Interferon in polycythemia vera and related

0 downloads 0 Views 264KB Size Report
May 21, 2015 - hydroxyurea (HU), can only be determined by ... Expert Review of Hematology Downloaded from informahealthcare.com by 2.104.105.187 on ...
Expert Review of Hematology Downloaded from informahealthcare.com by 2.104.105.187 on 05/21/15 For personal use only.

Perspective

Interferon in polycythemia vera and related neoplasms. Can it become the treatment of choice without a randomized trial? Expert Rev. Hematol. Early online, 1–7 (2015)

Hans Carl Hasselbalch*1 and Richard T Silver2 1 Department of Hematology, Roskilde Hospital, University of Copenhagen, Roskilde, Denmark 2 Myeloproliferative Disease Center, Division of Hematology-Oncology, Weill Cornell Medical Center, New York, USA *Author for correspondence: [email protected]

Recently, it was concluded that the optimal therapy for essential thrombocythemia and polycythemia vera, either recombinant interferon alpha (rIFNa) or hydroxyurea can only be determined by the completion of a randomized clinical trial. We present our recommendations for the use of rIFNa for those patients who are not candidates for the randomized trial. We argue for rethinking the approach whether we should continue to wait for the results from a randomized trial before recommending treatment with rIFNa for those unable and unwilling to enter these trials. The interferon story shows that clinical experience may be an alternative path to follow when making treatment decisions and recommendations in orphan diseases. KEYWORDS: clinical experience . essential thrombocythemia . hydroxyurea . interferon . polycythemia vera .

randomized trial

An Editorial/Perspective article recently published in Haematologica concluded that the optimal therapy for essential thrombocythemia (ET) and polycythemia vera (PV), either recombinant interferon alpha (rIFNa) or hydroxyurea (HU), can only be determined by the completion of a randomized clinical trial [1]. On behalf of the many hundreds or thousands of patients worldwide not receiving treatment with rIFNa because their physicians are awaiting the results from this randomized trial, we present our recommendations based on decades of experience for the use of rIFNa for those patients with ET and PV who are not candidates for this randomized trial. This issue is most relevant and timely to discuss now when the use of rIFNa for the treatment of these neoplasms is increasing significantly in several centers in some parts of the world, whereas at other myeloproliferative neoplasms (MPN) centers of excellence it is not recommended as first-line therapy [2,3]. Since the ultimate goal as physicians is to serve our MPN patients according to the most effective and safe therapy available, the unmet need informahealthcare.com

10.1586/17474086.2015.1045409

for a large proportion of MPN patients to access rIFNa is discussed from a patient perspective as well. Discussion

Recombinant IFNa was used efficaciously for the first time in the treatment of PV and ET about 25 years ago [4,5]. Since then, its efficacy and safety have been confirmed in a large number of non-randomized studies [6–18], which have been reviewed recently, highlighting the fact that rIFNa is likely to become the drug of choice for the treatment of patients with ET, PV and hyperproliferative myelofibrosis [19–24]. In this context, it has been argued that when used from the time of diagnosis, when there is ‘minimal disease’ and the tumor burden is at a minimum, interferon treatment may have the best chance of a successful outcome [20–22,24,25] with subsequent normalization of the marrow in a subset of patients with PV [26,27]. In PV, there is, therefore, a steady decrease in high WBC and platelet counts and establishment of a phlebotomy free state within weeks or months.

 2015 Informa UK Ltd

ISSN 1747-4086

1

Expert Review of Hematology Downloaded from informahealthcare.com by 2.104.105.187 on 05/21/15 For personal use only.

