Statins in the treatment of polycythaemia vera and allied disorders: An antithrombotic and cytoreductive potential?

Statins in the treatment of polycythaemia vera and allied disorders: An antithrombotic and cytoreductive potential?

Leukemia Research 30 (2006) 1217–1225 Open forum Statins in the treatment of polycythaemia vera and allied disorders: An antithrombotic and cytoredu...

156KB Sizes 19 Downloads 49 Views

Leukemia Research 30 (2006) 1217–1225

Open forum

Statins in the treatment of polycythaemia vera and allied disorders: An antithrombotic and cytoreductive potential? Hans Carl Hasselbalch ∗ , Caroline H. Riley Department of Haematology, Odense University Hospital, University of Southern Denmark, Sdr. Boulevard 29, 5000 Odense, Denmark Received 2 December 2005; accepted 22 December 2005 Available online 17 February 2006

Abstract Thrombohaemorrhagic complications are major clinical problems in the classical chronic Ph-negative myeloproliferative disorders (CMPDs), polycytaemia vera (PV), essential thrombocythaemia (ET) and idiopathic myelofibrosis (IMF), contributing significantly to morbidity and mortality. Pathophysiologically these disorders are characterized by clonal myeloproliferation, myeloaccumulation and a propensity to develop myelofibrosis and neoangiogenesis in both the bone marrow and spleen. Based upon in vitro and in vivo studies of the effects of statins (antithrombotic, antiproliferative, proapoptotic and antiangiogenic), this review focuses on the translation of these effects into potential clinical benefits of statin therapy in patients with CMPDs. © 2006 Elsevier Ltd. All rights reserved. Keywords: Statins; Polycythaemia vera; Antiproliferative; Proapoptotic; Antithrombotic

1. Introduction The classical, chronic Ph-negative myeloproliferative disorders (CMPDs) encompass polycythaemia vera (PV), essentiel thrombocythaemia (ET) and idiopathic myelofibrosis (IMF) [1–4]. In PV and ET thrombohaemorrhagic complications are the major determinants of morbidity [5]. High platelet counts in high risk patients of thrombosis and haemorrhage, hypermetabolic symptoms or symptoms of myeloid metaplasia (e.g. abdominal discomfort due to an enlarged spleen) have been conventionally treated with busulphan or hydroxyurea. Both drugs are potentially leukemogenic at least when being used sequentially in this patient group [6–8]. In younger patients alpha-interferon, PEG-Intron or anagrelide may be useful alternatives [9–13]. Since all these agents have side effects – and some even potentially leukemogenic – clinical trials are being conducted to find alternative and better drug formulations [14–22]. These pilot studies have included signal transduction inhibition (STI) with tyrosine kinase inhibitors (STI571; imatinib mesylate, Gleevec) ∗

Corresponding author. Tel.: +45 465412135; fax: +45 44923346. E-mail address: [email protected] (H.C. Hasselbalch).

0145-2126/$ – see front matter © 2006 Elsevier Ltd. All rights reserved. doi:10.1016/j.leukres.2005.12.018

[15–19] and inhibition of the enzyme farnesyltransferase in the mevalonate pathway [20–22]. Most recently, the farnesyl transferase inhibitor tipifarnib has been shown to preferentially inhibit in vitro autonomous erythropoiesis of PV patient cells implying that this treatment modality may be efficious in PV [23]. In recent years, several studies have indicated that statins – besides a cholesterol lowering effect – possess anticancer properties as well by inhibition of the enzyme 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase upstream in the mevalonate pathway [24–41]. The statin family of drugs consists of those derived from fungal fermentation (pravastatin, simvastatin and lovastatin) and those which are fully synthetic (atorvastatin, fluvastatin, rosuvastatin, pitavastatin and cerivastatin (withdrawn from clinical use in 2001) [42,43]. Since statins besides antiproliferative, proapoptotic and antiangiogenic effects in addition have antithrombotic effects [44–53], including potential improvement of endothelial dysfunction [48,54–56] these agents may have a therapeutic potential in the CMPDs being featured by myeloproliferation, myeloaccumulation (decreased apoptosis), marked angiogenesis and an increased risk of thrombotic and haemorrhagic complications conse-

1218

H.C. Hasselbalch, C.H. Riley / Leukemia Research 30 (2006) 1217–1225

quent to platelet and endothelial cell activation/dysfunction [5,57–59]. Based upon in vitro and in vivo studies of the effects of statins (antithrombotic, antiproliferative, proapoptotic and antiangiogenic), this review focuses on the translation of these effects into potential clinical benefits of statin therapy in patients with CMPDs.

2. The mevalonate pathway 3-Hydroxy-3-methylglutaryl-coenzyme A reductase catalyses the conversion of HMG-CoA into mevalonate. This key enzyme is the rate-limiting step in cholesterol synthesis and the formation of isoprenoids such as farnesylpyrophosphate (FPP; C15) and geranylgeranylpyrophosphate (GGPP; C20) in the mevalonate pathway [60]. The enzyme is inhibited by statins giving rise to intracellular depletion of FPP and GGPP. Both intermediates of the mevalonate pathway are critical for activation of a variety of proteins involved in intracellular signaling. This activation step involves the transfer and subsequent binding of the farnesyl or geranylgeranylmoieties to the protein, resulting in a farnesylated or geranylgeranylated protein. This kind of protein activitation involving the covalent attachment of hydrophobic isoprenoid molecules to target proteins is referred to as prenylation [61]. The processes are catalysed by farnesyltransferase and geranylgeranyltransferase, respectively. Prenylation is essential for membrane attachment and the subsequent participation of prenylated proteins in diverse signaling pathways regulating cell growth and survival [61–64]. Proteins that require farnesylation or geranylgeranylation for their function include guanosine triphosphate (GTP)—binding proteins such as Ras (farnesylation) [62–64] and the Rho family members Rac1 and RoA [65]. Accordingly, blockade of the mevalonate pathway may take place at several sites either upstream by inhibition of HMG-CoA reductase (statins), by inhibition of the activity of farnesyl pyrophosphate synthase (zoledronic acid and related amino-biphosphonates) and consequent depletion of FPP and GGPP or by direct inhibition of enzymes involved in the farnesylation of the proteins (farnesyltransferase/geranylgeranyltransferase inhibitors).

3. Antiproliferative and proapoptotic effects During recent years the potential of statins in the treatment of haematological malignant diseases has been studied extensively [24–38,40]. As described above statins inhibit the prenylation of several important signaling proteins, including the Ras proteins. By inhibiting prenylation of Ras signal transduction in response to GF-stimulation the downstream activity in several important signaling pathways is inhibited with decreased proliferation and increased apoptosis [27,66–68]. The sensitivity of AML cells to statins appears not to be dependent on activating Ras mutations or overex-

pression and may also be dissociated both geranylgeranylation as well as farnesylation [34]. However, others have found that non-Ras geranylgeranylated proteins are important for statin-induced apoptosis of human AML cells [28]. Yet another mechanism responsible for the toxic effects of statins on AML cells seems to imply inhibition of intracellular cholesterol increments, which have been shown to be a protective cellular mechanism against injury [33,36]. Statins also profoundly modulate post-translational glycosylation of membrane targeted proteins consequent to depletion of dolichol. This compound is of major importance for the Nlinked glycosylation of membrane targeted proteins. In particular, insulin and insulin-like growth factor (IGF) signaling appear to be disrupted, since the intracellular processing of these receptors requires dolichol for correct N-glycosylation, implying the formation of intact and functional type 1 IGF and insulin receptors. Accordingly, deficient intracellular glycosylation of these receptors is followed by proreceptor retention within the endoplasmatic reticulum, prevention of their transfer to the cell membrane and consequently impairment of IGF-signaling and DNA-synthesis. Depletion of farnesyl pyrophosphate is also associated with impairment of mitogen signaling induced by both type 1 IGF receptor and the insulin receptor. Thus, HMG-CoA reductase inhibition and mevalonate depletion affects IGF and insulin signaling via at least two distinct but related and synergistic mechanisms: reduced prenylation of signaling intermediates such as Ras and disruption of the IGF and insulin proreceptor processing [69].

