当前位置: 首页 > 期刊 > 《临床肿瘤学》 > 2005年第11期 > 正文
编号:11332120
Pathogenesis of Myelofibrosis With Myeloid Metaplasia
http://www.100md.com 《临床肿瘤学》
     the Division of Hematology, Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN

    ABSTRACT

    The primary disease process in myelofibrosis with myeloid metaplasia (MMM) is clonal myeloproliferation with varying degrees of phenotypic differentiation. This is characteristically accompanied by secondary intramedullary collagen fibrosis, osteosclerosis, angiogenesis, and extramedullary hematopoiesis. Modern clonality studies have confirmed the multipotent stem-cell origin of the neoplastic process in MMM. The nature of the specific oncogenic mutation(s) is currently being unraveled with the recent discovery of an association between a somatic point mutation of JAK2 tyrosine kinase (V617F) and bcr/abl-negative myeloproliferative disorders, including MMM. The pathogenetic mechanisms that underlie the secondary bone marrow stromal changes in MMM are also incompletely understood. Mouse models of this latter disease aspect have been constructed by either in vivo overexpression of thrombopoietin (TPOhigh mice) or megakaryocyte lineage restricted underexpression of the transcription factor GATA-1 (GATA-1low mice). Gene knockout experiments using such animal models have suggested the essential role of hematopoietic cell-derived transforming growth factor beta1 in inducing bone marrow fibrosis and stromal cell–derived osteoprotegerin in promoting osteosclerosis. However, experimental myelofibrosis in mice does not recapitulate clonal myeloproliferation that is fundamental to human MMM. Other cytokines that are implicated in mediating myelofibrosis and angiogenesis in MMM include basic fibroblast, platelet-derived, and vascular endothelial growth factors. It is currently assumed that such cytokines are abnormally released from clonal megakaryocytes as a result of a pathologic interaction with neutrophils (eg, emperipolesis). This latter phenomenon, through neutrophil-derived elastase, could also underlie the abnormal peripheral-blood egress of myeloid progenitors in MMM.

    INTRODUCTION

    Myelofibrosis with myeloid metaplasia (MMM) was first described in 18791 and is currently classified as a myeloproliferative disorder (MPD).2 The disease presents either de novo (agnogenic myeloid metaplasia) or in the setting of both polycythemia vera (PV; postpolycythemic myeloid metaplasia) and essential thrombocythemia (ET; post-thrombocythemic myeloid metaplasia).3,4 The clinical phenotype includes progressive anemia, massive splenomegaly, a leukoerythroblastic blood smear, profound constitutional symptoms, cachexia, and hepatosplenic as well as nonhepatosplenic extramedullary hematopoiesis.5 Bone marrow histology displays substantial collagen fibrosis, osteosclerosis, and angiogenesis.6 Most patients are diagnosed after age 60 years,7 and reported median survival ranges from 3.5 to 10 years.8-11 Causes of death include infection, bleeding, organ failure, portal hypertension, and leukemic transformation.9-11 The latter occurs in 8% to 23% of the patients in the first decade of the disease. Current drug therapy has not altered the natural history of the disease.12 Although the prospect of cure is possible with allogeneic hematopoietic stem-cell transplantation, the majority of affected patients are not suitable candidates for the particular treatment modality because of either advanced age or the presence of comorbid conditions.13 Therefore, rational treatment approaches in MMM await elucidation of the underlying pathogenetic mechanisms of both clonal myeloproliferation14 and the reactive bone marrow stromal changes that are believed to be cytokine mediated.15

    CLONALITY STUDIES

    Normal hematopoiesis in large animals is polyclonal, and individual multipotent stem cells are capable of both myeloid and lymphoid lineage differentiation.16 In contrast, hematopoiesis in patients with MPD, including those with MMM, is monoclonal and is supported by a genetically transformed mutant clone that retains the capacity to differentiate across multiple cell lineages.17 In general, clonality studies in MPD have mostly been based on allelic polymorphisms between the paternally and maternally derived X chromosomes in females.18 In a normal (polyclonal) somatic cell population, the random X chromosome inactivation process has affected both the maternal and paternal X chromosomes.19 In a clonal cell population, in contrast, because of the single-cell derivation of the neoplastic process, only the maternal or the paternal X chromosome is affected (not both). This difference in X chromosome inactivation pattern between so-called normal and clonal cell populations can be demonstrated in informative (heterozygous at the genetic locus of interest) females, at either the DNA (on the basis of methylation differences between active and inactive genes),20 or post-DNA level (on the basis of the fact that RNA and enzyme expression is restricted to the active chromosome).18,21,22 Some of the classic studies in this regard have used glucose-6-phosphate dehydrogenase isoenzyme analysis (ie, protein level investigation),14,23,24 whereas more recent studies have focused on X-linked DNA25,26 and transcript21 analysis.

    Monoclonality in MMM was suggested as early as 1968 based on the clonal occurrence of an abnormal chromosome marker.27 Subsequent glucose-6-phosphate dehydrogenase–based clonality studies in the 1970s documented clonal involvement of bone marrow mononuclear cells as well as peripheral-blood granulocytes, erythrocytes, and platelets, but not bone marrow fibroblasts.14,28 Similarly, X-linked DNA analysis has revealed monoclonal X chromosome inactivation pattern in peripheral-blood leukocytes,29 granulocytes,26,30 bone marrow mononuclear cells,26 and, in some cases, T lymphocytes.30 These observations were supported by cytogenetics-based demonstration of clonality in erythroid,31,32 granulocyte-monocyte,31,32 and granulocyte-monocyte-erythroid progenitors,32 but not in fibroblasts.33,34 Clonality in MMM has also been investigated using N-ras mutation as a marker and showing its presence in granulocytes, monocytes, and erythroblasts, as well as B and T lymphocytes.35 Similarly, a recent study used a combination of immunomagnetic cell separation technique and interphase cytogenetics to demonstrate clonal involvement of T and B lymphocytes (Fig 1). 17 Therefore, current evidence strongly supports the true stem-cell nature of the mutant clone in MMM, and within the confines of the available testing methods, interpatient heterogeneity in the extent of clonal involvement by a specific cell type has been demonstrated.17,30