Perspective

Hasselbalch & Silver

Similar to the decline in the JAK2V617F-allele burden in patients with PV and ET [10–15,18], treatment with interferon is associated with a decline in the CALR mutation in patients with ET likewise treated [28]. All cancers are characterized by increasing genomic instability, subclone formation, resistance to treatment and ultimately metastasis. Although the evidence for increasing genomic instability from early cancer stage (ET/PV) to the advanced cancer stage – myelofibrosis – has to be definitely established (as assessed by rate of accumulation of new mutations and transformation to acute myeloid leukemia (AML) in a large series of patients from early cancer stage (ET/PV) to advanced cancer stage (myelofibrosis/AML)), it is reasonable to assume that the MPNs do not differ substantially from any other cancer in regard to the biology of an untreated cancer – disease progression due to clonal evolution with increasing genomic instability, subclone evolution through the acquisition of additional driver mutations, resistance to treatment and metastasis. Thus, in MPNs, disease progression and leukemic transformation has been shown to be associated with clonal evolution, subclonal mutations (ASXL1, SRSF2, CBL, IDH1/IDH2, TP53 and SRSF2) being independently associated with leukemic transformation and poor survival. Although the mutation rate is low, it has been shown that the presence of two or more somatic mutations (the presence of at least a subclonal mutation) significantly reduces overall survival and increases the risk for leukemic transformation in patients with MPNs [29]. Since MPNs and AML transformation are associated with elevated biomarkers of chronic inflammation [30–37], we speculate if chronic inflammation with increased oxidative stress in the bone marrow and in the circulation might be involved in MPN pathogenesis and in MPN-associated complications and comorbidities [38–40]. In this regard, chronic inflammation may also have a great impact upon mutagenesis and the mutational load in MPNs [39]. The identification of additional mutations already at the time of diagnosis does not argue against this concept of chronic inflammation as a driving force for mutagenesis, since most recent studies have shown that many patients with MPNs have had their disease for several years before MPN diagnosis [41]. Considering that the JAK2V617F mutation induces accumulation of reactive oxygen species (ROS) [42] and ROS in several other cancers induce genomic instability (DNA damage), there is compelling evidence that chronic inflammation with excessive ROS may be harmful for the stem cell compartment in MPNs. Since interferon – with no alternatives – has been shown in several studies to reduce the JAK2V617F mutation, which is an inductor of ROS accumulation [42], a marker of tumor burden and a risk marker of thrombosis [43], it is reasonable to assume that this beneficial effect may be partly related to disruption of ROS accumulation in the bone marrow although several other mechanisms are likely operative – all to be explored in future studies. Until results from such studies are available, current evidence of a link between chronic inflammation, genomic instability and mutational load doi: 10.1586/17474086.2015.1045409

remains weak, but supported by most recent observations that MPNs are associated with massive deregulation of oxidative stress genes and antioxidative defense genes [44] – likely mediated and driven by chronic inflammation [38,39,44]. Conceptually, it is reasonable to treat the MPNs early, since patients with severe myelofibrosis and marked splenomegaly (considered analogous to the ‘metastatic stage’) do not respond to therapy with rIFNa. The beneficial effects of rIFNa in ET and PV are noteworthy. In PV and ET, the decline in the JAK2-allele burden is physiologically important since the JAK2V617F allele appears to be a thrombosis promoter per se [43] and is perhaps a tumor promoter as well [45]. Furthermore, the JAK2V617F mutation negatively regulates p53 stabilization [46], thereby increasing genomic instability [47] predisposing to additional mutations, subclone formation, resistance to treatment and ultimately leukemic and myelofibrotic transformation. This evolution may depict the biological continuum from early to an advanced ‘cancer stage’ [48], in which chronic inflammation is likely a very important driver of clonal evolution and the development of second cancers as well [36,38,40]. Although rIFNa may induce molecular remissions, it is important to underscore that the molecular heterogeneity of MPN is complex, implying variability of molecular responses to rIFNa. Thus, TET2 clones persist in some patients during treatment with rIFNa [49] and additional clones may impair the response to rIFNa as well [50]. Regarding the differential response to interferon between JAK2V617F-positive ET and PV patients, this may be partly explained by differential interferon signaling and STAT1 phosphorylation observed in vitro in erythroid ET and PV cells [51], implying increased STAT1 activity to be associated with an ET-like phenotype and downregulation of STAT1 activity in JAK2V617F-heterozygous ET progenitors to be associated with a PV-like phenotype. Accordingly, the consequences of JAK2V617F are determined by the balance between STAT5 and STAT1 activation [51]. For those advocating delayed therapy, a risk stratification system is followed, allowing patients with low-risk ET (10 years [68,69], which might in part explain the lack of evidence for HU as leukemogenic [68,69]. An additional argument for the lack of leukemogenicity of HU is the often-cited observation that no increased risk for leukemia has been observed in sickle cell anemia patients treated with HU [1]. However, sickle cell anemia is not a stem cell disease, whereas MPNs, therefore, are more likely to develop genomic instability, when being exposed to an agent which itself impairs DNA-repair mechanisms. In Denmark, a multicenter study was launched in 2012 enrolling newly diagnosed patients with ET, PV and hyperproliferative myelofibrosis in a randomized study, comparing lowdose Pegasys to PegIntron in patients 5 years) is associated with deep molecular remissions (5 years) from the time of diagnosis.