4. Angiogenic and antiangiogenic effects Statins have biphasic effects on endothelial proliferation, high doses of statins inhibiting angiogenesis [55,56,70,71]. These biphasic actions may be explained by dose-related activation of different pathways. Thus, high dose effects have been shown to be related to inhibition of geranylgeranylation, whereas the low-dose effects were mediated by activation of PI3K/Akt pathway [55,71]. A proangiogenic effect has been shown to be associated with increased activity of endothelial nitric oxide synthase (eNOS) [54]. Other studies have shown that statins impair angiogenesis, being related to a different mechanism implying inhibition of RhoA [72,73]. It is tempting to speculate if the antiangiogenic effect may also be mediated by disruption of IGF-I signaling consequent to depletion of dolichol in endothelial cells, since IGF-I-in addition to directly stimulating angiogenesis [74] may also promote angiogenesis by stimulating the production of vascular endothelial growth factor (VEGF) [75,76].

5. Antithrombotic effects Recently statins have been shown to exert direct cardiovascular effects, which are unrelated to cholesterol lowering [48,50,77]. As outlined above these include among others

H.C. Hasselbalch, C.H. Riley / Leukemia Research 30 (2006) 1217–1225

antithrombotic effects and effects upon endothelial function as well. In addition to a reduction in serum cholesterol levels statins may also protect against thrombosis by decreasing platelet reactivity consequent to a reduction in the production of thromboxan A2 (TXA2 ) [78] or a reduction in the cholesterol content of platelet and erythrocyte membranes with ultimative decrease in the thrombogenic potential of these cells [77,79]. Furthermore, statins may decrease platelet activation by upregulating type III nitric oxide synthase in platelets [47], reducing circulating levels of sCD40 which are released from platelets upon activation [80] and together with other substances released from activated platelets, including IL-1beta [81] may elicit a prothrombotic state [82]. Furthermore, statins have been shown to reduce CD40 expression on vascular cells and circulating platelets [83,84] and inhibit CD40-ligand-dependent induction of COX-2 expression in endothelial cells by activated platelets [85]. Statins also decrease risk of thrombosis by increasing the vasodilatory, antithrombotic or inflammatory properties of the vascular endothelium by upregulating the enzyme generating nitric oxide (NO) (endothelial nitric oxide synthase) [54] with ensuing increased NO production from endothelial cells. The NO enhancing effect of statins is of major importance in their protective effect against atherothrombosis. The endothelium is preserved and NO also retards platelet aggregation and leucocyte adherence to the endothelium via the endothelial cell adhesion molecules, P-selectin and intercellular adhesion molecule-1 (ICAM-1) [86–88]. A similar vasculoprotective statin effect is also achieved by increasing COX-2 expression and prostacyclin PGI2 formation [89]. Statins may also decrease the risk of thrombosis by modulating coagulation and fibrinolytic pathways [90]. Thus, statins enhance the fibrinolytic activity within the vessel wall as reflected by the findings of reduced levels of plasminogen activator inhibitor-I (PAI-I) and increased levels of tissue plasminogen activator (t-PA) within smooth muscle and endothelial cells [91]. Additionally, statins have been shown to inhibit tissue factor expression by monocytes and macrophages [77,92]. Statins also strongly enhance endothelial anticoagulant and fibrinolytic properties by increasing thrombomodulin expression and function in human endothelial cells [93].

6. Antiinflammatory effects Statins have been shown to markedly attenuate leucocyte adherence to the endothelium being partly related to downregulation of endothelial adhesion cell molecules (i.e. P-selectin, ICAM-1) [86–88] but also to similar antiinflammatory effects upon leucocyte adhesion molecules with downregulation of CD11b (i.e. the alpha-chain of the beta2 -integrins) and CD18 (i.e. the beta-chain of the beta2 integrins) [94]. All these antiadhesion molecule effects are dependent upon NO [88,94], and together they impair adherence of leucocytes to endothelium.

1219

Statins also effectively lower plasma levels of C-reactive protein (CRP) [95,96] and increased CRP-levels have been shown to be predictive of an increased risk for coronary artery disease (CAD) in apparently healthy subjects [97–100]. Furthermore, the proinflammtory cytokines–tumor necrosis factor-alpha (TNF-alpha) and interleukin-6 (IL-6), which stimulate CRP-production in the liver, are also decreased following treatment with statins [100,101]. This antiinflammatory effect may also positively influence the phenotype of the endothelium which otherwise in response to inflammatory mediators such as TNF-alpha and interleukin-1beta (IL-1beta) changes from a non-adhesive, non-thrombogenic cellular interface to one that expresses and secretes several adhesion molecules. Considering platelets as inflammatory cells statins accordingly have a combined antiinflammatory and antithrombogenic effect by decreasing circulating levels of sCD40 [80].

7. Discussion Several of the effects of statins may be translated into clinical benefits to patients with CMPDs. First, statins have potent antiproliferative and proapoptotic effects and accordingly may decrease clonal myeloproliferation and accumulation of myeloid cells consequent to impaired apoptosis [24–27,33,35,36,40]. Second, in high doses statins have an antiangiogenic effect, which may be beneficial in these disorders as well [72,73,102]. Thus, endothelial proliferation in bone marrow and spleen is a prominent feature in the advanced myelofibrotic phase of PV and IMF [103–108]. In line with the development of bone marrow fibrosis the neovascularisation in the bone marrow and spleen is considered to develop consequent to the release of various growth factors from the megakaryocyte cell lineage including VEGF and bFGF [109,110]. Increased circulating levels of VEGF have been recorded in all three disease categories, being most pronounced in myelofibrosis patients [111–113]. Vascular endothelial growth factor is considered a key mediator of the abnormal angiogenesis in many other haematological malignancies [114,115] and is a survival factor for endothelial cells. It is intriguing to consider the possibility that VEGF may act as a growth and survival factor in CMPDs as well. If so, statins may interfere with the growth and survival promoting activity of VEGF as well as other important growth factors, including PDGF and bFGF [38,116–120]. Furthermore, statins through their antiproteolytic effect also decrease the release of several growth factors (e.g. VEGF, bFGF and TGFbeta) from the ECM of importance for cell proliferation, apoptosis and angiogenesis [121]. Besides the VEGF-system, the insulin growth factor system may be of particular pathogenetic importance in the CMPDs. The IGF-system consists of a family of insulin related proteins, including two growth factors (IGF-1 and IGF-2), their receptors (IGF-1R and IGF-2R) and various high-affinity binding proteins that regulate bioavailability and