    THE SEARCH FOR DISEASE-SPECIFIC MUTATIONS

    Cytogenetic Studies

    Despite the abundant evidence for clonal hematopoiesis in MMM, the nature of the disease-causing genetic mutation remains elusive. The detection of a consistent cytogenetic abnormality in myeloid malignancies is not only diagnostically useful,36,37 but may also provide pathogenetic clues.38,39 Unfortunately, the majority of myeloid disorders do not bear a specific karyotypic marker, and observed abnormalities often represent secondary subclones possibly linked to the genetic instability of the mutant clone.40,41 Nevertheless, the cytogenetics of MMM has been extensively studied, and a wide spectrum of structural and numerical chromosome abnormalities have been described.42-63 In general, recurrent cytogenetic abnormalities occur in approximately 50% of chemotherapy-naive patients with MMM, and the most prevalent lesions, representing more than 80% of the abnormal cases, include del(20)(q11;q13), del(13)(q12;q22), trisomy 8, trisomy 9, del(12)(p11;p13), monosomy or long-arm deletions involving chromosome 7, and partial trisomy 1q (Fig 2). 57,63 However, the individual lesions occur in only the minority of patients (10% to 25% of the abnormal cases), and none are specific to MMM (Fig 2).63 Furthermore, the application of molecular cytogenetic studies by fluorescent in situ hybridization did not disclose additional, karyotypically occult, structural lesions.64 Incidentally, the pattern of cytogenetic abnormalities is similar between the three subtypes of MMM: agnogenic myeloid metaplasia, postpolycythemic myeloid metaplasia, and post-thrombocythemic myeloid metaplasia.63

    As mentioned above, although not disease specific, 13q-, 20q-, and abnormalities of chromosome 1 are characteristically prevalent in MMM and constitute a focus of interest regarding pathogenetic insight. Molecular characterization of 13q- in bcr/abl-negative MPD has revealed microdeletions that did not involve the retinoblastoma (RB1) tumor suppressor gene locus,65 whereas the RB1 region is commonly deleted in 13q- associated with multiple myeloma.66,67 Consistent with this observation, the particular cytogenetic abnormality seems to confer good prognosis in MMM but is associated with adverse prognosis in multiple myeloma.63,68 In regard to 20q-, similar molecular studies have suggested commonly deleted regions that were specific to either MPD or myelodysplastic syndrome/acute myeloid leukemia.69 Accordingly, further inquiry into the specific genes that are located in the commonly deleted regions of MPD-associated 20q- might shed additional pathogenetic insight on MMM. Partial trisomy 1 that is associated with certain derivative chromosomes is arguably the most specific cytogenetic abnormality in MMM. Interestingly, one particular such lesion, der(6)t(1;6) (q21-23;p21-23), involves a chromosomal region (6p21) that houses the FK-506 binding protein 5 (FKBP51) gene, which was recently shown to be overexpressed in CD34+ cell-derived megakaryocytes of patients with MMM and potentially confer an antiapoptotic effect through inhibition of calcineurin.70 Molecular studies are currently underway to map the precise breakpoints of these intriguing translocations and learn their pathogenetic role in MMM.

    Mutation Screening Studies

    The pathogenesis of MMM and related bcr/abl-negative MPD might be explained, in part, by a somatic point mutation on exon 14 (V617F) of the JAK2 kinase gene that is located on chromosome 9p24.71-73 The recurrent JAK2 mutation was identified by either a candidate gene approach71,73 or a high-throughput DNA sequencing of the functional domains of 85 human tyrosine kinases.72 The incidence of the mutant allele in granulocytes collected from MPD patients was 35% to 50% in MMM, 32% to 57% in ET, and 74% to 97% in PV.72,73 No mutation was detected in either normal individuals or patients with secondary erythrocytosis.71-73 The JAK2 V617F mutation occurs within the auto-inhibitory JH2 domain, resulting in dysregulation of kinase activity that resides in the JH1 domain. Supporting evidence for this comes from the demonstration of mutant allele-mediated activation of both JAK2- and STAT5-mediated transcription, as well as the induction of either Epo hypersensitivity or growth factor independence in cell lines.71,72 Furthermore, erythrocytosis was induced in mice transplanted with bone marrow cells carrying the mutant allele.71 However, direct mutation screening for other candidate genes, including type III receptor tyrosine kinases (c-kit, c-fms, flt-3), has generally yielded negative results.74,75

    Loss-of-Heterozygosity Studies

    In certain human cancers, loss of heterozygosity (LOH) of chromosomal regions harboring mutated tumor suppressor genes is either a key oncogenic event or contributes to disease progression. Genome-wide patterns of LOH can be studied using polymorphic genetic markers, and the principle behind the particular molecular investigation is that the loss of the normal allele of a tumor suppressor gene in the presence of a mutated allele results in homozygous loss of protective gene function.76

