A JH CME Information: Primary myelofibrosis: 2014 update on diagnosis, risk-stratification and management Author: Ayalew Tefferi M.D. Editor: Carlo Brugnara M.D.

If you wish to receive credit for this activity, please refer to the website: www.wileyhealthlearning.com

䊏 Accreditation and Designation Statement: Blackwell Futura Media Services is accredited by the Accreditation Council for Continuing Medical Education to provide continuing medical education for physicians. Blackwell Futura Media Services designates this journal-based CME for a maximum of 1 AMA PRA Category 1 CreditTM. Physicians should only claim credit commensurate with the extent of their participation in the activity.

䊏 Educational Objectives Upon completion of this educational activity, participants will be better able to identify appropriate management of primary myelofibrosis.

䊏 Activity Disclosures No commercial support has been accepted related to the development or publication of this activity. Author: Ayalew Tefferi, M.D. has no relevant financial relationships to disclose. Editor: Carlo Brugnara, M.D. has no relevant financial relationships to disclose. This activity underwent peer review in line with the standards of editorial integrity and publication ethics maintained by American Journal of Hematology. The peer reviewers have no conflicts of interest to disclose. The peer review process for American Journal of Hematology is single blinded. As such, the identities of the reviewers are not disclosed in line with the standard accepted practices of medical journal peer review. Conflicts of interest have been identified and resolved in accordance with Blackwell Futura Media Services’s Policy on Activity Disclosure and Conflict of Interest. The primary resolution method used was peer review and review by a non-conflicted expert.

䊏 Instructions on Receiving Credit This activity is intended for physicians. For information on applicability and acceptance of continuing medical education credit for this activity, please consult your professional licensing board. This activity is designed to be completed within one hour; physicians should claim only those credits that reflect the time actually spent in the activity. To successfully earn credit, participants must complete the activity during the valid credit period, which is up to two years from initial publication. Additionally, up to 3 attempts and a score of 70% or better is needed to pass the post test. Follow these steps to earn credit:       

Log on to www.wileyhealthlearning.com Read the target audience, educational objectives, and activity disclosures. Read the activity contents in print or online format. Reflect on the activity contents. Access the CME Exam, and choose the best answer to each question. Complete the required evaluation component of the activity. Claim your Certificate.

This activity will be available for CME credit for twelve months following its launch date. At that time, it will be reviewed and potentially updated and extended for an additional twelve months.

C 2014 Wiley Periodicals, Inc. V

doi:10.1002/ajh.00038

American Journal of Hematology, Vol. 89, No. 9, September 2014

915

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

AJH Educational Material

A JH

Primary myelofibrosis: 2014 update on diagnosis, risk-stratification, and management

Ayalew Tefferi* Disease overview: Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by stem cellderived clonal myeloproliferation, abnormal cytokine expression, bone marrow fibrosis, anemia, splenomegaly, extramedullary hematopoiesis (EMH), constitutional symptoms, cachexia, leukemic progression, and shortened survival. Diagnosis: Diagnosis is based on bone marrow morphology. The presence of JAK2, CALR, or MPL mutation is supportive but not essential for diagnosis; approximately 90% of patients carry one of these mutations and 10% are “triple-negative.” None of these mutations are specific to PMF and are also seen in essential thrombocythemia (ET). Prefibrotic PMF mimics ET in its presentation and the distinction, enabled by careful bone marrow morphological examination, is prognostically relevant. Differential diagnosis also includes chronic myeloid leukemia, myelodysplastic syndromes, chronic myelomonocytic leukemia, and acute myeloid leukemia. Risk Stratification: The Dynamic International Prognostic Scoring System-plus (DIPSS-plus) uses eight predictors of inferior survival: age >65 years, hemoglobin 25 3 109/L, circulating blasts 1%, constitutional symptoms, red cell transfusion dependency, platelet count 99th percentile of reference range for age, sex, or altitude of residence or red cell mass > 25% above mean normal predicted or Hgb > 17 g/dL (men)/ > 15 g/dL (women) if associated with a sustained increase of 2 g/dL from baseline that can not be attributed to correction of iron deficiency. c Small to large megakaryocytes with aberrant nuclear/cytoplasmic ratio and hyperchromatic and irregularly folded nuclei and dense clustering. d or in the absence of reticulin fibrosis, the megakaryocyte changes must be accompanied by increased marrow cellularity, granulocytic proliferation, and often decreased erythropoiesis (i.e., pre-fibrotic PMF). BM, bone marrow; Hgb, hemoglobin; Hct, hematocrit; Epo, erythropoietin; EEC, endogenous erythroid colony; WHO, World Health Organization; CML, chronic myelogenous leukemia; PV, polycythemia vera; PMF, primary myelofibrosis; MDS, myelodysplastic syndromes; LDH, lactate dehydrogenase.