1220

H.C. Hasselbalch, C.H. Riley / Leukemia Research 30 (2006) 1217–1225

action of IGF [122]. Similar to VEGF’s, IGF’s are major proliferative/antiapoptotic cytokines and accordingly relevant therapeutic targets in several haematological neoplasias featured by enhanced proliferation and decreased apoptosis [123–125]. These growth factors promote cell proliferation and differentiation via specific receptors and are very important for erythropoiesis stimulating both erythroid and myeloid progenitor cells [122]. IGF-1 has an erythropoietin (Epo)like activity that targets circulating early progenitor cells via an Epo-independent pathway [126] and most recently, IGF2 has been shown to be a growth factor for haematopoietic stem cells [127]. The overproduction of red cells in PV has been explained by the combination of hypersensitivity of erythroid progenitor cells to IGF-I [128] and high circulating IGFBP-1 levels, the latter having a stimulating effect upon erythroid burst formation in vitro [129]. Recently, aberrant expression of IGF-2 has been described in Ph-chromosome negative CMPDs with a highly significant increase in IMF patients [130]. This observation further supports the concept of a significant role of this GF-system in CMPDs, being further underscored in the context of IGF-2 as a growth factor of haematopoietic stem cells [127]. Most recently, a major break-through in the molecular pathogenesis of the Ph-negative CMPDs has been described by the identification of a single point mutation (Val617Phe) in JAK2 in more than half of patients with CMPD being most prevalent in PVpatients (up to 97%) and present in about 50–60% of patients with ET and IMF as well [131–135]. JAK2 is a cytoplasmatic tyrosine kinase with a key role in signal transduction from multiple haematopoietic growth factor receptors [136]. Accordingly, the JAK2 mutation is thought to contribute to the sustained myeloproliferation and myeloaccumulation in the Ph-negative CMPDs. Regarding the mevalonate pathway as a therapeutic target in the treatment of CMPDs and the IGF-system having an important role in sustaining myeloproliferation and decreased apoptosis it is of particular importance that interruption of the mevalonate pathway also profoundly affects IGF-signaling [69] and inhibition of IGF-signaling by mevalonate depletion may influence VEGF-signaling as well [137]. Comparative studies of the potency of the various statins in terms of their antiproliferative and proapoptotic effects have shown that the fungal-derived agents (pravastatin, simvastatin and lovastatin) have virtually identical antiproliferative properties on AML-cells [24–26]. The synthetic statins (cerivastatin, fluvastatin, lovastatin and atorvastatin) have all shown to be cytotoxic to AML cell lines as well but amongst them cerivastatin was found to be at least 10 times more potent in triggering apoptosis. Next to cerivastatin, the AML cell lines were most sensitive to fluvastatin, lovastatin and atorvastatin [29]. Since then cerivastatin has been withdrawn from clinical use due to an increased incidence of rhabdomyolysis. The differences in their proapoptotic potential may be related to different chemical, pharmacokinetic and pharmacodynamic properties. In particular, the degree of lipophilicity may be of importance determining the propen-

sity of the drug to pass the cell membranes of neoplastic cells. Indeed, among the various liphophilic statins (cerivastatin, atorvastatin, simvastatin, lovastatin, fluvastatin and pitavastatin) cerivastatin has been shown to possess the greatest degree of lipophilicity [42,43] but also the most efficient statin to induce tumor-specific apoptosis [29]. On the contrary, among the hydrophilic statins (pravastatin and rosuvastatin), pravastatin has been found not to influence cell proliferation, which may be attributed to the hydrophilic nature of pravastatin and accordingly low penetration of the cells by the drug [41,43,44]. In the latter study human lymphoblast and myeloma cell lines were used, showing atorvastatin, lovastatin and simvastatin to possess almost identical cytotoxic effects, albeit the proapoptotic potential of atorvastatin and lovastatin was more pronounced than simvastatin [41]. Thrombosis and bleeding are frequent complications during the course of the CMPD’s. Besides abnormal rheology due to a raised haematocrit in PV-patients these events are primarily attributed to platelet and endothelial cell activation and dysfunction [5,57–59]. Other well-known risk factors for thrombosis in the general population – smoking, hypertension and diabetes mellitus – may further aggravate the thrombotic risk in these patients. Accordingly these risk factors are included in the risk-stratification of the individual patient when deciding the optimal treatment and when to start cytoreductive treatment [138]. Increased platelet activity has been found in several studies of patients with CMPDs [5] and is a well-known risk factor of acute coronary syndromes. In patients with hypercholesterolaemia, statins have been shown to decrease platelet activation being partly explained by lowering of plasma lipids. However, the antiplatelet effect also occurs independent of lipid lowering [77]. In patients with CMPDs, a direct antiplatelet effect of statins may be exerted by decreasing circulating levels of sCD40 [80] and accordingly also a CD40-mediated prothrombotic state [82–85]. Most recently, atorvastatin has been shown to reduce platelet activity by lowering the platelet-oxidized-LDL receptor expression in hypercholesterolaemic patients. Platelet deactivation occurred before significant LDL changes. Accordingly, the antiatherothrombotic action of atorvastatin was considered to be related to a cholesterol-independent reduction of the expression of a receptor molecule (oxidized-LDL) of utmost importance for platelet activation and development of thrombosis [139]. Endothelial activation and dysfunction has also been recorded in these patients as evidenced by increased plasma levels of the soluble adhesion molecule E-selectin (sELAM) and thrombomodulin [57,59] and a markedly impaired endothelial-dependent flow-mediated vasodilation [58]. The highest levels of sELAM were recorded in CMPD patients with thrombosis [57]. Thus, it has been suggested that dysfunctional endothelium with loss of its thromboresistant properties might also account for the increased risk of thrombosis in CMPD patients [57]. Statins may improve and restore endothelial function in CMPDs by stimulating endothelial nitric oxide synthase activity and consequently enhancing