    In a recent study, a genome-wide screening for LOH using 86 polymorphic markers encompassing all 22 autosomes was performed on CD34+ cells obtained from 29 patients with MMM.77 Allelic loss was compared with that of normal T lymphocytes from the same patients. After combing the results of marker studies on the same arm of individual chromosomes, the most frequent allelic loss was observed on 1q (25%), 3p (24%), and 3q (22%).77 Interestingly, the LOH patterns were not correlated with structural lesions detected by cytogenetic studies. Using 23 additional LOH markers, a detailed mapping of the chromosome 3p arm between D3S1597 and D3S1578 revealed a CDR between D3S1583 and D3S1609 at 3p24 that houses the retinoic acid receptor tumor suppressor gene. Subsequent experiments revealed that RAR2 gene expression was markedly decreased in all tested patients with MMM, even in the absence of LOH at the particular locus. Additional experiments suggested that this was the result of an epigenetic mechanism mediated by abnormal methylation of the gene promoter P2.77 Although RAR is considered important in myeloid differentiation, additional studies are needed to decipher the specific pathogenetic role of the observed phenomenon in MMM. In the meantime, the particular work provides support for evaluating the therapeutic value of demethylating as well as retinoic acid-based agents in MMM.78,79

    Gene Expression Studies

    In gene expression analysis, an altered gene or biochemical pathway associated with a particular disease may be revealed by the identification of a consistently upregulated or downregulated gene across a cohort of patients with the same disease.80 Global gene expression profiling has been shown to accurately distinguish among previously established phenotypic categories of hematologic malignancies as well as reveal additional molecular classes.81 More importantly, considerable pathogenetic insight may be obtained by comparing gene expression patterns of diseased and normal tissue.

    To date, only one study has evaluated MMM by oligonucleotide microarray analysis.82 Gene expression by CD34+ cells in eight patients with MMM was compared with that of six normal controls. Upregulated genes in MMM included several transcription factors (JUNB at 19p13.2, GATA2 at 3q21, N-myc at 2p24.1), granulocyte-macrophage colony-stimulating factor (CSF1 at 5q33), interleukin-8 (IL8 at 4q13-21), IL1B at 2q14, platelet-derived endothelial cell growth factor (PDECGF at 22q13.33), platelet factor 4 at 4q12-21, and delta, Drosophila homolog-like 1 (DLK1 at 14q32). Downregulated genes included connective tissue growth factor (CTGF) that is located on chromosome 6q23.1, osteopontin (OPN) located at 4q21-25, FK-506 binding protein 5 (FKBP5) located at 6p21, bone morphogenic protein 2B (BMP2B) located at 14q22-23, macrophage inhibitory cytokine-1 (MIC-1) located at 19p13, BCL-1 located at 11q13, cell division cycle 2 (cdc2) located 10q21.1, and cdc20 located at 9q13-21.

    Some of the upregulated transcription factors (eg, N-myc) may be relevant to cell cycle progression and, along with some of the overexpressed growth factors (eg, CSF1, PDECGF) and pleiotropic cytokines (eg, IL-8, IL-1B), might contribute to the myeloproliferation and stromal reaction in MMM.83-85 Dlk1 is a transmembrane protein belonging to the superfamily of epidermal growth factor–like proteins and is essential for normal hematopoiesis, and its abnormal expression has also been demonstrated in myelodysplastic syndrome.86 Osteopontin is a secreted cell attachment phosphoprotein that might participate in cell-matrix interaction and might facilitate fibroblast, osteoblast, and osteoclast contribution to the pathology of the stromal reaction in MMM.87 BMP2B might play a similar role in the associated changes in bone remodeling.88 The results of the aforementioned DNA microarray study regarding FKBP51 gene expression is at odds with another study that used a differential display approach and demonstrated overexpression in megakaryocytes derived from CD34+ cells of patients with MMM.70 The discrepancy might have arisen from either the distinct cell populations studied in the two different reports (peripheral-blood CD34+ cells82 v CD34-derived megakaryocytes70) or the associated experimental conditions in the latter study (comparison of cytokine-prepared normal megakaryocytes v spontaneously grown MMM megakaryocytes).70 Regardless, the systematic evaluation of data coming out of various types of gene expression studies, including DNA microarray analysis, should help generate specific hypothesis for more focused molecular investigations.

    Other Molecular and Biologic Studies

    Another biologic feature of megakaryocytes in MMM is their endogenous (cytokine-independent) growth capability as well as their hypersensitivity to various growth factors, including thrombopoietin (TPO).89-94 The specific in vitro growth characteristic might also affect other cell lineages95,96 and is not specific to MMM, as it is readily seen in other MPD, including PV97-99 and ET.96,100-102 Such growth factor independence is not seen in either normal subjects or reactive myeloproliferation103 and, in MPD, has not been attributed to mutations in ligand receptor89,104 or receptor-associated signal transducer molecules.105 One suggested mechanism of intrinsic growth factor hypersensitivity in MMM involves megakaryocyte overexpression of FKBP51 that is associated with constitutive JAK2/STAT5 activation.70,106 This possibility is consistent with the recent discovery of an activation mutation of JAK2 (V617F) in MPD.71-73 However, megakaryocyte FKBP51 gene as well as protein expression in MMM was shown to be similar to that of normal megakaryocytes in another study.107

    Another study has suggested the constitutive activation of the c/JAK3/STAT3 pathway in CD34+ cells from patients with MMM that was coupled with the inability to differentiate into natural-killer cells.108 Obviously, such examples of altered cellular biology are by no means specific to MMM and could represent a nonspecific feature that is common to many neoplastic cell processes.109,110 The same can be said about the stem-cell leukemia gene, the expression of which in blood mononuclear cells from patients with MMM was shown to be increased.111 Markedly decreased megakaryocyte/platelet expression of the TPO receptor (Mpl) is another nonspecific112,113 cellular feature in MMM114 that is also seen in ET115 and PV.114 The particular phenomenon has been blamed for the normal or elevated serum concentrations of TPO in MMM116 that is unexpected in lieu of the disease-associated increased megakaryocyte mass.