TABLE IV. IWG-MRT Recommended Criteria for Post-Polycythemia Vera and Post-Essential Thrombocythemia Myelofibrosis [12] Criteria for post-polycythemia vera myelofibrosis Required criteria: 1 Documentation of a previous diagnosis of polycythemia vera as defined by the WHO criteria (see Table II) 2 Bone marrow fibrosis grade 2–3 (on 0–3 scale) or grade 3–4 (on 0–4 scale) (see footnote for details) Additional criteria (two are required): 1 Anemia or sustained loss of requirement for phlebotomy in the absence of cytoreductive therapy 2 A leukoerythroblastic peripheral blood picture 3 Increasing splenomegaly defined as either an increase in palpable splenomegaly of 5 cm (distance of the tip of the spleen from the left costal margin) or the appearance of a newly palpable splenomegaly 4 Development of 1 of three constitutional symptoms: >10% weight loss in 6 months, night sweats, unexplained fever (>37.5 C) Criteria for post-essential thrombocythemia myelofibrosis Required criteria: 1 Documentation of a previous diagnosis of essential thrombocythemia as defined by the WHO criteria (see Table II) 2 Bone marrow fibrosis grade 2–3 (on 0–3 scale) or grade 3–4 (on 0–4 scale) (see footnote for details) Additional criteria (two are required): 1 Anemia and a 2 g/dL decrease from baseline hemoglobin level 2 A leukoerythroblastic peripheral blood picture 3 Increasing splenomegaly defined as either an increase in palpable splenomegaly of 5 cm (distance of the tip of the spleen from the left costal margin) or the appearance of a newly palpable splenomegaly 4 Increased lactate dehydrogenase 5 Development of 1 of three constitutional symptoms: >10% weight loss in 6 months, night sweats, unexplained fever (>37.5 C) Grade 2–3 according to the European classification [16]: diffuse, often coarse fiber network with no evidence of collagenization (negative trichrome stain) or diffuse, coarse fiber network with areas of collagenization (positive trichrome stain). Grade 3–4 according to the standard classification [17]: diffuse and dense increase in reticulin with extensive intersections, occasionally with only focal bundles of collagen and/or focal osteosclerosis or diffuse and dense increase in reticulin with extensive intersections with coarse bundles of collagen, often associated with significant osteosclerosis.

Most recently, Tefferi et al. studied 254 patients with PMF and reported mutational frequencies of 58% for JAK2, 25% CALR, 8% MPL, and 9% wild-type for all three mutations (i.e., triple-negative) [15]. CALR mutational frequency in JAK2/MPL-unmutated cases was 74%. CALR mutations were associated with younger age, higher platelet count, and lower DIPSS-plus score. CALR-mutated patients were doi:10.1002/ajh.23703

also less likely to be anemic, require transfusions, or display leukocytosis. Spliceosome mutations were infrequent in CALR-mutated patients. In a subsequent international study of 570 patients [43], the authors reported the longest survival in CALR1ASXL12 patients (median 10.4 years) and shortest in CALR2ASXL11 patients (median 2.3 years). CALR1ASXL11 and CALR2ASXL12 patients had similar American Journal of Hematology, Vol. 89, No. 9, September 2014

919

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

Tefferi

Figure 1. DIPSS-plus (dynamic international prognostic scoring system 1 karyotype 1 platelet count 1 transfusion status) risk stratification in 793 patients with primary myelofibrosis seen at Mayo Clinic Rochester (with permission from Gangat et al.) [30] [Color figure can be viewed in the online issue, which is available at wileyonlinelibrary.com.].

survival and were grouped together in an intermediate risk category (median survival 5.8 years). Guglielmelli et al. subsequently demonstrated the additional value of the number of prognostically detrimental mutations [44]. Risk factors for leukemia-free survival include 3% circulating blasts, platelet count 9%, leukocytes 40 3 109/L, or other unfavorable karyotype warrant immediate consideration of (ASCT) (Fig. 2). ASCT is also indicated in the presence of high risk molecular profile (i.e., CALR2/ASXL11 mutational status). Transplant. In considering ASCT as a treatment modality, one should be acutely aware of the risks involved. In one of the largest studies of ASCT in PMF [52], 5-year disease-free survival (DFS) and treatment-related mortality were 33% and 35% for matched related doi:10.1002/ajh.23703

and 27% and 50% for unrelated transplants, respectively. Of note, outcome did not appear to be favorably affected by reduced intensity conditioning (RIC). In another RIC transplant study, 5-year DFS was estimated at 51%; chronic graft-versus-host disease (cGVHD) occurred in 49% of the patients and relapse (29%) was predicted by high-risk disease and prior splenectomy [64]. In the earlier study [52], the respective cGVHD and relapse rates for matched related transplants were 40% and 32% and history of splenectomy did not affect outcome. More recent outcome reports on ASCT in MF were more encouraging: 100-day mortality 13%, a relapse rate of 11%, and a 7-year survival of 61% [65]. There is currently much interest in evaluating the use of JAK inhibitors before transplant with favorable or unfavorable experiences reported by different investigators. I do not currently advise the use of JAK inhibitors in the context of transplant until more information on its value becomes available. Investigational drug therapy. Several experimental drugs have been and are currently being evaluated in PMF, post-PV/ET MF, and other related MPN [66]. These include pomalidomide, JAK inhibiting ATP mimetics, and mTOR inhibitors. Although not further elaborated in the current review, it is important to note that JAK-STAT can be inhibited by many other classes of drugs, which have been evaluated for the treatment of MF and related MPN; these include histone deacetylase inhibitors, such as panobinostat (LBH589) and givinostat (ITF2357) [67, 68]. Pomalidomide. Pomalidomide is a second generation immunomodulatory drug and in a phase-2 randomized study, 25% of patients with anemia responded to the drug used alone (2 mg/day) or in combination with prednisone (0.5 or 2 mg/day) [69]. In a subsequent phase-2 study of single agent pomalidomide (0.5 mg/day) [70], anemia American Journal of Hematology, Vol. 89, No. 9, September 2014