doi: 10.1586/17474086.2015.1045409

Expert Rev. Hematol.

Interferon in PV & related neoplasms

patients with advanced essential thrombocythemia and polycythemia vera. J Clin Oncol 2009;27:5418-24

References Papers of special note have been highlighted as: . of interest .. of considerable interest 1.

Expert Review of Hematology Downloaded from informahealthcare.com by 2.104.105.187 on 05/21/15 For personal use only.

2.

Mascarenhas J, Mesa R, Prchal J, et al. Optimal therapy for polycythemia vera and essential thrombocythemia can only be determined by the completion of randomized clinical trials. Haematologica 2014;99(6):945-9 Barbui T, Barosi G, Birgegard G, et al. Philadelphia-negative classical myeloproliferative neoplasms: critical concepts and management recommendations from European Leukemia Net. J Clin Oncol 2011;29(6):761-70

3.

Tefferi A. Polycythemia vera and essential thrombocythemia: 2013 update on diagnosis, risk-stratification, and management. Am J Hematol 2013;88(6): 507-16

4.

Linkesch W, Gisslinger H, Ludwig H, et al. Therapy with interferon (recombinant IFNalpha-2C) in myeloproliferative diseases with severe thrombocytoses [in German]. Acta Med Austriaca 1985;12:123-7

5.

Silver RT. Recombinant interferon-alpha for treatment of polycythaemia vera. Lancet 1988;2(8607):403

6.

Silver RT. Interferon alfa: effects of long-term treatment for polycythemia vera. Semin Hematol 1997;34(1):40-50

7.

Gilbert HS. Long term treatment of myeloproliferative disease with interferon alpha-2b: Feasibility and efficacy. Cancer 1998;83:1205-13

8.

9.

10.

11.

12.

Silver RT. Long-term effects of the treatment of polycythemia vera with recombinant interferon-alpha. Cancer 2006;107:451-8 Samuelsson J, Hasselbalch H, Bruserud O, et al. A phase II trial of pegylated interferon alpha-2b therapy for polycythemia vera and essential thrombocythemia: Feasibility, clinical and biologic effects, and impact on quality of life. Cancer 2006;106:2397-405 Kiladjian JJ, Cassinat B, Turlure P, et al. High molecular response rate of polycythemia vera patients treated with pegylated interferon alpha-2a. Blood 2006; 108(6):2037-40 Jabbour E, Kantarjian H, Cortes J, et al. PEG-IFN-alpha-2b therapy in BCR-ABL-negative myeloproliferative disorders: Final results of a phase 2 study. Cancer 2007;110:2012-18 Quintas- Cardama A, Kantarjian H, et al. Pegylated interferon alfa-2a yields high rates of hematologic and molecular response in

informahealthcare.com

13.

..

14.

15.

16.