    PATHOGENETIC MECHANISM OF BONE MARROW STROMAL REACTION

    MMM is typically characterized by bone marrow collagen fibrosis, new bone formation (osteosclerosis), and angiogenesis.6,117 As elaborated in the previous section on clonality studies, bone marrow fibroblasts in MMM are polyclonal33,34,118 and exhibit normal function and in vitro growth characteristics.118 This information, coupled with the observation that both cellular and extracellular levels of various fibrogenic and angiogenic cytokines are altered in patients with MMM, strongly supports the contention that the bone marrow histologic changes in MMM are reactive and cytokine mediated.118-120 In this regard, several patient-centered studies have suggested a pathogenetic role for transforming growth factor beta (TGF-?),121-123 platelet-derived growth factor,123 basic fibroblast growth factor,124-126 vascular endothelial growth factor (VEGF),127 and tissue inhibitors of matrix metalloproteinases.128,129 Similar circumstantial evidence has long suggested that megakaryocytes122,130 and activated monocytes120 might be the source of the aforementioned nosogenic cytokines. Furthermore, a pathologic interaction between megakaryocytes and neutrophils (emperipolesis), induced by altered megakaryocyte P-selectin distribution, might underlie the abnormal cytokine release mechanism.131,132 Neutrophil-derived enzymes, including elastase, might also participate in the pathologic process by facilitating peripheral-blood egress of myeloid progenitors.133-135

    Mouse models of myelofibrosis have provided additional information regarding the role of certain cytokines in the pathogenesis of the stromal reaction in MMM.136,137 There are currently two established models of experimental myelofibrosis in mice: TPOhigh and GATA-1low mice. The former is constructed by the systemic overexpression of thrombopoietin137-140 and the latter by megakaryocyte lineage restricted underexpression of the transcription factor GATA-1.141 The common feature to both experimental models is tissue accumulation of megakaryocytes, a result of TPO-driven proliferation in TPOhigh mice142 and impaired megakaryocyte maturation in GATA-1low mice.143 However, considerable heterogeneity exists among different mouse models in terms of both phenotype and disease tempo (Fig 3). 138,140,141,144-146 In immune-deficient mice, myelofibrosis and osteosclerosis developed only in the severe combined immunodeficient (T and B cell deficient) model attributed to both the high degree of TPO expression, compared with the nude (T cell deficient) model, as well as the retention of intact monocyte-macrophage function, compared with the nonobese diabetic severe combined immunodeficient (T and B as well as multiple other defects in innate immunity, including NK and monocyte-macrophage function) model.146,147 Furthermore, experimental myelofibrosis in TPOhigh mice has been shown to be reversible either by transplantation139 or cessation of the systemic administration of TPO.148

    The role of megakaryocyte-derived TGF-? in myelofibrosis was first suggested by the in vitro observation that the addition of a neutralizing anti–TGF-? antibody inhibited the stimulatory effects of a megakaryoblast-conditioned medium on collagen synthesis in bone marrow fibroblasts from a patient with acute megakaryocytic leukemia.121 Interestingly, the particular cytokine has also been implicated in hairy cell leukemia–associated reticulin and collagen fibrosis.149 Consistent with these observations, TGF-?1 null murine hematopoietic stem cells retrovirally infected with murine TPO failed to produce bone marrow fibrosis when transplanted into lethally irradiated wild-type mice, despite the development of all other components of the complete phenotype.136 Similarly, the osteosclerotic component of experimental myelofibrosis in TPOhigh mice is aborted by knocking out the stromal cell-derived osteoprotegerin gene.150 Similarly, an age-dependent increase in osteoprotegerin accompanied the development of osteosclerosis in one of the aforementioned transgenic TPO models.145 TGF-?1 seems to be similarly important in an the alternative model of GATA-1low mice.141,151 In this instance, treatment with TPO improved the associated thrombocytopenia as well as stabilized bone marrow fibrosis through downregulation of TGF-?1 expression.152

    THERAPEUTIC IMPLICATIONS

    The aforementioned pathogenetic information regarding the stromal reaction in MMM underlies the rationale for testing a series of drugs at my institution over the last 12 years. Accordingly, specific drugs that have been evaluated included those that were directed at reducing megakaryocyte bulk (interferon alfa-2a,153 cladribine154,155), impairing megakaryocyte differentiation and thus possibly interfering with megakaryocyte-neutrophil interaction (anagrelide156), inhibiting TGF-?–mediated fibroblast proliferation and collagen synthesis (suramin,157 pirfenidone158), inhibiting platelet-derived growth factor receptor–associated tyrosine kinase activity (imatinib mesylate159), and interfering with angiogenesis and TNF- production (thalidomide,160-163 etanercept164). Unfortunately, none of these agents were shown to induce favorable changes in bone marrow stroma, although clinical benefit was demonstrated with cladribine, thalidomide, and etanercept.155,160,164 On the basis of these findings, we are currently running treatment trials with a more potent thalidomide analog (CC-5013; lenalidomide; Revlimid; Celgene, Summit, NJ) as well as combination therapy with thalidomide and etanercept. Revlimid is the lead compound among the immunomodulatory analogs of thalidomide (ImiDs), and its ex vivo antiangiogenic as well as anti-tumor necrosis factor property is estimated to be at least 50-fold higher than that of thalidomide.165,166 Within the context of myeloid malignancies, the drug was recently shown to have excellent therapeutic activity in MDS associated with a 5q- chromosomal abnormality.167

    There are several other antiangiogenic agents that might be considered for future therapeutic trials in MMM. In this regard, we have recently completed a phase II study (unpublished) with a farnesyl transferase inhibitor (R115777) that is known to downregulate VEGF expression.168 Some promise of therapeutic activity in this regard has already been communicated by other investigators.169 In another upcoming pilot study, we plan to evaluate the therapeutic activity of bortezomib,170 a proteosome inhibitor, which in addition to its indirect antiangiogenic effect, might overcome the antiapoptotic effect of FKBP51 overexpression in MMM.70,106 Other antiangiogenic agents that are undergoing clinical trials and are potent inhibitors of VEGF receptors, platelet-derived growth factor receptor, and Kit include SU-5416, SU-6668, and PTK-787.171-173 In general, these agents were well tolerated, but their activity has so far been underwhelming.171,173 Rapamycin is another antiangiogenic drug174 that might be worth looking into because it also interferes with collagen synthesis175 and abnormal granulocyte migration.135,176