921

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

Tefferi

response was documented only in the presence of JAK2V617F (24% vs. 0%) and predicted by the presence of pomalidomide-induced basophilia (38% vs. 6%) or absence of marked splenomegaly (38% vs. 11%). Platelet response was seen in 58% of patients but the drug had limited activity in reducing spleen size. Drug-associated neuropathy or myelosuppression was infrequent but possible. A phase-1 study did not uncover better activity at higher doses (>2 mg/day), which were instead associated with increased myelosuppression [71]. Most recently, the results of a phase-3 study comparing pomalidomide with placebo did not show significant difference in anemia response, which was approximately 16% for each arm, whereas platelet response was significantly better with pomalidomide [72]. JAK inhibitor ATP mimetics. JAK inhibitor ATP mimetics that have undergone clinical trials in MPN include ruxolitinib (INCB018424), fedratinib (SAR302503), momelotinib (CYT387), lestaurtinib (CEP701), pacritinib (SB1518), AZD1480, BMS911543, LY2784544, and XL019 (clinicalTrials.gov). Results of these studies so far suggest substantial differences among these drugs in their toxicity and efficacy profiles, some of which might be linked to their variable in vitro activity against other JAK and non-JAK kinase targets. For the purposes of this review, I will focus on ruxolitinib (now FDA approved for use in MF), fedratinib (phase-3 study completed but drug withdrawn because of side effects), and momelotinib and pacritinib (phase-3 study ongoing vs. ruxolitinib or best available therapy, respectively). Ruxolitinib is a JAK1/JAK2 inhibitor. The drug was initially evaluated in 153 patients with PMF or post-PV/ET MF, in a phase-1/2 study [73]. DLT was thrombocytopenia and the MTD was either 25 mg twice-daily or 100 mg once-daily. Adverse events included thrombocytopenia, anemia, and a “cytokine rebound reaction” upon drug discontinuation, characterized by acute relapse of symptoms and splenomegaly [51, 74]. Non-hematologic adverse events were infrequent. Grade 3/4 thrombocytopenia or anemia (in transfusionindependent patients at baseline), respectively, occurred in 39% and 43% of patients receiving the drug at 25 or 10 mg twice daily. Among all evaluable patients, 44% experienced 50% decrease in palpable spleen size. Improvement in constitutional symptoms (fatigue, pruritus, abdominal discomfort, early satiety, night sweats, and exercise tolerance) and weight gain were seen in the majority of patients. Four (14%) of twenty-eight transfusion-dependent patients became transfusion-independent. The drug’s effect on JAK2V617F allele burden or bone marrow pathology was negligible but a major reduction in proinflammatory cytokines (e.g., IL-1RA, IL-6, TNF-a, and MIP1b) was documented and coincided with improvement in constitutional symptoms. Two randomized studies comparing ruxolitinib with either placebo or best supportive care have now been published [75, 76]. In the COMFORT-1 trial that compared the drug with placebo (n 5 309) [75], the spleen response rate was approximately 42% for ruxolitinib versus 20%, 9 (39%) had 50% decrease in allele burden. Effect on bone marrow pathology was limited. In general, response was not affected by the presence of JAK2V617F. Most recently, the results of a phase-3 study (n 5 289) comparing fedratinib at two different doses (500 or 400 mg/day) with placebo were disclosed and confirmed the efficacy of the drug in inducing spleen (49%, 47%, and 1%, respectively) and symptom response. However, reports of encephalopathy associated with the use of the drug resulted in the withdrawal of the drug from further development. Other side effects included anemia (grade 3 or 4 in 43–60%), thrombocytopenia (grade 3 or 4 in 17–27%), and diarrhea (56–66%) [79]. Momelotinib (MMB, GS-0387, CYT387) is a JAK1 and JAK2 inhibitor. Among the first 60 patients treated in a phase-1/2 study [80], MTD was 300 mg/day and DLT included grade 3 headache and hyperlipasemia. Anemia and spleen responses were 59% and 48%, respectively. Among 33 patients who were red cell-transfused in the month prior to study entry, 70% achieved a minimum 12-week period without transfusions (range 4.7 to >18.3 months). Most patients experienced constitutional symptoms improvement. Grade 3/ 4 adverse reactions included thrombocytopenia (32%), hyperlipasemia (5%), elevated liver transaminases (3%), and headache (3%). Newonset treatment-related peripheral neuropathy was observed in 22% of patients (sensory symptoms, grade 1). The study was subsequently expanded to include 166 patients treated at either 150 mg or 300 mg once-daily, or 150 mg twice-daily for 9 months, with similar results [81]. The drug is currently undergoing phase-3 study compared to ruxolitinib. Pacritinib (SB1518) is a JAK2/FLT3 inhibitor. In phase-1 studies, myelosuppression was minimal and 400 mg/day was chosen as the recommended dose for phase-2 study, which included 34 patients [82]. The most common treatment-related adverse events were gastrointestinal, especially diarrhea. Spleen response rate was 44% (32% by MRI) and accompanied by symptoms response. Anemia response was reported in 2 (6%) patients. The drug is currently undergoing phase-3 study compared to best available therapy. The above observations demonstrate major differences in toxicity and activity profile among several JAK inhibitor small molecules and underscore the need to evaluate more such drugs before making any conclusions regarding the value of anti-JAK2 therapy in MF or related MPN. It is also becoming evident that some of the salutary doi:10.1002/ajh.23703