H.C. Hasselbalch, C.H. Riley / Leukemia Research 30 (2006) 1217–1225

NO production from endothelial cells [54,140]. Increased NO production also has an antithrombotic effect by impairment of both platelet aggregation and leucocyte adherence to the endothelium [87,102], the latter effect being elicited by downregulation of endothelial (P-selectin and ICAM1) and leucocyte (CD11b and CD18) adhesion molecules [86–88,94,102]. Furthermore, statins also strongly upregulate endothelial cell thrombomodulin expression and activity thereby enhancing the anticoagulant and antiinflammatory properties of the vascular endothelium [93]. Recently, aspirin has been shown to reduce the risk of thrombosis in PV patients, but still a large proportion of the patients experience thrombotic events [141], which may not solely be attributed to platelet hyperactivity but also to elevated leucocyte counts and leucocyte activation [5,142]. The role of polymorphonuclear leucocytes in thrombogenesis has been recognized for several years [143]. Direct interaction of neutrophils and platelets are mediated through P-selectin (CD62P), which represents the first step in the formation of a leucocyte–platelet thrombus in vivo. Indeed, increased circulating platelet–leucocyte aggregates have been found in a significant proportion of patients with CMPDs being correlated with previous thrombosis, platelet activation and platelet count [144]. The statin fluvastatin reduces expression of P-selectin on platelets from hypercholesterolemic patients [88] and inhibit platelet–neutrophil interaction, this effect being mediated by inhibition of Rho-GTPases [145]. Based upon the observed alterations in platelet and endothelial function in patients with CMPDs and the above mentioned beneficial effects of statins upon platelet, leucocyte and endothelial interactions statins may certainly confer a protective role against thrombosis in CMPDs, in which increased thromboxan biosynthesis have been recorded in a large proportion of patients with PV [146,147]. The clinical benefits of statins may be evident even in patients receiving aspirin, reflecting that additional mechanisms other than those related to reduction in thromboxane are responsible [148]. Thus, statin therapy has been shown to reduce C-reactive protein levels independently of its effect on cholesterol [95,96]. A chronic low-level activation of the acute-phase response, either due to chronic inflammation, smoking, or biologically with increasing age is known to be associated with an increased long-term risk of atherosclerosis (angina pectoris), atherothrombosis (myocardial infarction) and death in the general population, even in persons with normal lipid levels [98–100,149]. Furthermore, statin therapy seems to be effective in the primary prevention of coronary events among persons with relatively low lipid levels but with elevated levels of C-reactive protein [150]. In addition, a reduced rate of progression of atherosclerosis is asssociated with intensive statin treatment [151]. Finally, persons with low CRP levels after statin treatment have been shown to have a better outcome than those with higher CRP levels irrespective of the resultant level of LDL cholesterol [152]. All these data provide a rationale for statin therapy of patients with CMPDs, at least in those with chronic, low-level activation of the acute-

1221

phase response as reflected in elevated C-reactive protein levels. 8. Conclusions and perspectives Statins have potent antiplatelet and antiinflammatory effects, which together with restoration of endothelial dysfunction may decrease the risk of thrombotic complications in patients with CMPDs. In addition, statins have been shown to inhibit leukaemic cell proliferation, increase apoptosis of leukaemic cells and have demonstrated antiangiogenic effects. Given these observations a potential role of statins as cytoreductive and antithrombotic agents in patients with CMPDs may be anticipated. If so, these effects may be further enhanced by combinational therapy with zoledronic acid, considering their different site of action within the mevalonate pathway [153]. Acknowledgement This work is unfunded with no commercial relationship. References [1] Reilly JT. Idiopathic myelofibrosis: pathogenesis, natural history and management. Blood Rev 1997;11(4):233–42. [2] Tefferi A. The chronic myeloproliferative disorders. Semin Hematol 1999;36(Suppl. 2):3–8. [3] Tefferi A. Myelofibrosis with myeloid metaplasia. N Engl J Med 2000;342:1255–65. [4] Spivak JL. The chronic myeloproliferative disorders: clonality and clinical heterogeneity. Semin Hematol 2004;41(2 Suppl. 3):1–5. [5] Elliot MA, Tefferi A. Thrombosis and haemorrhage in polycythaemia vera and essential thrombocythaemia. Br J Haematol 2005;128:275–90. [6] Finazzi G, Ruggeri M, Rodeghiero F, Barbui T. Second malignancies in patients with essential thrombocythaemia treated with busulphan and hydroxyurea: long-term follow-up of a randomised clinical trial. Br J Haematol 2000;110:577–83. [7] Nielsen I, Hasselbalch HC. Acute leukemia and myelodysplasia in patients with philadelphia chromosome negative chronic myeloproliferative disorder treated with hydroxyurea alone or with hydroxyurea after busulphan. Am J Hematol 2003;74:26–31. [8] Finnazi G, Caruso V, Marchiola R, Capnist G, Chiesi T, Finelli C, et al. Acute leukemia in polycythemia vera. An analysis of 1,638 patients enroled in a prospective observational study. Blood 2005;105:2664–70. [9] Gilbert HS. Long term treatment of myeloproliferative disease with interferon-alpha-2b: feasibility and efficacy. Cancer 1998;83:1205–13. [10] Bachleitner-Hofmann T, Gisslinger H. The role of interferonalpha in the treatment of idiopathic myelofibrosis. Ann Hematol 1999;78:533–8. [11] Radin AI, Kim HT, Grant BW, Bennett JM, Kirkwood JM, Stewart JA, et al. Phase II study of alpha2 interferon in the treatment of the chronic myeloproliferative disorders. Cancer 2003;98:100–9. [12] Birgegard G, Bjorkholm M, Kutti J, Larfars G, Lofvenberg E, Markevarn B, et al. Adverse effects and benefits of two years treatment for thrombocythemia in chronic myeloproliferative disorders. Haematologica 2004;89:520–7.

1222

H.C. Hasselbalch, C.H. Riley / Leukemia Research 30 (2006) 1217–1225

[13] Steurer M, Gastl G, Jedrzejczak WW, Pytlik R, Lin W, Schlogl E, et al. Anagrelide for thrombocytosis in myeloproliferative disorders: a prospective study to assess efficacy and adverse event profile. Cancer 2004;101:2239–46. [14] Barosi G, Michelle E, Letizia C, Fillipo B, Paulo PP, Giuseppe V, et al. Thalidomide in myelofibrosis with myeloid metaplasia: a pooled analysis of individual patient data from five studies. Leuk Lymphoma 2002;43:2301–17. [15] Tefferi A, Mesa RA, Gray LA, et al. Phase 2 trial of imatiib mesylate in myelofibrosis with myeloid metaplasia. Blood 2002;99:3854–6. [16] Hasselbalch HC, Bjerrum OW, Jensen BA, Tøffner Clausen N, Hansen PB, Birgens H, et al. Imatinib mesylate in idiopathic and postpolycythemic myelofibrosis. Am J Hematol 2003;74:238–42. [17] Jones CM, Dickinson TM. Polycythemia vera responds to imatinib mesylate. Am J Med Sci 2003;325:149–52. [18] Silver RT, Silver RT. Imatinib mesylate (GleevecTM ) reduces phlebotomy requirements in polycythemia vera. Leukemia 2003;17:1186–7. [19] Hasselbalch HC, Larsen TS, Henriksen AS, Christensen JH, Hansen PG, Pedersen NT, et al. Imatinib mesylate in polycythemia vera. A heterogeneous response pattern but a consistent reduction in phlebotomy requirements. Blood 2004;104 [Abstract # 4747]. [20] Mesa RA, Camoriano JK, Geyer SM, Kaufmann SH, Ehrlichman C, Juckett M, et al. A phase 2 consortium (P2C) trial of R115777 (Zarnestra) in myelofibrosis with myeloid metaplasia. Blood 2004;102 [Abstract # 3428]. [21] Cortes J, Albitar M, Thomas D, Giles F, Kurzrock R, Thibault A, et al. Efficacy of the farnesyl transferase inhibitor R115777 in chronic myeloid leukemia and other hematologic malignancies. Blood 2003;101:1692–7. [22] Jabbour E, Kantarjian H, Cortes J. Clinical activity of farnesyltransferase inhibitors in hematologic malignancies: possible mechanisms of action. Leuk Lymphoma 2004;45:2187–95. [23] Larghero J, Gervais N, Cassinat B, Rain JD, Schlageter MH, Padua RA, et al. Farnesyltransferase inhibitor tipifarnib (Zarnestra R115777) preferentially inhibits in vitro autonomous erythropoiesis of polycythemia vera patient cells. Blood 2005;105:3743–5. [24] Newman A, Clutterbuck RD, Powles RL, Millar JL. Selective inhibition of primary acute myeloid leukaemia cell growth by simvastatin. Leukemia 1994;8:2023–9. [25] Newman A, Clutterbuck RD, Powles RL, et al. A comparison of the effect of the 3-hydroxy-3-methylglutaryl coenzyme A (HMGCoA) reductase inhibitors simvastatin, lovastatin, and pravastatin on leukaemic and normal bone marrow progenitors. Leuk Lymphoma 1997;24:533–7. [26] Dimitroulakos J, Nohynek D, Backway KL, et al. Increased sensitivity of acute myeloid leukemias to lovastatin-induced apoptosis: a potential therapeutic approach. Blood 1999;93:1308–18. [27] Dimitroulakos J, Thai S, Wasfy GH, Hedley DW, Minden MD, Penn LZ. Lovastatin induces a pronounced differentiation response in acute myeloid leukaemias. Leuk Lymphoma 2000;40: 167–78. [28] Xia Z, Tan MM, Wong WW, Dimitroulakos J, Minden MD, Penn LZ. Blocking protein geranylgeranylation is essential for lovastatin-induced apoptosis of human acute myeloid leukemia cells. Leukemia 2001;15:1398–407. [29] Wong WW, Tan MM, Xia Z, Dimitroulakos J, Minden MD, Penn LZ. Cerivastatin triggers tumor-specific apoptosis with higher efficacy than lovastatin. Clin Cancer Res 2001;7:2067–75. [30] Chapman-Shimshoni D, Yuklea M, Radney J, Shapiro H, Lischner M. Simvastatin induces apoptosis of B-CLL cells by activation of mitochrondial caspase 9. Exp Hematol 2003;31:779–83. [31] Kaushal V, Kohli M, Mehta P. Potential anticancer effects of statins: facts or fiction? Endothelium 2003;10:49–58. [32] Chan KKW, Oza AM, Siu LL. The statins as anticancer agents. Clin Cancer Res 2003;9:10–9.