    Another venue of therapeutic trials might target the downstream effectors of TGF-?.177 One such candidate molecule is connective tissue growth factor,178,179 and human monoclonal antibodies against connective tissue growth factor are already being evaluated in clinical trials involving other fibrotic disorders.180 One can also envision treatment molecules that target either the TGF-? receptor181,182 or the postreceptor signaling intermediates, the Smad proteins.183,184 In the end, it is likely that a combination of drugs directed at different molecules might be needed for effective control of the composite stromal aberration in MMM. In this regard, the recently discovered, MPD-associated JAK2 V617F might constitute a molecular target for the development of rational drug therapy.185

    In summary, based on the composite set of information obtained from laboratory investigation of patient samples as well as experimental myelofibrosis in mice, a working model of the stromal reaction in MMM is proposed (Fig 4). In this model, the central event is the accumulation of bone marrow megakaryocytes that is clonal17 in man but cytokine-mediated in experimental myelofibrosis.137-141 These megakaryocytes, both in man and mice, are also qualitatively abnormal and exhibit abnormal distribution of P-selectin131,132 and decreased expression of Mpl.114 The former abnormality promotes a pathologic interaction with neutrophils (emperipolesis) and the latter an in situ increase in TPO concentration. The excess TPO might enhance the underlying clonal myeloproliferation as well as induce stromal cells to produce fibrogenic, osteogenic, and angiogenic cytokines. Such cytokines are also abnormally released as a result of the aforementioned emperipolesis as well as activation of clonal monocytes.120 The different components of the overall stromal reaction (fibrosis, osteosclerosis, angiogenesis, CD34 cell egress) might be linked to one or more specific cytokines.136,150

    It is underscored that experimental myelofibrosis in mice does not recapitulate clonal myeloproliferation that is fundamental to human MMM.17 However, it does capture the stromal aberration that is integral to the human disease.186-193 Similarly, alterations in either gene structure or gene expression involving TPO,194 c-Mpl,89,195 GATA-1,196,197 or FOG-1 (the transcriptional regulator of GATA-1)198 have not been detected in human MMM. Regardless, the availability of animal models of myelofibrosis should facilitate in vivo preclinical drug testing as well as identification of crucial molecules that can be therapeutically targeted.

    Author's Disclosures of Potential Conflicts of Interest

    The author indicated no potential conflicts of interest.

    NOTES

    Author's disclosures of potential conflicts of interest are found at the end of this article.

    REFERENCES

    Heuck G: Zwei Falle von Leukamie mit eigenthumlichem Blut- resp: Knochenmarksbefund. Arch Pathol Anat Physiol Virchows 78:475-496, 1879

    Dameshek W: Some speculations on the myeloproliferative syndromes. Blood 6:372-375, 1951

    Passamonti F, Malabarba L, Orlandi E, et al: Polycythemia vera in young patients: A study on the long-term risk of thrombosis, myelofibrosis and leukemia. Haematologica 88:13-18, 2003

    Cervantes F, Alvarez-Larran A, Talarn C, et al: Myelofibrosis with myeloid metaplasia following essential thrombocythaemia: Actuarial probability, presenting characteristics and evolution in a series of 195 patients. Br J Haematol 118:786-790, 2002

    Tefferi A: Myelofibrosis with myeloid metaplasia. N Engl J Med 342:1255-1265, 2000

    Mesa RA, Hanson CA, Rajkumar SV, et al: Evaluation and clinical correlations of bone marrow angiogenesis in myelofibrosis with myeloid metaplasia. Blood 96:3374-3380, 2000

    Mesa RA, Silverstein MN, Jacobsen SJ, et al: Population-based incidence and survival figures in essential thrombocythemia and agnogenic myeloid metaplasia: An Olmsted County study, 1976-1995. Am J Hematol 61:10-15, 1999

    Dupriez B, Morel P, Demory JL, et al: Prognostic factors in agnogenic myeloid metaplasia: A report on 195 cases with a new scoring system. Blood 88:1013-1018, 1996

    Cervantes F, Pereira A, Esteve J, et al: Identification of ‘short-lived’ and ‘long-lived’ patients at presentation of idiopathic myelofibrosis. Br J Haematol 97:635-640, 1997

    Rupoli S, Da Lio L, Sisti S, et al: Primary myelofibrosis: A detailed statistical analysis of the clinicopathological variables influencing survival. Ann Hematol 68:205-212, 1994

    Okamura T, Kinukawa N, Niho Y, et al: Primary chronic myelofibrosis: Clinical and prognostic evaluation in 336 Japanese patients. Int J Hematol 73:194-198, 2001

    Tefferi A: Treatment approaches in myelofibrosis with myeloid metaplasia: The old and the new. Semin Hematol 40:18-21, 2003

    Deeg HJ, Gooley TA, Flowers ME, et al: Allogeneic hematopoietic stem cell transplantation for myelofibrosis. Blood 102:3912-3918, 2003

    Jacobson RJ, Salo A, Fialkow PJ: Agnogenic myeloid metaplasia: A clonal proliferation of hematopoietic stem cells with secondary myelofibrosis. Blood 51:189-194, 1978

    Groopman JE: The pathogenesis of myelofibrosis in myeloproliferative disorders. Ann Intern Med 92:857-858, 1980

    Kim HJ, Tisdale JF, Wu T, et al: Many multipotential gene-marked progenitor or stem cell clones contribute to hematopoiesis in nonhuman primates. Blood 96:1-8, 2000