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES effects of these drugs might be the result of a potent anti-cytokine activity. mTOR inhibitors. JAK-STAT activation leads to Akt/mTOR activation as well and it is therefore reasonable to evaluate the therapeutic activity of Akt and mTOR inhibitors. In a phase 1/2 study involving the mTOR inhibitor everolimus including 39 MF patients, the commonest toxicity was grade 1–2 stomatitis. A >50% reduction in splenomegaly occurred in 20% of the patients evaluated and the constitutional symptoms response was 69%; 80% experienced complete resolution of pruritus [83]. Drug effect on cytosis or anemia was modest and on JAK2V617F burden negligible. Overall IWG-MRT response rate was 23%. Recommendations: Considering the lack of effective drug therapy in PMF, the risk of transplant-related complications might be justified in those patients in whom median survival is expected to be 20%. These include DIPSS-plus high or intermediate-2 risk patients as well as those with ASXL11/ CALR2 mutational status. Non-transplant candidates are best managed with experimental drug therapy. I have yet to be satisfied by the value of any currently available JAK inhibitor and strongly advise patients to continue participating in clinical trials.

Management of refractory disease and specific disease complications Hydroxyurea-refractory splenomegaly is often managed by splenectomy [84]. Other indications for splenectomy include symptomatic portal hypertension, thrombocytopenia, and frequent red blood cell transfusions. In a recent report of 314 splenectomized patients with MF [85], more than 75% benefited from the procedure and the benefit lasted for a median of 1 year; specific benefits included becoming transfusion-independent and resolution of severe thrombocytopenia. Perioperative complications occurred in 28% of the patients and included infections, abdominal vein thrombosis, and bleeding. Overall perioperative mortality rate was 9%. Approximately 10% of patients

䊏 References 1. Vardiman JW, Thiele J, Arber DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: Rationale and important changes. Blood 2009;114:937–951. Prepublished on 2009/04/10 as DOI 10.1182/blood-2009-03209262. 2. Tefferi A, Vardiman JW. Classification and diagnosis of myeloproliferative neoplasms: The 2008 World Health Organization criteria and point-of-care diagnostic algorithms. Leukemia 2008;22:14–22. 3. Tefferi A. Novel mutations and their functional and clinical relevance in myeloproliferative neoplasms: JAK2, MPL, TET2, ASXL1, CBL, IDH and IKZF1. Leukemia 2010;24:1128–1138. 4. Tefferi A, Thiele J, Vannucchi AM, Barbui T. An overview on CALR and CSF3R mutations and a proposal for revision of WHO diagnostic criteria for myeloproliferative neoplasms. Leukemia 2014. Prepublished on 2014/01/21 as DOI 10.1038/leu.2014.35. 5. Tefferi A, Finke CM, Lasho TL, et al. U2AF1 mutations in primary myelofibrosis are strongly associated with anemia and thrombocytopenia despite clustering with JAK2V617F and normal karyotype. Leukemia 2014;28:431–433. Prepublished on 2013/10/08 as DOI 10.1038/ leu.2013.286. 6. Pardanani A, Lasho TL, Finke C, et al. Prevalence and clinicopathologic correlates of JAK2 exon 12 mutations in JAK2V617F-negative polycythemia vera. Leukemia 2007;21:1960– 1963. 7. Lasho TL, Finke CM, Hanson CA, et al. SF3B1 mutations in primary myelofibrosis: Clinical,

doi:10.1002/ajh.23703

8. 9. 10.

11.

12.

13. 14.

15.

experienced progressive hepatomegaly and 29% thrombocytosis after splenectomy. Median survival after splenectomy was 19 months. Leukemic transformation was documented in 14% of patients whose survival was not different than that of patients without “leukemic transformation,” although others had suggested otherwise [86]. Splenic irradiation (100 cGy in 5–10 fractions) induces transient reduction in spleen size but can be associated with severe pancytopenia [87]. Non-hepatosplenic EMH might involve the vertebral column, lymph nodes, pleura, and peritoneum (ascites) and is effectively treated with low-dose radiotherapy (100–1,000 cGy in 5–10 fractions) [88]. Diagnosis of MF-associated pulmonary hypertension is confirmed by a technetium 99m sulfur colloid scintigraphy and treatment with single-fraction (100 cGy) whole-lung irradiation has been shown to be effective [89]. Single fraction of 100–400 cGy involved field radiotherapy has also been shown to benefit patients with MFassociated extremity pain [90]. Transjugular intrahepatic portosystemic shunt might be considered to alleviate symptoms of portal hypertension. Recent technical advances in the procedure and the introduction of specially coated stents have greatly improved shunt patency and clinical efficacy of TIPS in general. Current TIPS indications include recurrent variceal bleeding and refractory ascites, both of which could accompany advanced MF. The therapeutic value of TIPS has not been systematically studied in MF but relevant information is available from several case reports that confirm feasibility and efficacy [91]. Recommendations: At present, my first-line choice for the management of drug-refractory splenomegaly is participation in experimental drug therapy. Both splenectomy and low-dose radiotherapy are reasonable alternative treatment options. Prophylactic therapy with hydroxyurea is advised to prevent post-splenectomy thrombocytosis [84]. Postsplenectomy thrombosis might be prevented by instituting short term systemic anticoagulation. Laparoscopic splenectomy is not advised in the setting of MF [92] and data on the value of splenic artery embolization are limited [93–95]. I do not believe that splenectomy increases the risk of leukemic transformation [86].