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;112:3065-72 An important trial describing the high-response rates in patients with PV treated with IFN-a2. Gowin K, Thapaliya P, Samuelson J, et al. Experience with pegylated interferon a-2a in advanced myeloproliferative neoplasms in an international cohort of 118 patients. Haematologica 2012;97(10):1570-3 Larsen TS, Iversen KF, Hansen E, et al. Long term molecular responses in a cohort of Danish patients with essential thrombocythemia, polycythemia vera and myelofibrosis treated with recombinant interferon alpha. Leuk Res 2013;37(9): 1041-5 Silver RT, Vandris K, Goldman JJ. Recombinant interferon-a may retard progression of early primary myelofibrosis: a preliminary report. Blood 2011;117(24): 6669-72

17.

Ianotto JC, Boyer-Perrard F, Gyan E, et al. Efficacy and safety of pegylated-interferon a-2a in myelofibrosis: a study by the FIM and GEM French cooperative groups. Br J Haematol 2013(6):783-91

18.

Quinta´s-Cardama A, Abdel-Wahab O, Manshouri T, et al. Molecular analysis of patients with polycythemia vera or essential thrombocythemia receiving pegylated interferon a-2a. Blood 2013;122(6): 893-901

19.

20.

Kiladjian JJ, Chomienne C, Fenaux P. Interferon-alpha therapy in bcr-ablnegative myeloproliferative neoplasms. Leukemia 2008;22:1990-8 Hasselbalch HC, Larsen TS, Riley CH, et al. Interferon-alpha in the treatment of Philadelphia-negative chronic myeloproliferative neoplasms: Status and perspectives. Curr Drug Targets 2011;12: 392-419

21.

Hasselbalch HC. A new era for IFN-a in the treatment of Philadelphia-negative chronic myeloproliferative neoplasms. Expert Rev Hematol 2011;4(6):637-55

22.

Hasselbalch HC, Kiladjian JJ, Silver RT. Interferon alfa in the treatment of Philadelphia-negative chronic myeloproliferative neoplasms. J Clin Oncol 2011;29(18):e564-5

Perspective

23.

Kiladjian JJ, Mesa RA, Hoffman R. The renaissance of interferon therapy for the treatment of myeloid malignancies. Blood 2011;117(18):4706-15

24.

Silver RT, Kiladjian JJ, Hasselbalch HC. Interferon and the treatment of polycythemia vera, essential thrombocythemia and myelofibrosis. Expert Rev Hematol 2013;6(1):49-58

..

This article is a comprehensive review describing the promising efficacy of recombinant IFN-a in the treatment of patients with myeloproliferative neoplasms (MPNs).

25.

Hasselbalch HC. Myelofibrosis with myeloid metaplasia: The advanced phase of an untreated disseminated hematological cancer—Time to change our therapeutic attitude with early upfront treatment? Leuk Res 2009;33:11-18

26.

Larsen TS, Bjerrum OW, Pallisgaard N, et al. Sustained major molecular response on interferon alpha-2b in two patients with polycythemia vera. Ann Hematol 2008; 87(10):847-50

27.

Larsen TS, Møller MB, de Stricker K, et al. Minimal residual disease and normalization of the bone marrow after long-term treatment with alpha-interferon2b in polycythemia vera: A report on molecular response patterns in seven patients in sustained complete hematological remission. Hematology 2009;14:331-4

28.

Cassinat B, Verger E, Kiladjian JJ. Interferon alfa therapy in CALR-mutated essential thrombocythemia. N Engl J Med 2014;371(2):188-9

29.

Lundberg P, Karow A, Nienhold R, et al. Clonal evolution and clinical correlates of somatic mutations in myeloproliferative neoplasms.Blood. 2014;123(14):2220-8

30.

Barbui T, Carobbio A, Finazzi G, et al. Inflammation and thrombosis in essential thrombocythemia and polycythemia vera: different role of C-reactive protein and pentraxin 3. Haematologica 2011;96(2): 315-18

31.

Tefferi A, Vaidya R, Caramazza D, et al. 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(10):1356-63

32.

Vaidya R, Gangat N, Jimma T, et al. Plasma cytokines in polycythemia vera: phenotypic correlates, prognostic relevance, and comparison with myelofibrosis. Am J Hematol 2012;87(11):1003-5

doi: 10.1586/17474086.2015.1045409

Perspective 33.