    Reeder TL, Bailey RJ, Dewald GW, et al: Both B and T lymphocytes may be clonally involved in myelofibrosis with myeloid metaplasia. Blood 101:1981-1983, 2003

    Beutler E, Collins Z, Irwin LE: Value of genetic variants of glucose-6-phosphate dehydrogenase in tracing the origin of malignant tumors. N Engl J Med 276:389-391, 1967

    Goto T, Monk M: Regulation of X-chromosome inactivation in development in mice and humans. Microbiol Mol Biol Rev 62:362-378, 1998

    Vogelstein B, Fearon ER, Hamilton SR, et al: Use of restriction fragment length polymorphisms to determine the clonal origin of human tumors. Science 227:642-645, 1985

    Prchal JT, Guan YL: A novel clonality assay based on transcriptional analysis of the active X chromosome. Stem Cells 11:62-65, 1993 (suppl 1)

    Gilliland DG, Blanchard KL, Levy J, et al: Clonality in myeloproliferative disorders: Analysis by means of the polymerase chain reaction. Proc Natl Acad Sci U S A 88:6848-6852, 1991

    Fialkow PJ, Faguet GB, Jacobson RJ, et al: Evidence that essential thrombocythemia is a clonal disorder with origin in a multipotent stem cell. Blood 58:916-919, 1981

    Adamson JW, Fialkow PJ, Murphy S, et al: Polycythemia vera: Stem-cell and probable clonal origin of the disease. N Engl J Med 295:913-916, 1976

    Elkassar N, Hetet G, Briere J, et al: Clonality analysis of hematopoiesis in essential thrombocythemia: Advantages of studying T lymphocytes and platelets. Blood 89:128-134, 1997

    Kreipe H, Jaquet K, Felgner J, et al: Clonal granulocytes and bone marrow cells in the cellular phase of agnogenic myeloid metaplasia. Blood 78:1814-1817, 1991

    Engel W, Merker H, Schneider G, et al: Clonal occurrence of a chromosome Dq–in myelosclerosis with myeloid metaplasia. Humangenetik 6:335-337, 1968

    Kahn A, Bernard JF, Cottreau D, et al: Gd(–) Abrami: A deficient G-6PD variant with hemizygous expression in blood cells of a woman with primary myelofibrosis. Humangenetik 30:41-46, 1975

    Anger B, Janssen JW, Schrezenmeier H, et al: Clonal analysis of chronic myeloproliferative disorders using X-linked DNA polymorphisms. Leukemia 4:258-261, 1990

    Tsukamoto N, Morita K, Maehara T, et al: Clonality in chronic myeloproliferative disorders defined by X-chromosome linked probes: Demonstration of heterogeneity in lineage involvement. Br J Haematol 86:253-258, 1994

    Ruutu T, Partanen S, Knuutila S: Clonal karyotype abnormalities in erythroid and granulocyte-monocyte precursors in polycythaemia vera and myelofibrosis. Scand J Haematol 31:253-256, 1983

    Sugiyama H, Ichiba S, Okuno Y, et al: Cytogenetic evidence for a clonal disorder involving CFU-GEMM, BFU-E and CFU-C in patients with myeloproliferative disorders. Nippon Ketsueki Gakkai Zasshi 52:1022-1032, 1989

    Greenberg BR, Woo L, Veomett IC, et al: Cytogenetics of bone marrow fibroblastic cells in idiopathic chronic myelofibrosis. Br J Haematol 66:487-490, 1987

    Wang JC, Lang HD, Lichter S, et al: Cytogenetic studies of bone marrow fibroblasts cultured from patients with myelofibrosis and myeloid metaplasia. Br J Haematol 80:184-188, 1992

    Buschle M, Janssen JW, Drexler H, et al: Evidence for pluripotent stem cell origin of idiopathic myelofibrosis: Clonal analysis of a case characterized by a N-ras gene mutation. Leukemia 2:658-660, 1988

    Nowell PC, Hungerford DA: A minute chromosome in human chronic granulocytic leukemia. J Natl Cancer Inst 25:85, 1960

    Larson RA, Kondo K, Vardiman JW, et al: Evidence for a 15;17 translocation in every patient with acute promyelocytic leukemia. Am J Med 76:827-841, 1984

    Daley GQ, Van Etten RA, Baltimore D: Induction of chronic myelogenous leukemia in mice by the P210bcr/abl gene of the Philadelphia chromosome. Science 247:824-830, 1990

    de The H, Chomienne C, Lanotte M, et al: The t(15;17) translocation of acute promyelocytic leukaemia fuses the retinoic acid receptor alpha gene to a novel transcribed locus. Nature 347:558-561, 1990

    Lowenberg B, Hagemeijer A, Swart K: Karyotypically distinct subpopulations in acute leukemia with specific growth requirements. Blood 59:641-645, 1982

    Tamura S, Kanamaru A, Takemoto Y, et al: Clonal evolutions during long-term cultures of bone marrow from de novo acute myeloid leukaemia with trilineage myelodysplasia and with myelodysplastic remission marrow. Br J Haematol 84:219-226, 1993

    Nowell P, Jensen J, Gardner F, et al: Chromosome studies in "preleukemia". III. Myelofibrosis. Cancer 38:1873-1881, 1976

    Hsu LY, Pinchiaroli D, Gilbert HS, et al: Partial trisomy of the long arm of chromosome 1 in myelofibrosis and polycythemia vera. Am J Hematol 2:375-383, 1977

    Whang-Peng J, Knutsen T, Chang P, et al: Cytogenetic studies in patients with myelofibrosis and myeloid metaplasia. Leuk Res 2:41-56, 1978

    Nowell P, Finan J: Chromosome studies in preleukemic states, IV. Myeloproliferative versus cytopenic disorders. Cancer 42:2254-2261, 1978