histopathology and genetic correlates among 155 patients. Leukemia 2012;26:1135–1137. Prepublished on 2011/11/09 as DOI 10.1038/ leu.2011.320. Tefferi A. Myelofibrosis with myeloid metaplasia. N Engl J Med 2000;342:1255–1265. Tefferi A. Pathogenesis of myelofibrosis with myeloid metaplasia. J Clin Oncol 2005;23:8520– 8530. Mesa RA, Li CY, Ketterling RP, et al. Leukemic transformation in myelofibrosis with myeloid metaplasia: A single-institution experience with 91 cases. Blood 2005;105:973–977. 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. Barosi G, Mesa RA, Thiele J, et al. Proposed criteria for the diagnosis of post-polycythemia vera and post-essential thrombocythemia myelofibrosis: A consensus statement from the International Working Group for Myelofibrosis Research and Treatment. Leukemia 2008;22: 437–438. Kvasnicka HM, Thiele J. Prodromal myeloproliferative neoplasms: The 2008 WHO classification. Am J Hematol 2010;85:62–69. Hussein K, Van Dyke DL, Tefferi A. Conventional cytogenetics in myelofibrosis: Literature review and discussion. Eur J Haematol 2009;82: 329–338. Tefferi A, Lasho TL, Finke CM, et al. CALR vs JAK2 vs MPL-mutated or triple-negative myelofibrosis: Clinical, cytogenetic and molecular

16.

17. 18.

19.

20. 21.

22.

23.

24.

comparisons. Leukemia 2014. Prepublished on 2014/01/10 as DOI 10.1038/leu.2014.3. Thiele J, Kvasnicka HM, Facchetti F, et al. European consensus on grading bone marrow fibrosis and assessment of cellularity. Haematologica 2005;90:1128–1132. Manoharan A, Horsley R, Pitney WR. The reticulin content of bone marrow in acute leukaemia in adults. Br J Haematol 1979;43:185–190. Tefferi A, Pardanani A. Genetics: CALR mutations and a new diagnostic algorithm for MPN. Nat Rev Clin Oncol 2014. Prepublished on 2014/02/12 as DOI 10.1038/nrclinonc.2014.16. Barbui T, Thiele J, Passamonti F, et al. Survival and disease progression in essential thrombocythemia are significantly influenced by accurate morphologic diagnosis: An international study. J Clin Oncol 2011;29:3179–3184. Prepublished on 2011/07/13 as DOI 10.1200/JCO.2010.34.5298. Steensma DP, Hanson CA, Letendre L, Tefferi A. Myelodysplasia with fibrosis: A distinct entity? Leuk Res 2001;25:829–838. Patnaik MM, Parikh SA, Hanson CA, Tefferi A. Chronic myelomonocytic leukaemia: A concise clinical and pathophysiological review. Br J Haematol 2014. Prepublished on 2014/01/29 as DOI 10.1111/bjh.12756. Orazi A, O’Malley DP, Jiang J, et al. Acute panmyelosis with myelofibrosis: An entity distinct from acute megakaryoblastic leukemia. Mod Pathol 2005;18:603–614. 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. Passamonti F, Cervantes F, Vannucchi AM, et al. A dynamic prognostic model to predict

American Journal of Hematology, Vol. 89, No. 9, September 2014

923

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES

Tefferi

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

37.

38.