Expert Review of Hematology Downloaded from informahealthcare.com by 2.104.105.187 on 05/21/15 For personal use only.

34.

35.

Hasselbalch & Silver

Skov V, Thomassen M, Riley CH, et al. Gene expression profiling with principal component analysis depicts the biological continuum from essential thrombocythemia over polycythemia vera to myelofibrosis. Exp Hematol 2012;40(9):771-80 Skov V, Larsen TS, Thomassen M, et al. Molecular profiling of peripheral blood cells from patients with polycythemia vera and related neoplasms: identification of deregulated genes of significance for inflammation and immune surveillance. Leuk Res 2012;36(11):1387-92

Hasselbalch HC. The role of cytokines in the initiation and progression of myelofibrosis. Cytokine Growth Factor Rev 2013;24(2):133-45

37.

Bjørn ME, Andersen CL, Jensen MK, et al. Circulating YKL-40 in myelofibrosis a potential novel biomarker of disease activity and the inflammatory state. Eur J Haematol 2014;93(3):224-8

39.

Vannucchi AM. JAK2 mutation and thrombosis in the myeloproliferative neoplasms Curr Hematol Malig Rep. 2010; 5(1):22-8

54.

Balci K, Utku U, Asil T, et al. Deep cerebral vein thrombosis associated with iron deficiency anaemia in adults. J Clin Neurosci 2007;14(2):181-4

44.

Hasselbalch HC, Thomassen M, Riley CH, et al. Whole blood transcriptional profiling reveals deregulation of oxidative and antioxidative defence genes in myelofibrosis and related neoplasms. Potential implications of downregulation of Nrf2 for genomic instability and disease progression. PLoS One 2014;9(11):e112786

55.

Kiladjian JJ, Rain JD, Bernard JF, et al. Long-term incidence of hematological evolution in three French prospective studies of hydroxyurea and pipobroman in polycythemia vera and essential thrombocythemia. Semin Thromb Hemost 2006;32:417-21

56.

45.

Nielsen C, Birgens HS, Nordestgaard BG, et al. Diagnostic value of JAK2 V617F somatic mutation for myeloproliferative cancer in 49 488 individuals from the general population. Br J Haematol 2013; 160(1):70-9

Kiladjian JJ, Chevret S, Dosquet C, et al. Treatment of polycythemia vera with hydroxyurea and pipobroman: final results of a randomized trial initiated in 1980. J Clin Oncol 2011;29(29):3907-13

..

This important study analyzed the results from the French Polycythemia Study. Group investigating the use of HU versus pipobroman as first-line therapy in patients with PV.

57.

Spivak JL, Hasselbalch H. Hydroxycarbamide: a user’s guide for chronic myeloproliferative disorders. Expert Rev Anticancer Ther 2011;11(3):403-14

58.

Latagliata R, Spadea A, Cedrone M, et al. Symptomatic mucocutaneous toxicity of hydroxyurea in Philadelphia chromosome-negative myeloproliferative neoplasms: the Mister Hyde face of a safe drug. Cancer 2012;118(2):404-9

59.

Este`ve E, Georgescu V, Heitzmann P, et al. Multiple skin and mouth squamous cell carcinomas related to long-term treatment with hydroxyurea. Ann Dermatol Venereol 2001;128(8-9):919-21

60.

BC Cancer Agency Cancer Drug Manual Developed: Hydroxyurea. September 1994 Revised: December 2006, 1 October 2013

61.

Kissova J, Ovesna P, Penka M, et al. Second malignancies in Philadelphia-negative myeloproliferative neoplasms-single-center experience. Anticancer Res 2014;34(5):2489-96

62.

Frederiksen H, Farkas DK, Christiansen CF, et al. Chronic myeloproliferative neoplasms and subsequent cancer risk: a Danish population-based cohort study. Blood 2011;118:6515-20

63.

Pettersson H, Knutsen H, Holmberg E, et al. Increased incidence of another cancer in myeloproliferative neoplasms patients at the time of diagnosis. Eur J Haematol 2014; Epub ahead of print

64.