[33] Li HY, Appelbaum FR, Willman CL, Zager RA, Banker DE. Cholesterol-modulating agents kill acute myeloid leukaemia cells and sensitize them to therapeutics by blocking adaptive cholesterol responses. Blood 2003;101:3628–34. [34] Stirewalt DL, Appelbaum FR, Willman CL, Zager RA, Banker DE. Mevastatin can increase toxicity in primary AMLs exposed to standard therapeutic agents, but statin efficacy is not simply associated with ras hotspot mutations or overexpression. Leuk Res 2003;27:133–45. [35] Grant S. Reducing cholesterol and overcoming drug resistance. Blood 2004;104:1598–9. [36] Banker DE, Mayer SJ, Li HY, Willman CL, Appelbaum FR, Zager RA. Cholesterol synthesis and import contribute to protective cholesterol increments in acute myeloid leukaemia cells. Blood 2004;104:1816–24. [37] Schmidmaier R, Baumann P, Simsek M, Dayyani F, Emmerich B, Meinhardt G. The HMG-CoA reductase inhibitor simvastatin overcomes cell-adhesion-mediated drug resistance in multiple myeloma by geranylgeranylation of Rho protein and activation of Rho kinase. Blood 2004;104:1825–32. [38] Graff MR, Richel DJ, van Noorden CJF, Guchelaar HJ. Effects of statins and farnesyltransferase inhibitors on the development and progression of cancer. Cancer Treat Rev 2004;30:609–41. [39] Poynter JN, Gruber SB, Higgins PD, Almog R, Bonner JD, Rennert HS, et al. Stains and the risk of cancer. N Engl J Med 2005;352:2184–92. [40] Lewis KA, Holstein SA, Hohl RJ. Lovastatin alters isoprenoid biosynthetic pathway in acute myelogenous leukemia cells in vivo. Leuk Res 2005;29:527–33. [41] Cafforio P, Dammaco F, Gernone A, Silvestris F. Statins activate the mitochondrial pathway of apoptosis in human lymphoblasts and myeloma cells. Carcinogenesis 2005;26:883–91. [42] Corsini A, Maggi FM, Catapano AL. Pharmacology of competetive inhibitors of HMG-CoA reductase. Pharmacol Res 1995;31:9–27. [43] Schacter M. Chemical, pharmacokinetic and pharmacodynamic properties of statins: an update. Fundam Clin Pharmacol 2005;1:117–25. [44] Rosenson RS, Tangney CC. Anti-atherothrombotic properties of statins-implications for cardiovascular event reduction. JAMA 1998;279:1643–50. [45] Fenton Jr JW, Shen GX. Statins as cellular antithrombotics. Haemostasis 1999;29:166–9. [46] Kearney D, Fitzgerald D. The anti-thrombotic effects of statins. J Am Coll Cardiol 1999;33:1305–7. [47] Laufs U, Gertz K, Huang P, Nickenig G, Bohm M, Dirnagl U, et al. Atorvastatin upregulates type III nitric oxide synthase in thrombocytes, decreases platelet activation, and protects from cerebral ischemia in normocholesterolemic mice. Stroke 2000;31:2437–49. [48] Lefer AM, Scalia R, Lefer DJ. Vascular effects of HMG CoA-reductase inhibitors (statins) unrelated to cholesterol lowering: new concepts for cardiovascular disease. Cardiovasc Res 2001;49:281–7. [49] LaRosa JC. Pleiotropic effects of statins and their clinical significance. Am J Cardiol 2001;88:291–3. [50] Liao JK. Beyond lipid lowering: the role of statins in vascular protection. Int J Cardiol 2002;86:5–18. [51] Bonetti PO, Lerman LO, Napoli C, Lerman A. Statin effects beyond lipid lowering—are they clinically relevant? Eur Heart J 2003;24:225–48. [52] Tekten T, Ceyhan C, Ercan E, Onbasili AO, Turkoglu C. The effect of atorvastatin on platelet function in patients with coronary artery disease. Acta Cardiol 2004;59:311–5. [53] Hwang YS, Tsai WC, Lu YH, Lin CC, Chen YF. Effect of atorvastatin on the expression of CD40 ligand and P-selectin on platelets in patients with hypercholesterolemia. Am J Cardiol 2004;94:364–6. [54] Laufs U, Fata VL, Liao JK. Inhibition of 3-hydroxy-3methylglutaryl (HMG)-CoA reductase blocks hypoxia-mediated

H.C. Hasselbalch, C.H. Riley / Leukemia Research 30 (2006) 1217–1225

[55] [56] [57]

[58]

[59]

[60] [61] [62] [63] [64]

[65] [66]

[67]

[68]

[69]

[70]

[71]

[72]

[73]

[74]

