias common 
Elliott et al38  23 277 7.5 93.9% decrease in spleen size; median duration of response 6 months; toxicities: cytopenias, complicated subsequent splenectomy 
Bouabdallah et al39  15 980 22 59% response rate; toxicities: cytopenias 
McFarland et al40  450 All patients with a palliative response; toxicities: cytopenias 

NR indicates not reported.

Candidates for splenic radiotherapy.

Splenic radiation is effective for palliating MF-associated splenomegaly, but the effects are transient. In addition, the abscopal effects of the radiotherapy can lead to significant myelosuppression and profound thrombocytopenia. Splenic radiotherapy is of greatest utility for patients with considerable symptoms and an adequate platelet count who, because of age or comorbidities, probably would not undergo splenectomy in the future.

Reported outcomes for splenic radiotherapy.

Several reports (Table 3) have described the palliative benefit of external beam radiotherapy for improving symptomatic splenomegaly in MF.38,39  The Mayo Clinic experience38  included a group of 23 MF patients (who received a total of 49 courses of radiotherapy) who received a median radiation course of 277 cGy in a median of 8 fractions. An objective decrease in spleen size was noted after 94% of radiotherapy courses; however, 44% of patients experienced posttreatment cytopenias (26% were severe, 13% fatal). Splenic radiation also seemed to increase morbidity and mortality of subsequent splenectomy when undertaken. This latter effect seems related to the development of splenic adhesions to the abdominal wall and surrounding viscera, leading to greater complexity with subsequent attempts at surgical extirpation. In addition, in irradiated areas, delayed hemorrhage was common, resulting in blunt dissection of the spleen away from other structures.

JAK2 inhibitors

The discovery of several key myeloproliferative disorder-associated mutations has broadened the therapeutic horizons forMF significantly (Table 5).42  Discovery of the JAK2V617F mutation in the 14th exon of JAK2 was followed by the 10 mutations currently described in the 12th exon of JAK2. In addition, 5 constitutively activating mutations thus far have identified in the thrombopoietin receptor, MPL, which signals through JAK2. All of these mutations seem to feed into a final common pathway of cellular activation through the PI3 kinase pathway, the STAT pathway, and the MAP kinase pathway.42 

Table 5

Experimental therapies for myelofibrosis with reported efficacy for symptomatic splenomegaly

AnemiaSplenomegalyConstitutional symptomsPruritusReference
INCB018424 < 10% 43 
CEP-701 < 10% — 47 
XL019 — 46 
TG101348 — 45 
ITF2357 — 48 
AnemiaSplenomegalyConstitutional symptomsPruritusReference
INCB018424 < 10% 43 
CEP-701 < 10% — 47 
XL019 — 46 
TG101348 — 45 
ITF2357 — 48 

— indicates not applicable; and X, instance.

The best-studied clinical experience for a JAK2 inhibitor is for INCB018424 (Incyte, Wilmington, DE), which is selective against JAK1 and JAK2; this agent leads to significant reduction in splenomegaly and dramatic improvement in constitutional symptoms43  with toxicities of thrombocytopenia and anemia43  (Table 5). In addition, the adverse increase in inflammatory cytokines observed in myelofibrosis seem to improve with INCB018424 therapy and correlate with symptom improvement.44  Other drugs currently being tested include TG101348, a selective JAK2 inhibitor (TarGen, San Francisco, CA)45 ; XL019, a selective JAK2 inhibitor (Exelexis, San Francisco, CA)46 ; CEP-701, a tyrosine kinase inhibitor of JAK2 and FLT3 (Cephalon, Frazer, PA)47 ; and ITF2357, a histone deacetylase inhibitor (Italfarmaco, Milan, Italy).48  With each agent, preliminary results also report improvements in splenomegaly and symptoms in MF patients (Table 4), although various toxicities seen (ie, gastrointestinal, neuropathy; XL019) all share the potential for causing anemia and/or thrombocytopenia. To date, no JAK2 inhibitor has reported a significant ability to improve cytopenias, fibrosis, or histologic changes associated with MF. It should be mentioned that the JAK2 inhibitor trials are all in the preliminary stages of reporting (ie, abstracts from the last American Society of Hematology meeting), with all trials remaining active and results not published in manuscript form at this time.