924

survival in primary myelofibrosis: A study by the IWG-MRT (International Working Group for Myeloproliferative Neoplasms Research and Treatment). Blood 2010;115:1703–1708. Hussein K, Pardanani AD, Van Dyke DL, et al. International prognostic scoring systemindependent cytogenetic risk categorization in primary myelofibrosis. Blood 2010;115:496–499. Caramazza D, Begna KH, Gangat N, et al. Refined cytogenetic-risk categorization for overall and leukemia-free survival in primary myelofibrosis: A single center study of 433 patients. Leukemia 2011;25:82–88. Prepublished on 2010/ 10/15 as DOI leu2010234 [pii]10.1038/ leu.2010.234. Tefferi A, Siragusa S, Hussein K, et al. Transfusion-dependency at presentation and its acquisition in the first year of diagnosis are both equally detrimental for survival in primary myelofibrosis-prognostic relevance is independent of IPSS or karyotype. Am J Hematol 2009; 85:14–17. Elena C, Passamonti F, Rumi E, et al. Red blood cell transfusion-dependency implies a poor survival in primary myelofibrosis irrespective of International Prognostic Scoring System and Dynamic International Prognostic Scoring System. Haematologica 2011;96:167–170. Patnaik MM, Caramazza D, Gangat N, et al. Age and platelet count are IPSS-independent prognostic factors in young patients with primary myelofibrosis and complement IPSS in predicting very long or very short survival. Eur J Haematol 2010;84:105–108. Gangat N, Caramazza D, Vaidya R, et al. DIPSS plus: A refined dynamic international prognostic scoring system for primary myelofibrosis that incorporates prognostic information from karyotype, platelet count, and transfusion status. J Clin Oncol 2011;29:392–397. Prepublished on 2010/12/15 as DOI JCO.2010.32.2446 [pii] 10.1200/JCO.2010.32.2446. Tefferi A, Jimma T, Gangat N, et al. Predictors of greater than 80% 2-year mortality in primary myelofibrosis: A Mayo Clinic study of 884 karyotypically annotated patients. Blood 2011;118: 4595–4598. Prepublished on 2011/09/02 as DOI 10.1182/blood-2011-08-371096. Tefferi A, Lasho TL, Patnaik MM, et al. JAK2 germline genetic variation affects disease susceptibility in primary myelofibrosis regardless of V617F mutational status: Nullizygosity for the JAK2 46/1 haplotype is associated with inferior survival. Leukemia 2010;24:105–109. Tefferi A, Lasho TL, Huang J, et al. Low JAK2V617F allele burden in primary myelofibrosis, compared to either a higher allele burden or unmutated status, is associated with inferior overall and leukemia-free survival. Leukemia 2008;22:756–761. Guglielmelli P, Barosi G, Specchia G, et al. Identification of patients with poorer survival in primary myelofibrosis based on the burden of JAK2V617F mutated allele. Blood 2009;114: 1477–1483. Tefferi A, Jimma T, Sulai NH, et al. IDH mutations in primary myelofibrosis predict leukemic transformation and shortened survival: Clinical evidence for leukemogenic collaboration with JAK2V617F. Leukemia 2012;26:475–480. Prepublished on 2011/09/ 14 as DOI 10.1038/leu.2011.253. Guglielmelli P, Biamonte F, Score J, et al. EZH2 mutational status predicts poor survival in myelofibrosis. Blood 2011;118:5227–5234. Prepublished on 2011/09/17 as DOI blood-2011-06363424 [pii] 10.1182/blood-2011-06-363424. Lasho TL, Jimma T, Finke CM, et al. SRSF2 mutations in primary myelofibrosis: Significant clustering with IDH mutations and independent association with inferior overall and leukemiafree survival. Blood 2012;120:4168–4171. Prepublished on 2012/09/13 as DOI 10.1182/blood2012-05-429696. Vannucchi AM, Lasho TL, Guglielmelli P, et al. Mutations and prognosis in primary myelofibrosis. Leukemia 2013;27:1861–1869. Prepublished on 2013/04/27 as DOI 10.1038/leu.2013.119.

39. Pardanani A, Guglielmelli P, Lasho TL, et al. Primary myelofibrosis with or without mutant MPL: Comparison of survival and clinical features involving 603 patients. Leukemia 2011;25: 1834–1839. Prepublished on 2011/06/22 as DOI 10.1038/leu.2011.161. 40. Tefferi A, Pardanani A, Lim KH, et al. TET2 mutations and their clinical correlates in polycythemia vera, essential thrombocythemia and myelofibrosis. Leukemia 2009;23:905–911. 41. Pardanani A, Lasho TL, Finke CM, et al. Polyclonal immunoglobulin free light chain levels predict survival in myeloid neoplasms. J Clin Oncol 2012;30:1087–1094. Prepublished on 2012/02/15 as DOI 10.1200/JCO.2011.39.0310. 42. Tefferi A, Vaidya R, Caramazza D, et al. Circulating interleukin (IL)28, IL-2R, IL-12, and IL15 levels are independently prognostic in primary myelofibrosis: A comprehensive cytokine profiling study. J Clin Oncol 2011;29:1356–1363. Prepublished on 2011/02/09 as DOI 10.1200/ JCO.2010.32.9490. 43. Tefferi A, Guglielmelli P, Lasho TL, et al. CALR and ASXL1 mutations-based molecular prognostication in primary myelofibrosis: An international study of 570 patients. Leukemia 2014. Prepublished on 2014/02/06 as DOI 10.1038/ leu.2014.57. 44. Guglielmelli P, Lasho TL, Rotunno G, et al. The number of prognostically detrimental mutations and prognosis in primary myelofibrosis: An international study of 797 patients. Leukemia 2014. Prepublished on 2014/02/20 as DOI 10.1038/leu.2014.76. 45. Tam CS, Kantarjian H, Cortes J, et al. Dynamic model for predicting death within 12 months in patients with primary or post-polycythemia vera/essential thrombocythemia myelofibrosis. J Clin Oncol 2009;27:5587–5593. 46. Huang J, Li CY, Mesa RA, et al. Risk factors for leukemic transformation in patients with primary myelofibrosis. Cancer 2008;112:2726–2732. 47. Passamonti F, Cervantes F, Vannucchi AM, et al. Dynamic International Prognostic Scoring System (DIPSS) predicts progression to acute myeloid leukemia in primary myelofibrosis. Blood 2010;116:2857–2858. 48. Cervantes F, Vannucchi AM, Kiladjian JJ, et al. Three-year efficacy, safety, and survival findings from COMFORT-II, a phase 3 study comparing ruxolitinib with best available therapy for myelofibrosis. Blood 2013;122:4047–4053. Prepublished on 2013/11/01 as DOI 10.1182/blood2013-02-485888. 49. Tefferi A. Challenges facing JAK inhibitor therapy for myeloproliferative neoplasms. N Engl J Med 2012;366:844–846. Prepublished on 2012/ 03/02 as DOI 10.1056/NEJMe1115119. 50. Tefferi A. JAK inhibitors for myeloproliferative neoplasms: Clarifying facts from myths. Blood 2012;119:2721–2730. Prepublished on 2012/01/ 27 as DOI 10.1182/blood-2011-11-395228. 51. Tefferi A, Litzow MR, Pardanani A. Long-term outcome of treatment with ruxolitinib in myelofibrosis. N Engl J Med 2011;365:1455–1457. Prepublished on 2011/10/15 as DOI 10.1056/ NEJMc1109555. 52. Ballen KK, Shrestha S, Sobocinski KA, et al. Outcome of transplantation for myelofibrosis. Biol Blood Marrow Transplant 2010;16:358– 367. 53. Silverstein MN. Agnogenic Myeloid Metaplasia. Acton, MA: Publishing Science Group; 1975. p 126. 54. Cervantes F, Mesa R, Barosi G. New and old treatment modalities in primary myelofibrosis. Cancer J 2007;13:377–383. 55. Elliott MA, Mesa RA, Li CY, et al. Thalidomide treatment in myelofibrosis with myeloid metaplasia. Br J Haematol 2002;117:288–296. 56. Thomas DA, Giles FJ, Albitar M, et al. Thalidomide therapy for myelofibrosis with myeloid metaplasia. Cancer 2006;106:1974–1984. 57. 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.