[75]

down-regulation of endothelial nitric oxide synthase. J Biol Chem 1997;272:31725–9. Weis M, Heeschen C, Glassford AJ, Cooke JP. Statins have biphasic effects on angiogenesis. Circulation 2002;105:739–45. Ferrara DE, Pierangeli SS. Diverse effects of statins on endothelial cells? Thromb Haemost 2005;93:186–8. Musolino C, Alonci A, Allegra A, Spatari G, Bellomo G, Tringali O, et al. Increased levels of the soluble adhesion molecule Eselectin in patients with chronic myeloproliferative disorders and thromboembolic complications. Am J Hematol 1998;57:109–12. Neunteufl T, Heher S, Stefenelli T, Pabinger I, Gisslinger H. Endothelial dysfunction in patients with polycythaemia vera. Br J Haematol 2001;115:354–9. Jensen MK, de Nully Brown P, Thorsen S, Hasselbalch HC. Frequent occurrence of anticardiolipin antibodies, factor V Leiden mutation, and perturbed endothelial function in chronic myeloproliferative disorders. Am J Hematol 2002;69:185–91. Goldstein JL, Brown MS. Regulation of the mevalonate pathway. Nature 1990;343:425–30. Gelb MH. Protein prenylation, et cetera: signal transduction in two dimensions. Science 1997;275:1750–1. Rebello A, Martinez A. Ras proteins: recent advances and new functions. Blood 1999;94:2971–80. Magee T, Marshall C. New insights into the interaction of Ras with the cell membrane. Cell 1999;98:9–12. Reuter CW, Morgan MA, Bergmann L. Targeting the Ras signalling pathway: a rational, mechanism-based treatment of hematologic malignancies? Blood 2000;96:1655–69. Bishop AL, Hall A. Rho GTPases and their effector proteins. Biochem J 2000;348:241–55. Park WH, Lee YY, Kim ES, et al. Lovastatin-induced inhibition of HL-60 cell proliferation via cell cycle arrest and apoptosis. Anticancer Res 1999;19:3133–40. Agarwal B, Bhendwal S, Halmros B, Moss SF, Ramey WG, Holt PR. Lovastatin augments apoptosis induced by chemotherapeutic agents in colon cancer cells. Clin Cancer Res 1999;5:2223–9. Blanco-Colio LM, Villa A, Ortego M, et al. 3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, atorvastatin and simvastatin, induce apoptosis of vascular smooth muscle cells by downregulation of Bcl-2 expression and Rho A prenylation. Atherosclerosis 2002;161:17–26. Siddals KW, Marshman E, Westwod M, Gibson M. Abrogation of insulin-like growth factor-I (IGF-I) and insulin action by mevalonate depletion. J Biol Chem 2004;279:38353–9. Kureishi Y, Luo Z, Shiojima I, Bialik A, Fulton D, Lefer DJ, et al. The HMG-CoA reductase inhibitor simvastatin activates the protein kinase Akt and promotes angiogenesis in normocholesterolemic animals. Nat Med 2000;6:1004–10. Urbich C, Dernbach E, Zeither AM, Dimmeler S. Double-edged role of statins in angiogenesis signaling. Circ Res 2002;90: 737–44. Vincent L, Soria C, Mirshahi F, Opolon P, Mishal Z, Vannier JP, et al. Cerivastatin, an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibits endothelial proliferation induced by angiogenic factors in vitro and angiogenesis in in vivo models. Arterioscler Thromb Vasc Biol 2002;22:623–9. Vincent L, Albanese P, Bompais H, Uzan G, Vannier JP, Steg PG, et al. Insights in the molecular mechanisms of the anti-angiogenic effects of an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A reductase. Thromb Haemost 2003;89:530–7. Nicosia RF, Nicosia SV, Smith M. Vascular endothelial growth factor, platelet-derived growth factor, and insulin-like growth factor-1 promote rat aortic angiogenesis in vitro. Am J Pathol 1994;145:1023–9. Warren RS, Yuan H, Matli MR, Ferrara N, Donner DB. Induction of vascular endothelial growth factor by insulin-like growth factor I in colorectal carcinoma. J Biol Chem 1996;271:29483–8.

1223

[76] Akagi Y, Liu W, Zebrowski B, Xie K, Ellis LM. Regulation of vascular endothelial growth factor expression in human colon cancer by insulin-like growth factor-I. Cancer Res 1998;58:4008–14. [77] Liao JK. Effects of statins on 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibition beyond low-density lipoprotein cholesterol. Am J Cardiol 2005;96:24F–33F. [78] Notarbartolo A, Davi G, Averna M, Bargallo CM, Ganci A, Giammarresi C, et al. Inhibition of thromboxane biosynthesis and platelet function by simvastatin in type IIa hypercholesterolemia. Arterioscler Thromb Vasc Biol 1995;15:247–51. [79] Lijnen P, Celis H, Fagard R, Staessen J, Amey A. Influence of cholesterol lowering on plasma membrane lipids and cationic transport systems. J Hypertens 1994;12:59–64. [80] Malyszko J, Malyszko JS, Hryszko T, Mysliwiec M. Increased soluble CD40L levels are reduced by long term simvastatin in peritoneally dialyzed patients. Blood Coagul Fibrinolysis 2004;15:463–7. [81] Lindemann S, Tolley ND, Dixon DA, et al. Activated platelets mediate inflammatory signaling by regulated interleukin 1beta synthesis. J Cell Biol 2001;154:485–90. [82] Cippolone F, Mezzetti A, Porreca E, Di Febbo C, Nutini M, Fazia M, et al. Association between enhanced soluble CD40L and prothrombotic state in hypercholesterolemia: effects of statin therapy. Circulation 2002;106:399–402. [83] Mulhaupt F, Matter CM, Kwak BR, Pelli G, Veillard NR, Burger F, et al. Statins (HMG-CoA reductase inhibitors) reduce CD40 expression in human vascular cells. Cardiovasc Res 2003;59:755–66. [84] Hwang YS, Tsai WC, Lu YH, Lin CC, Chen YF. Effect of atorvastatin on the expression of CD40 ligand and P-selectin on platelets in patients with hypercholesterolemia. Am J Cardiol 2004;94:364–6. [85] Mosheimer BA, Kaneider NC, Feistritzer C, Djanani A, Sturn DH, Patsch JR, et al. CD40-ligand-dependent induction of COX2 expression in endothelial cells by activated platelets: inhibitory effects of atorvastatin. Blood Coagul Fibrinolysis 2005;16:105–10. [86] Niwa S, Totsuka T, Hayashi S. Inhibitory effect of fluvastatin, a HMG-CoA reductase inhibitor on the expression of adhesion molecules on human monocyte cell line. Int J Immunopharmacol 1996;18:669–75. [87] Pruefer D, Scalia R, Lefer AM. Simvastatin inhibits leukocyteendothelial cell interactions and protects against inflammatory processes in normocholesterolemic rats. Arterioscler Thromb Vasc Biol 1999;19:2894–900. [88] Romano M, Mezzetti A, Marulli C, et al. Fluvastatin reduces soluble P-selectin and ICAM-1 levels in hypercholestreolemic patients: role of nitric oxide. J Invest Med 2000;48:183–9. [89] Degraeve F, Bolla M, Blaie S, et al. Modulation of COX-2 expression by statins in human aortic smooth muscle cells: involvement of geranylgeranylated proteins. J Biol Chem 2001;276:46849–55. [90] Eto M, Luscher TF. Modulation of coagulation and fibrinolytic pathways by statins. Endothelium 2003;10:35–41. [91] Lopez S, Peiretti F, Bonardo B, Juhan-Vague I, Nalbone G. Effect of atorvastatin and fluvastatin on the expression of plasminogen activator inhibitor type-1 in cultured human endothelial cells. Atherosclerosis 2000;152:359–66. [92] Baetta R, Camera M, Comparato C, Altana C, Ezekowitz MD, Tremoli E. Fluvastatin reduces tissue factor expression and macrophage accumulation in carotid lesions of cholesterol-fed rabbits in the absence of lipid lowering. Arterioscler Thromb Vasc Biol 2002;22:692–8. [93] Shi J, Wang J, Zheng H, Ling W, Joseph J, Li D, et al. Statins increase thrombomodulin expression and function in human endothelial cells by a mitric oxide-dependent mechanism and counteract tumor necrosis factor-alpha-induced thrombomodulin downregulation. Blood Coagul Fibrinolysis 2003;14:575–85. [94] Lefer AM, Campbell B, Shin Y-K, Scalia R, Hayward R, Lefer DJ. Simvastatin preserves the ishemic-reperfused myocardium in normocholesterolemic rat hearts. Circulation 1999;100:178–84.