Patients with symptomatic splenomegaly who qualify for and have access to a JAK2 inhibitor trial should be considered for enrollment. This is particularly the case for MF patients without baseline anemia or thrombocytopenia, who have constitutional symptoms.

Current management of symptomatic splenomegaly in MF patients requires thoughtful consideration of prognosis and treatment goals. For those who are not candidates for transplantation, the palliative benefit of a specific therapeutic choice must be balanced against its potential risks. For those treating MF patients, the JAK2 inhibitors have brought great excitement because of their targeted approach. Preliminary data indicate a meaningful response for MF patients in terms of symptomatic splenomegaly. However, although the JAK2 inhibitors have provided a valuable and incremental benefit over existing options, particularly for symptoms and quality of life, they have not yet demonstrated a significant impact for anemia or more advanced disease features. Future advances for MF patients may require further vetting of current and future JAK2 inhibitors, trials of JAK2 inhibitors combined with other agents (ie, such as an IMiD against anemia), or further advances in our understanding of the pathogenesis of the disorder to open additional avenues of targeted therapies.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 USC section 1734.

The author acknowledges the longstanding collaboration and mentorship of Dr Ayalew Tefferi and the collaboration of his colleagues at Mayo Clinic during the author's 18 years there in the care and clinical investigation of patients with myelofibrosis.

Contribution: R.A.M. wrote the manuscript.

Conflict-of-interest disclosure: The author declares no competing financial interests.

Correspondence: Ruben A. Mesa, Division of Hematology-Oncology, Mayo Clinic Arizona, 13400 E Shea Blvd, Scottsdale, AZ 85259; e-mail: mesa.ruben@mayo.edu.