Hasselbalch HC. Editorial: Perspectives on the increased risk of second cancer in patients with essential thrombocythemia,

Barbui T, Carobbio A, Finazzi G, et al. Elevated C-reactive protein is associated with shortened leukemia-free survival in patients with myelofibrosis. Leukemia 2013; 27(10):2084-6

36.

38.

43.

Hasselbalch HC. Perspectives on chronic inflammation in essential thrombocythemia, polycythemia vera, and myelofibrosis: is chronic inflammation a trigger and driver of clonal evolution and development of accelerated atherosclerosis and second cancer? Blood 2012;119(14):3219-25 Hasselbalch HC. Chronic inflammation as a promotor of mutagenesis in essential thrombocythemia,polycythemia vera and myelofibrosis. A human inflammation model for cancer development? Leuk Res 2013;37(2):214-20

46.

47.

48.

49.

50.

Nakatake M, Monte-Mor B, Debili N, et al. JAK2(V617F) negatively regulates p53 stabilization by enhancing MDM2 via La expression in myeloproliferative neoplasms. Oncogene 2012;31(10):1323-33 Plo I, Nakatake M, Malivert L, et al. JAK2 stimulates homologous recombination and genetic instability: potential implication in the heterogeneity of myeloproliferative disorders. Blood 2008;112(4):1402-12 Nielsen C, Bojesen SE, Nordestgaard BG, et al. JAK2V617F somatic mutation in the general population: myeloproliferative neoplasm development and progression rate. Haematologica 2014;99(9):1448-55 Kiladjian JJ, Masse A, Cassinat B, et al. Clonal analysis of erythroid progenitors suggests that pegylated interferon alpha-2a treatment targets JAK2V617F clones without affecting TET2 mutant cells. Leukemia 2010;24(8):1519-23 Quinta´s-Cardama A, Abdel-Wahab O, Manshouri T, et al. Molecular analysis of patients with polycythemia vera or essential thrombocythemia receiving pegylated interferon a-2a. Blood 2013;122(6): 893-901

40.

Hasselbalch HC. Perspectives on the impact of JAK-inhibitor therapy upon inflammation-mediated comorbidities in myelofibrosis and related neoplasms. Expert Rev Hematol 2014;7(2):203-16

..

A comprehensive review describing the masssive inflammation-mediated comorbidity burden in MPNs and the rationales for combination therapy with interferon-alpha2, ruxolitinib and statins.

51.

Chen E, Beer PA, Godfrey AL, et al. Distinct clinical phenotypes associated with JAK2V617F reflect differential STAT1 signaling. Cancer Cell 2010;18(5): 524-35

41.

Enblom A, Lindskog E Hasselbalch HC, et al. High rate of abnormal blood values and vascular complications before diagnosis of myeloproliferative neoplasms. Eur J Intern Med 2015. [Epub ahead of print]

52.

Fretham SJ, Carlson ES, Georgieff MK. The role of iron in learning and memory. Adv Nutr 2011;2(2):112-21

53.

Avni T, Leibovici L, Gafter-Gvili A. Iron supplementation for the treatment of chronic heart failure and iron deficiency: Systematic review and meta-analysis. Eur J Heart Fail 2012;14(4):423-9

42.

Marty C, Lacout C, Droin N, et al. A role for reactive oxygen species in JAK2 V617F myeloproliferative neoplasm progression. Leukemia 2013;27(11):2187-95

doi: 10.1586/17474086.2015.1045409

Expert Rev. Hematol.

Interferon in PV & related neoplasms

Expert Review of Hematology Downloaded from informahealthcare.com by 2.104.105.187 on 05/21/15 For personal use only.

polycythemia vera and myelofibrosis. Eur J Haematol 2014;Epub ahead of print 65.

Riley CH, Jensen MK, Brimnes MK, et al. Increase in circulating CD4+CD25+Foxp3+ T cells in patients with Philadelphia-negative chronic myeloproliferative neoplasms during treatment with IFN-a. Blood 2011;118(8): 2170-3

66.