    Geraedts JP, den Ottolander GJ, Ploem JE, et al: An identical translocation between chromosome 1 and 7 in three patients with myelofibrosis and myeloid metaplasia. Br J Haematol 44:569-575, 1980

    Pakkala A, Ruutu T, Partanen S, et al: Peripheral blood cells in the study of chromosome aberrations of patients with chronic myeloid leukaemia or myelofibrosis. Acta Haematol 70:264-268, 1983

    Borgstrom GH, Knuutila S, Ruutu T, et al: Abnormalities of chromosome 13 in myelofibrosis. Scand J Haematol 33:15-21, 1984

    Miller JB, Testa JR, Lindgren V, et al: The pattern and clinical significance of karyotypic abnormalities in patients with idiopathic and postpolycythemic myelofibrosis. Cancer 55:582-591, 1985

    Castoldi G, Cuneo A, Tomasi P, et al: Chromosome abnormalities in myelofibrosis. Acta Haematol 78:104-108, 1987 (suppl 1)

    Smadja N, Krulik M, de Gramont A, et al: Cytogenetic studies in twelve patients with primary myelofibrosis and myeloid metaplasia. Cancer Genet Cytogenet 24:151-158, 1987

    Demory JL, Dupriez B, Fenaux P, et al: Cytogenetic studies and their prognostic significance in agnogenic myeloid metaplasia: A report on 47 cases. Blood 72:855-859, 1988

    Emilia G, Temperani P, Ferrari S, et al: Cytogenetic and molecular studies in primary myelofibrosis. Cancer Genet Cytogenet 38:101-113, 1989

    Donti E, Tabilio A, Bocchini F, et al: Partial trisomy 1q in idiopathic myelofibrosis. Leuk Res 14:1035-1040, 1990

    Mertens F, Johansson B, Heim S, et al: Karyotypic patterns in chronic myeloproliferative disorders: Report on 74 cases and review of the literature. Leukemia 5:214-220, 1991

    Werner M, Nolte M, Kaloutsi V, et al: Karyotype findings and molecular analysis of the bcr gene rearrangement supplementing the histologic classification of chronic myeloproliferative disorders. Lab Invest 72:405-410, 1995

    Reilly JT, Snowden JA, Spearing RL, et al: Cytogenetic abnormalities and their prognostic significance in idiopathic myelofibrosis: A study of 106 cases. Br J Haematol 98:96-102, 1997

    Rege-Cambrin G, Mecucci C, Tricot G, et al: A chromosomal profile of polycythemia vera. Cancer Genet Cytogenet 25:233-245, 1987

    Swolin B, Weinfeld A, Westin J: A prospective long-term cytogenetic study in polycythemia vera in relation to treatment and clinical course. Blood 72:386-395, 1988

    Diez-Martin JL, Graham DL, Petitt RM, et al: Chromosome studies in 104 patients with polycythemia vera. Mayo Clin Proc 66:287-299, 1991

    MacDonald DH, Lahiri D, Chase A, et al: A case of myelofibrosis with a t(4;13)(q25;q12): Evidence for involvement of a second 13q12 locus in chronic myeloproliferative disorders. Br J Haematol 105:771-774, 1999

    Emilia G, Sacchi S, Temperani P, et al: Progression of essential thrombocythemia to blastic crisis via idiopathic myelofibrosis. Leuk Lymphoma 9:423-426, 1993

    Tefferi A, Mesa RA, Schroeder G, et al: Cytogenetic findings and their clinical relevance in myelofibrosis with myeloid metaplasia. Br J Haematol 113:763-771, 2001

    Tefferi A, Meyer RG, Wyatt WA, et al: Comparison of peripheral blood interphase cytogenetics with bone marrow karyotype analysis in myelofibrosis with myeloid metaplasia. Br J Haematol 115:316-319, 2001

    Pastore C, Nomdedeu J, Volpe G, et al: Genetic analysis of chromosome 13 deletions in BCR/ABL negative chronic myeloproliferative disorders. Genes Chromosomes Cancer 14:106-111, 1995

    Tricot G, Barlogie B, Jagannath S, et al: Poor prognosis in multiple myeloma is associated only with partial or complete deletions of chromosome 13 or abnormalities involving 11q and not with other karyotype abnormalities. Blood 86:4250-4256, 1995

    Fonseca R, Oken MM, Harrington D, et al: Deletions of chromosome 13 in multiple myeloma identified by interphase FISH usually denote large deletions of the q arm or monosomy. Leukemia 15:981-986, 2001

    Fonseca R, Harrington D, Oken MM, et al: Biological and prognostic significance of interphase fluorescence in situ hybridization detection of chromosome 13 abnormalities (delta13) in multiple myeloma: An Eastern Cooperative Oncology Group study. Cancer Res 62:715-720, 2002

    Bench AJ, Nacheva EP, Hood TL, et al: Chromosome 20 deletions in myeloid malignancies: Reduction of the common deleted region, generation of a PAC/BAC contig and identification of candidate genes—UK Cancer Cytogenetics Group (UKCCG). Oncogene 19:3902-3913, 2000

    Giraudier S, Chagraoui H, Komura E, et al: Overexpression of FKBP51 in idiopathic myelofibrosis regulates the growth factor independence of megakaryocyte progenitors. Blood 100:2932-2940, 2002

    James C, Ugo V, Le Couedic JP, et al: A unique clonal JAK2 mutation leading to constitutive signaling causes polycythemia vera. Nature 434:1144-1148, 2005

    Levine RL, Wadleigh M, Cools J, et al: Activating mutation in the tyrosine kinase JAK2 in polycythemia vera, essential thrombocythemia, and myeloid metaplasia with myelofibrosis. Cancer Cell 7:387-397, 2005

    Baxter EJ, Scott LM, Campbell PJ, et al: Acquired mutation of the tyrosine kinase JAK2 in human myeloproliferative disorders. Lancet 365:1054-1061, 2005