1224

H.C. Hasselbalch, C.H. Riley / Leukemia Research 30 (2006) 1217–1225

[95] Ridker PM, Rifai N, Pfeiffer MA, Sacks F, Braunwald E. Longterm effects of pravastatin on plasma concentrations of C-reactive protein. Circulation 1999;100:230–5. [96] Ridker PM, Rifai N, Pitman Lowenthal S. Rapid reduction in Creactive protein with cerivastatin among 785 patients with primary hypercholesterolemia. Circulation 2001;103:1191–3. [97] Ridker PM, Glynn RJ, Hennekens CH. C-reactive protein adds to the predictive value of total and HDL cholesterol in determining risk of first myocardial infarction. Circulation 1998;97:2007–11. [98] Koenig W, Sund M, Frohlich M, et al. C-reactive protein, a sensitive marker of inflammation, predicts future risk of coronary heart disease in initially healthy middle-aged men: results from MONICA (Monitoring Trends and Determinants in Cardiovascular Disease) Augsburg Chort Study, 1984 to 1992. Circulation 1999;99:237–42. [99] Rohde LE, Hennekens CH, Ridker PM. Survey of C-reactive protein and cardiovascular risk factors in apparently healthy men. Am J Cardiol 1999;84:1018–22. [100] Ridker PM, Hennekens CH, Buring JE, Rifai N. C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women. N Engl J Med 2000;342:836–43. [101] Rosenson RS, Tangney CC, Casey LC. Inhibition of proinflammatory cytokine production by pravastatin. Lancet 1999;353:983–4. [102] Mason JC. Statins and their role in vascular protection. Clin Sci 2003;105:251–66. [103] Reilly JT, Nash JR, Mackie MJ, McVerry BA. Endothelial cell proliferation in myelofibrosis. Br J Haematol 1985;60:625–30. [104] Thiele J, Rompcik V, Wagner S, Fischer R. Vascular architecture and collagen type IV in primarymyelofibrosis and polycythaemia vera: an immunomorphomeric study on trephine biopsies of bone marrow. Br J Haematol 1992;80:227–34. [105] Lundberg LG, Lerner R, Sundelin R, Rogers R, Folkman J, Palmblad J. Bone marrow in polycythemia vera, chronic myelocytic leukemia and myelofibrosis has an increased vascularity. Am J Pathol 2000;157:15–9. [106] Panteli K, Zagorianakou N, Bai M, Katsaraki A, Agnantis NJ, Bourantas K. Angiogenesis in chronic myeloproliferative diseases detected by CD34 expression. Eur J Haematol 2004;72:410–5. [107] Barosi G, Vittorio R, Margherita M, Luca VG, Alessandro P, Vittorio N, et al. Spleen neoangiogenesis in patients with myelofibrosis with myeloid metaplasia. Br J Haematol 2004;124:618–25. [108] Arora B, Ho CL, Hoyer JD, Mesa RA, Tefferi A. Bone marrow angiogenesis and its clinical correlates in myelofibrosis with myeloid metaplasia. Haematologica 2004;89:1454–8. [109] Castro-Malaspina H. Pathogenesis of myelofibrosis: role of ineffective megakaryopoiesis and megakaryocyte components. In: Myelofibrosis and the biology of connective tissue. New York: Alan R. Liss, Inc.; 1984. p. 427–454. [110] Le Bousse-Kerdiles MC, Martyre MC. Dual implication of fibrogenic cytokines in the pathogenesis of fibrosis and myeloproliferation in myeloid metaplasia with myelofibrosis. Ann Hematol 1999;78:437–44. [111] Raimundo FD, Azzaro MP, Palumbo GA, Bagnoto S, Stagno F, Giustolisi GM, et al. Elevated vascular endothelial growth factor (VEGF) serum levels in idiopathic myelofibrosis. Leukemia 2001;15:976–80. [112] Murphy P, Ahmed N, Hassan HT. Increased serum levels of vascular endothelial growth factor correlate with splenomegaly in polycythemia vera. Leuk Res 2002;26:1007–10. [113] Musolino C, Calabro’ L, Bellomo G, Martello F, Loteta B, Pezzano C, et al. Soluble angiogenic factors: implications for chronic myeloproliferative disorders. Am J Hematol 2002;69: 159–63. [114] Mesters RM. Angiogenesis in hematological malignancies. Ann Hematol 2002;81:S72–4. [115] Podar K, Anderson KC. The pathophysiological role of VEGF in hematological malignancies: therapeutic implications. Blood 2005;105:1383–95.