1
Tefferi
A
Thiele
J
Orazi
A
et al
Proposals and rationale for revision of the World Health Organization diagnostic criteria for polycythemia vera, essential thrombocythemia, and primary myelofibrosis: recommendations from an ad hoc international expert panel.
Blood
2007
110
1092
1097
2
Mesa
RA
Niblack
J
Wadleigh
M
et al
The 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
3
Mesa
RA
Li
CY
Ketterling
RP
Schroeder
GS
Knudson
RA
Tefferi
A
Leukemic transformation in myelofibrosis with myeloid metaplasia: a single-institution experience with 91 cases.
Blood
2005
105
973
977
4
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
2901
5
Reilly
JT
Idiopathic myelofibrosis: pathogenesis, natural history and management.
Blood Rev
1997
11
233
242
6
Koch
CA
Li
CY
Mesa
RA
Tefferi
A
Nonhepatosplenic extramedullary hematopoiesis: associated diseases, pathology, clinical course, and treatment.
Mayo Clinic Proc
2003
78
1223
1233
7
Anger
BR
Seifried
E
Scheppach
J
Heimpel
H
Budd-Chiari syndrome and thrombosis of other abdominal vessels in the chronic myeloproliferative diseases.
Klin Wochenschr
1989
67
818
825
8
Deeg
HJ
Gooley
TA
Flowers
ME
et al
Allogeneic hematopoietic stem cell transplantation for myelofibrosis.
Blood
2003
102
3912
3918
9
Rondelli
D
Barosi
G
Bacigalupo
A
et al
Allogeneic hematopoietic stem cell transplantation with reduced intensity conditioning in intermediate or high risk patients with myelofibrosis with myeloid metaplasia.
Blood
2005
105
4115
4119
10
Lofvenberg
E
Wahlin
A
Management of polycythaemia vera, essential thrombocythaemia and myelofibrosis with hydroxyurea.
Eur J Haematol
1988
41
375
381
11
Chang
JC
Gross
HM
Remission of chronic idiopathic myelofibrosis to busulfan treatment.
Am J Med Sci
1988
295
472
476
12
Petti
MC
Latagliata
R
Spadea
T
et al
Melphalan treatment in patients with myelofibrosis with myeloid metaplasia.
Br J Haematol
2002
116
576
581
13
Levy
B
Vandris
K
Adriano
F
Goldman
J
Silver
RT
Recombinant interferon alpha (rIFNα) may retard progression of early primary myelofibrosis (PM) by reducing splenomegaly and by changing marrow morphology.
ASH Annual Meeting Abstracts
2008
112
1758
14
Mesa
RA
Steensma
DP
Pardanani
A
et al
A phase 2 trial of combination low-dose thalidomide and prednisone for the treatment of myelofibrosis with myeloid metaplasia.
Blood
2003
101
2534
2541
15
Tefferi
A
Cortes
J
Verstovsek
S
et al
Lenalidomide therapy in myelofibrosis with myeloid metaplasia.
Blood
2006
108
1158
1164
16
Faoro
LN
Tefferi
A
Mesa
RA
Long-term analysis of the palliative benefit of 2-chlorodeoxyadenosine for myelofibrosis with myeloid metaplasia.
Eur J Haematol
2005
74
117
120
17
Quintás-Cardama
A
Tong
W
Kantarjian
H
et al
A phase II study of 5-azacitidine for patients with primary and post essential thrombocythemia/polcythemia vera myelofibrosis.
Leukemia
2008
22
965
970
18
Mesa
RA
Verstovsek
S
Rivera
C
et al
5-Azacitidine has limited therapeutic activity in myelofibrosis.
Leukemia
2009
2
26
[Epub ahead of print]
19
Zhang
B
Lewis
SM
The splenomegaly of myeloproliferative and lymphoproliferative disorders: splenic cellularity and vascularity.
Eur J Haematol
1989
43
63
66
20
Tefferi
A
Barosi
G
Mesa
RA
et al
International Working Group (IWG) consensus criteria for treatment response in myelofibrosis with myeloid metaplasia, for the IWG for Myelofibrosis Research and Treatment (IWG-MRT).
Blood
2006
108
1497
1503
21
Silver
RT
Jenkins
DE
Jr
Engle
RL
Jr
Use of testosterone and busulfan in the treatment of myelofibrosis with myeloid metaplasia.
Blood
1964
23
341
353
22
Silver
RT
Woolf
SH
Hehlmann
R
et al
An evidence-based analysis of the effect of busulfan, hydroxyurea, interferon, and allogeneic bone marrow transplantation in treating the chronic phase of chronic myeloid leukemia: developed for the American Society of Hematology.
Blood
1999
94
1517
1536
23
Barosi
G
Grossi
A
Comotti
B
Musto
P
Gamba
G
Marchetti
M
Safety and efficacy of thalidomide in patients with myelofibrosis with myeloid metaplasia.
Br J Haematol
2001
114
78
83
24
List
A
Kurtin
S
Roe
DJ
et al
Efficacy of lenalidomide in myelodysplastic syndromes.
N Engl J Med
2005
352
549
557
25
Tefferi
A
Lasho
TL
Mesa
RA
Pardanani
A
Ketterling
RP
Hanson
CA
Lenalidomide therapy in del(5)(q31)-associated myelofibrosis: cytogenetic and JAK2V617F molecular remissions.
Leukemia