American Journal of Hematology, Vol. 89, No. 9, September 2014

58. Tefferi A, Cortes J, Verstovsek S, et al. Lenalidomide therapy in myelofibrosis with myeloid metaplasia. Blood 2006;108:1158–1164. 59. Quintas-Cardama A, Kantarjian HM, Manshouri T, et al. Lenalidomide plus prednisone results in durable clinical, histopathologic, and molecular responses in patients with myelofibrosis. J Clin Oncol 2009;27:4760–4766. 60. Huang J, Tefferi A. Erythropoiesis stimulating agents have limited therapeutic activity in transfusion-dependent patients with primary myelofibrosis regardless of serum erythropoietin level. Eur J Haematol 2009;83:154–155. 61. Tefferi A, Lasho TL, Mesa RA, et al. Lenalidomide therapy in del(5)(q31)-associated myelofibrosis: Cytogenetic and JAK2V617F molecular remissions. Leukemia 2007;21:1827–1828. 62. Martinez-Trillos A, Gaya A, Maffioli M, et al. Efficacy and tolerability of hydroxyurea in the treatment of the hyperproliferative manifestations of myelofibrosis: Results in 40 patients. Ann Hematol 2010;89:1233–1237. 63. 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. 64. Kroger N, Holler E, Kobbe G, et al. Allogeneic stem cell transplantation after reduced-intensity conditioning in patients with myelofibrosis: A prospective, multicenter study of the Chronic Leukemia Working Party of the European Group for Blood and Marrow Transplantation. Blood 2009;114:5264–5270. 65. Deeg HJ, Appelbaum FR. Indications for and current results with allogeneic hematopoietic cell transplantation in patients with myelofibrosis. Blood 2011;117:7185. Prepublished on 2011/07/02 as DOI 117/26/7185 [pii]10.1182/blood-2011-04349142. 66. Pardanani A. JAK2 inhibitor therapy in myeloproliferative disorders: Rationale, preclinical studies and ongoing clinical trials. Leukemia 2008;22:23–30. 67. Rambaldi A, Dellacasa CM, Finazzi G, et al. A pilot study of the histone-deacetylase inhibitor Givinostat in patients with JAK2V617F positive chronic myeloproliferative neoplasms. Br J Haematol 2010;150:446–455. 68. DeAngelo DJ, Spencer A, Fischer T, et al. Activity of oral panobinostat (LBH589) in patients with myelofibrosis. ASH Annual Meeting Abstracts 2009;114:2898. 69. Tefferi A, Verstovsek S, Barosi G, et al. Pomalidomide is active in the treatment of anemia associated with myelofibrosis. J Clin Oncol 2009;27:4563–4569. 70. Begna KH, Mesa RA, Pardanani A, et al. A phase-2 trial of low-dose pomalidomide in myelofibrosis. Leukemia 2011;25:301–304. Prepublished on 2010/11/06 as DOI 10.1038/leu.2010.254. 71. Mesa RA, Pardanani AD, Hussein K, et al. Phase1/-2 study of pomalidomide in myelofibrosis. Am J Hematol 2010;85:129–130. Prepublished on 2010/01/07 as DOI 10.1002/ajh.21598. 72. Tefferi A, Passamonti F, Barbui T, et al. Phase 3 study of pomalidomide in myeloproliferative neoplasm (MPN)-associated myelofibrosis with RBCtransfusion-dependence. Blood 2013;122:394. 73. Verstovsek S, Kantarjian H, Mesa RA, et al. Safety and efficacy of INCB018424, a JAK1 and JAK2 inhibitor, in myelofibrosis. N Engl J Med 2010;363:1117–1127. 74. Tefferi A, Pardanani A. Serious adverse events during ruxolitinib treatment discontinuation in patients with myelofibrosis. Mayo Clin Proc 2011;86:1188–1191. Prepublished on 2011/10/29 as DOI 10.4065/mcp.2011.0518. 75. Verstovsek S, Mesa RA, Gotlib J, et al. A double-blind, placebo-controlled trial of ruxolitinib for myelofibrosis. N Engl J Med 2012;366: 799–807. Prepublished on 2012/03/02 as DOI 10.1056/NEJMoa1110557. 76. Harrison C, Kiladjian JJ, Al-Ali HK, et al. JAK inhibition with ruxolitinib versus best available therapy for myelofibrosis. N Engl J Med 2012; 366:787–798. Prepublished on 2012/03/02 as DOI 10.1056/NEJMoa1110556.