[116] McGuire TF, Corey SJ, Sebti SM. Lovastatin inhibits plateletderived growth factor (PDGF) stimulation of phosphatidylinositol 3-kinase activity as well as association of p85 subunit to tyrosine-phosphorylated PDGF receptor. J Biol Biochem 1993;268:22227–30. [117] McGuire TF, Xu XQ, Corey SJ, Romero GG, Sebti SM. Lovastatin disrupts early events in insulin signaling: a potential mechanism of lovastatin’s anti-mitogenic activity. Biochem Biophys Res Commun 1994;204:399–406. [118] Iwama A, Sawamaru M, Nara Y, Yamori Y. Effect of lovastatin and fluoromevalonate on phosphatidylinositol 3-kinase activity stimulated with PDGF. Clin Exp Pharmacol Physiol 1995;22(Suppl. 1):318–20. [119] Xu XQ, McGuire TF, Blastkovich MA, Sebti SM, Romero G. Lovastatin inhibits the stimulation of mitogen-activated protein kinase by insulin in HIRcB fibroblasts. Arch Biochem Biophys 1996;326:233–7. [120] Skaletz-Rorowski A, Muller JG, Kroke A, et al. Lovastatin blocks basic fibroblast growth factor-induced mitogen-activated protein kinase signaling in coronary smooth muscle cells via phosphatase inhibition. Eur J Cell Biol 2001;80:207–12. [121] Alfano D, Franso P, Vocca I, Gambi N, Pisa V, Mancini A, et al. The urokinase plasminogen activator and its receptor: role in cell growth and apoptosis. Thromb Haemost 2005;93: 190–1. [122] Zumkeller W. The insulin-like growth factor system in haematopoietic cells. Leuk Lymphoma 2002;43:487–91. [123] LeRoith D, Roberts CT. The insulin-like growth factor system and cancer. Cancer Lett 2003;195:127–37. [124] Pollak MN, Schernhammer ES, Hankinson SE. Insulin-like growth factors and neoplasia. Nat Rev 2004;4:505–18. [125] Mitsiades CS, Mitsiades NS, McMullan CJ, Poulaki V, Shringarpure R, Akiyama M, et al. Inhibition of the insulin-like growth factor receptor-1 tyrosine kinase activity as a therapeutic strategy for multiple myeloma, other hematological malignancies, and solid tumors. Cancer Cell 2004;5:221–30. [126] Correa PN, Axelrrad AA. Production of erythropoietic bursts by progenitor cells from adult human peripheral blood in an improved serum-free medium: role of insulin like growth factor I. Blood 1991;78:2823–33. [127] Zhang CC, Lodish HF. Insulin-like growth factor 2 expressed in a novel fetal liver cell population is a growth factor for hematopoietic stem cells. Blood 2004;103:2513–21. [128] Correra PN, Eskinazi D, Axelrad AA. Circulating erythroid progenitors in polycythemia vera are hypersensitive to insulin-like growth factor-1 in vitro: studies in an improved serum-free medium. Blood 1994;83:99–112. [129] Mirza AM, Ezzat S, Axelrad AA. Insulin-like growth factor binding protein is elevated in patients with polycythemia vera and stimulates erythroid burst formation in vitro. Blood 1997;89:1862–9. [130] Bock O, Tessema M, Serinsoz E, von Wasielewski R, Busche G, Kreipe H. Aberrant expression of insulin-like growth factor-2 (IGF2) in Philadelphia chromosome negative chronic myeloproliferative disorders. Leuk Res 2004;28:1145–51. [131] Baxter EJ, Scott LM, Campbell PJ, East C, Fourouclas N, Swanton S, et al. Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 2005;365:1054–61. [132] James C, Ugo V, Le Couedic JP, Staerk J, Delhommeau F, Lacout C, et al. A unique clonal JAK2 mutation leading to constitutive signalling causes polycythaemia vera. Nature 2005;434:1144–8. [133] Kralovics R, Passamonti F, Buser AS, Teo SS, Tiedt R, Passweg JR, et al. A gain-of-function mutation of JAK2 in myeloproliferative disorders. N Engl J Med 2005;352:1779–90. [134] Levine RL, Wadleigh M, Cools J, Ebert BL, Wernig G, Huntley BJ, et al. Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocytosis, and myeloid metaplasia with myelofibrosis. Cancer Cell 2005;7:387–97.

H.C. Hasselbalch, C.H. Riley / Leukemia Research 30 (2006) 1217–1225 [135] Jones AV, Kreil S, Zoi K, Waghorn K, Curtis C, Zhang L, et al. Widespread occurrence of the JAK2 V617F mutation in chronic myeloproliferative disorders. Blood 2005;106:2162–2168. [136] Parganas E, Wang D, Stravopodis D, Topham DJ, Marine JC, Teglund S, et al. Jak2 is essential for signaling through a variety of cytokine receptors. Cell 1998;93:385–95. [137] Reinmuth N, Fan F, Liu W, Parikh AA, Stoeltzing O, Jung YD, et al. Impact of insulin-like growth factor receptor-1 function on angiogenesis, growth and metastasis of colon cancer. Lab Invest 2002;82:1377–89. [138] Fruchtman SM. Treatment paradigms in the management of chronic myeloproliferative disorders. Semin Hematol 2004;41:18–22. [139] Pucetti L, Sawamura T, Pasqui AL, Patorelli M, Auteri A, Bruni F. Atorvastatin reduces platelet-oxidized-LDL receptor expression in hypercholesterolaemic patients. Eur J Clin Invest 2005;35:47–51. [140] Laufs U, La Fata V, Plutzky J, Liao JK. Upregulation of endothelial nitric oxide synthase by HMG CoA reductase inhibitors. Circulation 1998;97:1129–35. [141] Landolphi R, Marchioli R, Kutti J, Gisslinger H, Tognoni G, Patrono C, et al., European Collaboration on Low-Dose Aspirin in Polycythemia Vera. Efficacy and safety of low dose aspirin in polycythemia vera. N Engl J Med 2004;350:114–24. [142] Falanga A, Marchetti M, Vignoli A, Balducci D, Barbui T. Leucocyte–platelet interaction in patients with essential thrombocythemia and polycythemia vera. Exp Hematol 2005;33:523–30. [143] de Gaetoano G, Cerletti C, Evangeslista V. Recent advances in platelet–polymorphonuclear leucocyte interaction. Haemostasis 1999;29:41–9. [144] Jensen MK, de Nully Brown P, Lund BV, Nielsen OJ, Hasselbalch HC. Increased circulating platelet–leucocyte aggregates in myeloproliferative disorders is correlated to previous thrombosis, platelet activation and platelet count. Eur J Haematol 2001;66:143–51.

1225

[145] Kaneider NC, Egger P, Dunzendorfer S, Wiedermann CJ. Rho-GTPase-dependent platelet–neutrophil interaction affected by HMG-CoA reductase inhibition with altered adenosine nucleotide release and function. Arterioscler Thromb Vasc Biol 2002;22:1029–35. [146] Landolphi R, Ciabattoni G, Patrignani P, Castellana MA, Pogliani E, Bizzi B, et al. Increased thromboxane biosynthesis in patients with polycythaemia vera: evidence for aspirin suppressible platelet activation in vivo. Blood 1992;80:1965–71. [147] Rocca B, Ciabattoni G, Tartaglione R, Cortelazzo S, Barbui T, Patrono C, et al. Increased thromboxane biosynthesis in essential thrombocythemia. Thromb Haemost 1995;74:1225–30. [148] Szczelik A, Musial J, Undas A, Gajewski P, Gora P, Swadazba J, et al. Inhibition of thrombin generation by simvastatin and lack of additive effects of aspirin in patients with marked hypercholesterolemia. J Am Coll Cardiol 1999;33:1286–93. [149] Munford RS. Statins and the acute-phase response. N Engl J Med 2001;344:2016–20. [150] Ridker PM, Rifai N, Clearfield M, Downs JR, Weis SE, Miles JS, et al. Measurement of C-reactive protein for the targeting of statin therapy in the primary prevention of acute coronary events. N Engl J Med 2001;344:1959–65. [151] Nissen SE, Tuzcu EM, Schoenhagen P, Crowe T, Sasiela WJ, Tsai J, et al. Statin therapy, LDL cholesterol, C-reactive protein, and coronary artery disease. N Engl J Med 2005;352:29–38. [152] Ridker PM, Cannon CP, Morrow D, Rifai N, Rose LM, McCabe CH, et al. C-reactive levels and outcomes after statin therapy. N Engl J Med 2005;352:20–8. [153] Vincenzi B, Santini D, Avvisati G, Baldi A, La Cesa A, Tonini G. Statins may potentiate biphosphonates anticancer properties: a new pharmacological approach? Med Hypotheses 2003;61:98– 101.