Skov V, Riley CH, Thomassen M, et al. Whole blood transcriptional profiling reveals significant down-regulation of human leukocyte antigen class I and II genes in essential thrombocythemia, polycythemia vera and myelofibrosis. Leuk Lymphoma 2013;54(10):2269-73

67.

68.

69.

70.

Riley CH, Hansen M, Brimnes MK, et al. Expansion of circulating CD56bright natural killer cells in patients with JAK2-positive chronic myeloproliferative neoplasms during treatment with interferon-alpha. Eur J Haematol 2015; 94(3):227-34 Finazzi G, Caruso V, Marchioli R, et al. Acute leukemia in polycythemia vera: an analysis of 1638 patients enrolled in a prospective observational study. Blood 2005; 105(7):2664-70 Bjo¨rkholm M, Derolf AR, Hultcrantz M, et al. Treatment-related risk factors for transformation to acute myeloid leukemia and myelodysplastic syndromes in myeloproliferative neoplasms. J Clin Oncol 2011;29(17):2410-15 Tefferi A, Rumi E, Finazzi G, et al. Survival and prognosis among 1545 patients with contemporary polycythemia vera:

informahealthcare.com

interferon-alpha2 in the treatment of MPN.

an international study. Leukemia 2013; 27(9):1874-81 71.

Bjørn ME, de Stricker K, Kjær L, et al. Combination therapy with interferon and JAK1-2 inhibitor is feasible. proof of concept with rapid reduction in JAK2V617F-allele burden in polycythemia vera. Leuk Res Reports 2014;3(2):73-5

72.

Sackett DL, Rosenberg WM, Gray JA, et al. Editorials Evidence based medicine: what it is and what it isn’t. Br Med J 1996;312: 71-2

73.

74.

Day S. Evidence-based medicine and rare diseases. Chapter 3 in. Posada de la Paz M, Groft SC, Editors. Rare diseases epidemiology. Advances in experimental medicine and biology 2010. p. 41-53 Huang BT, Zeng QC, Zhao WH, et al. Interferon-alpha2b gains high sustained response therapy for advanced essential thrombocythemia and polycythemia vera with JAK2V617F positive mutation. Leuk Res 2014;38(10):1177-83

75.

Simon R, Rothenberg ML, Sommer J, et al. The role of non-randomized trials in the evaluation of oncology drugs. Clin Pharmacol Ther 2015;97(5):502-7

..

This paper discusses the role of non-randomized studies in the evaluation of oncology drugs and describes when and how single-arm trials can be used to support full approval of oncology drugs. Clinical experience in a sufficient number of patients to allow adequate assessment of the risk:benefit relationship is highlighted – an issue which is highly relevant in the context of

Perspective

76.

Hasselbalch HC. The platelet-cancer loop in myeloproliferative cancer. Is thrombocythemia an enhancer of cancer invasiveness and metastasis in essential thrombocythemia, polycythemia vera and myelofibrosis? Leuk Res 2014;38(10): 1230-6

77.

Randomized trial of pegylated interferon alfa-2a versus hydroxyurea in polycythemia vera (PV) and essential thrombocythemia (ET). Available from: https://clinicaltrials. gov/ct2/show/NCT01259856

78.

Pegylated interferon alpha-2b versus hydroxyurea in polycythemia vera (PROUD-PV). Available from: https:// clinicaltrials.gov/ct2/show/NCT01949805

79.

A Danish study of low-dose interferon-alfa versus hydroxyurea in the treatment of Philadelphia chromosome negative (Ph-) chronic myeloproliferative neoplasms. A national randomized prospective study with focus upon efficacy, toxicity and quality of life. Available from: http://clinicaltrials.gov/ ct2/show/NCT01387763

80.

Vannucchi AM, Kiladjian JJ, Griesshammer M, et al. Ruxolitinib versus standard therapy for the treatment of polycythemia vera. N Engl J Med 2015; 372(5):426-35

..

The first randomized study of ruxolitinib versus hydroxyurea in patients being resistant to or intolerant to treatment with hydroxyurea.

doi: 10.1586/17474086.2015.1045409