2007
21
1827
1828
26
Kiladjian
JJ
Cassinat
B
Chevret
S
et al
Pegylated interferon-alfa-2a induces complete hematologic and molecular responses with low toxicity in polycythemia vera.
Blood
2008
112
3065
3072
27
Tefferi
A
Elliot
MA
Yoon
SY
et al
Clinical and bone marrow effects of interferon alfa therapy in myelofibrosis with myeloid metaplasia.
Blood
2001
97
1896
28
Verstovsek
S
Lawhorn
K
Giles
F
et al
PEG-intron for myeloproliferative diseases: an update of ongoing phase II study
Blood
2004
104
Abstract 633
29
Hickling
RA
Splenectomy in myeloid metaplasia.
Q J Med
1937
30
253
30
Silverstein
MN
ReMine
WH
Sex, splenectomy, and myeloid metaplasia.
JAMA
1974
227
424
426
31
Tefferi
A
Mesa
RA
Nagorney
DM
Schroeder
G
Silverstein
MN
Splenectomy in myelofibrosis with myeloid metaplasia: a single-institution experience with 223 patients.
Blood
2000
95
2226
2233
32
Mesa
RA
Nagorney
DS
Schwager
S
Allred
J
Tefferi
A
Palliative goals, patient selection, and perioperative platelet management: outcomes and lessons from 3 decades of splenectomy for myelofibrosis with myeloid metaplasia at the Mayo Clinic.
Cancer
2006
107
361
370
33
Li
Z
Gooley
T
Applebaum
FR
Deeg
HJ
Splenectomy and hemopoietic stem cell transplantation for myelofibrosis.
Blood
2001
97
2180
2181
34
Silverstein
MN
Control of hypersplenism and painful splenomegaly in myeloid metaplasia by irradiation.
Int J Radiat Oncol Biol Phys
1977
2
1221
1222
35
Greenberger
JS
Chaffey
JT
Rosenthal
DS
Moloney
WC
Irradiation for control of hypersplenism and painful splenomegaly in myeloid metaplasia.
Int J Radiat Oncol Biol Phys
1977
2
1083
1090
36
Slanina
J
Vondraczek
A
Wannenmacher
M
Symptomatische Milzbestrahlung bei fortgeschrittener Osteomyelosklerose.
Dtsch Med Wochenschr
1986
111
1144
1150
37
Wagner
H
Jr
McKeough
PG
Desforges
J
Madoc-Jones
H
Splenic irradiation in the treatment of patients with chronic myelogenous leukemia or myelofibrosis with myeloid metaplasia: results of daily and intermittent fractionation with and without concomitant hydroxyurea.
Cancer
1986
58
1204
1207
38
Elliott
MA
Chen
MG
Silverstein
MN
Tefferi
A
Splenic irradiation for symptomatic splenomegaly associated with myelofibrosis with myeloid metaplasia.
Br J Haematol
1998
103
505
511
39
Bouabdallah
R
Coso
D
Gonzague-Casabianca
L
Alzieu
C
Resbeut
M
Gastaut
JA
Safety and efficacy of splenic irradiation in the treatment of patients with idiopathic myelofibrosis: a report on 15 patients.
Leuk Res
2000
24
491
495
40
McFarland
JT
Kuzma
C
Millard
FE
Johnstone
PA
Palliative irradiation of the spleen.
Am J Clin Oncol
2003
26
178
183
41
Steensma
DP
Hook
CC
Stafford
SL
Tefferi
A
Low-dose, single-fraction, whole-lung radiotherapy for pulmonary hypertension associated with myelofibrosis with myeloid metaplasia.
Br J Haematol
2002
118
813
816
42
Pardanani
A
JAK2 inhibitor therapy in myeloproliferative disorders: rationale, preclinical studies and ongoing clinical trials.
Leukemia
2008
22
23
30
43
Verstovsek
S
Kantarjian
HM
Pardanani
AD
et al
The JAK inhibitor, INCB018424, demonstrates durable and marked clinical responses in primary myelofibrosis (PMF) and post-polycythemia/essential thrombocythemia myelofibrosis (post PV/ETMF).
Blood
2008
112
1762
44
Tefferi
A
Kantarjian
HM
Pardanani
AD
et al
The clinical phenotype of myelofibrosis encompasses a chronic inflammatory state that is favorably altered by INCB018424, a selective inhibitor of JAK1/2.
Blood
2008
112
2804
45
Pardanani
AD
Gotlib
J
Jamieson
C
et al
A phase I study of TG101348, an orally bioavailable JAK2-selective inhibitor, in patients with myelofibrosis.
Blood
2008
112
97
46
Shah
NP
Olszynski
P
Sokol
L
et al
A phase I study of XL019m a selective JAK2 inhibitor, in patients with primary myelofibrosis, post-polycythemia vera, or post-essential thrombocythemia myelofibrosis [abstract].
Blood
2008
112
Abstract 98
47
Verstovsek
S
Tefferi
A
Kornblau
S
et al
Phase II study of CEP701, an orally available JAK2 inhibitor, in patients with primary myelofibrosis and post polycythemia vera/essential thrombocythemia myelofibrosis [abstract].
Blood
2007
110
Abstract 3543
48
Rambaldi
A
Dellacasa
CM
Salmoiraghi
S
et al
A phase 2A study of the histone-deacetylase inhibitor ITF2357 in patients with Jak2V617F positive chronic myeloproliferative neoplasms.
Blood
2008
112
100
Sign in via your Institution