doi:10.1002/ajh.23703

ANNUAL CLINICAL UPDATES IN HEMATOLOGICAL MALIGNANCIES 77. Heine A, Brossart P, Wolf D. Ruxolitinib is a potent immunosuppressive compound: Is it time for anti-infective prophylaxis? Blood 2013;122: 3843–3844. Prepublished on 2013/11/30 as DOI 10.1182/blood-2013-10-531103. 78. Pardanani A, Gotlib JR, Jamieson C, et al. Safety and efficacy of TG101348, a selective JAK2 inhibitor, in myelofibrosis. J Clin Oncol 2011;29: 789–796. Prepublished on 2011/01/12 as DOI 10.1200/JCO.2010.32.8021. 79. Pardanani A, Harrison CN, Cortes JE, et al. Results of a randomized, double-blind, placebo-controlled phase III study (JAKARTA) of the JAK2-selective inhibitor fedratinib (SAR302503) in patients with myelofibrosis (MF). Blood 2013;122:393. 80. Pardanani A, Laborde RR, Lasho TL, et al. Safety and efficacy of CYT387, a JAK1 and JAK2 inhibitor, in myelofibrosis. Leukemia 2013;27:1322–1327. Prepublished on 2013/03/06 as DOI 10.1038/leu.2013.71. 81. Pardanani A, Gotlib J, Gupta V, et al. Update on the long-term efficacy and safety of momelotinib, a JAK1 and JAK2 inhibitor, for the treatment of myelofibrosis. Blood 2013;122:108. 82. Komrokji RS, Wadleigh M, Seymour JF, et al. Results of a phase 2 study of pacritinib (SB1518), a novel oral JAK2 inhibitor, in patients with primary, post-polycythemia vera, and post-essential thrombocythemia myelofibrosis. ASH Annual Meeting Abstracts 2011;118:282. 83. Guglielmelli P, Barosi G, Rambaldi A, et al. Safety and efficacy of everolimus, a mTOR inhibitor, as single agent in a phase 1/2 study in patients with myelofibrosis. Blood 2011;118:

doi:10.1002/ajh.23703

84.

85.

86.

87.

88.

89.

90.

2069–2076. Prepublished on 2011/07/05 as DOI 10.1182/blood-2011-01-330563. Tefferi A, Mesa RA, Nagorney DM, et al. Splenectomy in myelofibrosis with myeloid metaplasia: A single-institution experience with 223 patients. Blood 2000;95:2226–2233. Prepublished on 2000/03/25. Mesa RA, Nagorney DS, Schwager S, et al. 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. Barosi G, Ambrosetti A, Centra A, et al. Splenectomy and risk of blast transformation in myelofibrosis with myeloid metaplasia. Italian Cooperative Study Group on Myeloid with Myeloid Metaplasia. Blood 1998;91:3630–3636. 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. Prepublished on 1998/11/25. Koch CA, Li CY, Mesa RA, Tefferi A. Nonhepatosplenic extramedullary hematopoiesis: Associated diseases, pathology, clinical course, and treatment. Mayo Clin Proc 2003;78:1223–1233. Prepublished on 2003/10/09 as DOI 10.4065/78.10.1223. 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. Prepublished on 2002/08/16. Neben-Wittich MA, Brown PD, Tefferi A. Successful treatment of severe extremity pain in

91.

92.

93.

94.

95.

myelofibrosis with low-dose single-fraction radiation therapy. Am J Hematol 2010;85:808–810. Prepublished on 2010/08/28 as DOI 10.1002/ ajh.21819. Mishchenko E, Tefferi A. Treatment options for hydroxyurea-refractory disease complications in myeloproliferative neoplasms: JAK2 inhibitors, radiotherapy, splenectomy and transjugular intrahepatic portosystemic shunt. Eur J Haematol 2010;85:192–199. Prepublished on 2010/06/ 10 as DOI 10.1111/j.1600-0609.2010.01480.x. Feldman LS, Demyttenaere SV, Polyhronopoulos GN, Fried GM. Refining the selection criteria for laparoscopic versus open splenectomy for splenomegaly. J Laparoendosc Adv Surg Tech A 2008;18:13–19. Iwase K, Higaki J, Mikata S, et al. Laparoscopically assisted splenectomy following preoperative splenic artery embolization using contour emboli for myelofibrosis with massive splenomegaly. Surg Laparosc Endosc Percutan Tech 1999;9:197–202. Hiatt JR, Gomes AS, Machleder HI. Massive splenomegaly. Superior results with a combined endovascular and operative approach. Arch Surg 1990;125:1363–1367. Hocking WG, Machleder HI, Golde DW. Splenic artery embolization prior to splenectomy in end-stage polycythemia vera. Am J Hematol 1980;8:123–127.

American Journal of Hematology, Vol. 89, No. 9, September 2014

925

Primary myelofibrosis: 2014 update on diagnosis, risk-stratification, and management.

Primary myelofibrosis (PMF) is a myeloproliferative neoplasm characterized by stem cell-derived clonal myeloproliferation, abnormal cytokine expressio...
853KB Sizes 0 Downloads 6 Views