    Abu-Duhier FM, Goodeve AC, Care RS, et al: Mutational analysis of class III receptor tyrosine kinases (C-KIT, C-FMS, FLT3) in idiopathic myelofibrosis. Br J Haematol 120:464-470, 2003

    Pardanani A, Reeder TL, Kimlinger TK, et al: Flt-3 and c-kit mutation studies in a spectrum of chronic myeloid disorders including systemic mast cell disease. Leuk Res 27:739-742, 2003

    Thiagalingam S, Foy RL, Cheng KH, et al: Loss of heterozygosity as a predictor to map tumor suppressor genes in cancer: Molecular basis of its occurrence. Curr Opin Oncol 14:65-72, 2002

    Jones LC, Tefferi A, Idos GE, et al: RARbeta2 is a candidate tumor suppressor gene in myelofibrosis with myeloid metaplasia. Oncogene 23:7846-7853, 2004

    Villar-Garea A, Esteller M: DNA demethylating agents and chromatin-remodeling drugs: Which, how and why? Curr Drug Metab 4:11-31, 2003

    Kurie JM, Lotan R, Lee JJ, et al: Treatment of former smokers with 9-cis-retinoic acid reverses loss of retinoic acid receptor-beta expression in the bronchial epithelium: Results from a randomized placebo-controlled trial. J Natl Cancer Inst 95:206-214, 2003

    Tefferi A, Bolander ME, Ansell SM, et al: Primer on medical genomics: Part III. Microarray experiments and data analysis. Mayo Clin Proc 77:927-940, 2002

    Ebert BL, Golub TR: Genomic approaches to hematologic malignancies. Blood 104:923-932, 2004

    Jones LC, Tefferi A, Wachsman W, et al: Detection of aberrant signaling pathways in CD34+cells from patients with myelofibrosis with myeloid metaplasia using oligonucleotide microarrays. Blood 98:626a-626a, 2001

    Zajac-Kaye M: Myc oncogene: A key component in cell cycle regulation and its implication for lung cancer. Lung Cancer 34:S43-S46, 2001 (suppl 2)

    Schrader JW: Biological effects of myeloid growth factors. Baillieres Clin Haematol 5:509-531, 1992

    Di Raimondo F, Palumbo GA, Molica S, et al: Angiogenesis in chronic myeloproliferative diseases. Acta Haematol 106:177-183, 2001

    Miyazato A, Ueno S, Ohmine K, et al: Identification of myelodysplastic syndrome-specific genes by DNA microarray analysis with purified hematopoietic stem cell fraction. Blood 98:422-427, 2001

    Denhardt DT, Guo X: Osteopontin: A protein with diverse functions. Faseb J 7:1475-1482, 1993

    Schmitt JM, Hwang K, Winn SR, et al: Bone morphogenetic proteins: An update on basic biology and clinical relevance. J Orthop Res 17:269-278, 1999

    Taksin AL, Couedic JPL, Dusanter-Fourt I, et al: Autonomous megakaryocyte growth in essential thrombocythemia and idiopathic myelofibrosis is not related to a c-mpl mutation or to an autocrine stimulation by Mpl-L. Blood 93:125-139, 1999

    Ozaki S, Kosaka M, Ozaki K, et al: Thrombopoietin-responsive essential thrombocythaemia with myelofibrosis. Br J Haematol 97:449-452, 1997

    Li Y, Hetet G, Maurer AM, et al: Spontaneous megakaryocyte colony formation in myeloproliferative disorders is not neutralizable by antibodies against IL3, IL6 and GM-CSF. Br J Haematol 87:471-476, 1994

    Kobayashi S, Teramura M, Hoshino S, et al: Circulating megakaryocyte progenitors in myeloproliferative disorders are hypersensitive to interleukin-3. Br J Haematol 83:539-544, 1993

    Han ZC, Briere J, Nedellec G, et al: Characteristics of circulating megakaryocyte progenitors (CFU-MK) in patients with primary myelofibrosis. Eur J Haematol 40:130-135, 1988

    Juvonen E: Megakaryocyte colony formation in chronic myeloid leukemia and myelofibrosis. Leuk Res 12:751-756, 1988

    Lutton JD, Levere RD: Endogenous erythroid colony formation by peripheral blood mononuclear cells from patients with myelofibrosis and polycythemia vera. Acta Haematol 62:94-99, 1979

    Juvonen E, Partanen S, Ruutu T: Colony formation by megakaryocytic progenitors in essential thrombocythaemia. Br J Haematol 66:161-164, 1987

    Prchal JF, Axelrad AA: Letter: Bone-marrow responses in polycythemia vera. N Engl J Med 290:1382, 1974

    Fisher MJ, Prchal JF, Prchal JT, et al: Anti-erythropoietin (EPO) receptor monoclonal antibodies distinguish EPO-dependent and EPO-independent erythroid progenitors in polycythemia vera. Blood 84:1982-1991, 1994

    Juvonen E, Partanen S, Ikkala E, et al: Megakaryocytic colony formation in polycythaemia vera and secondary erythrocytosis. Br J Haematol 69:441-444, 1988

    Juvonen E, Ikkala E, Oksanen K, et al: Megakaryocyte and erythroid colony formation in essential thrombocythaemia and reactive thrombocytosis: Diagnostic value and correlation to complications. Br J Haematol 83:192-197, 1993

    Battegay EJ, Thomssen C, Nissen C, et al: Endogenous megakaryocyte colonies from peripheral blood in precursor cell cultures of patients with myeloproliferative disorders. Eur J Haematol 42:321-326, 1989

    Florensa L, Besses C, Woessner S, et al: Endogenous megakaryocyte and erythroid colony formation from blood in essential thrombocythaemia. Leukemia 9:271-273, 1995

    Reid CD: The significance of endogenous erythroid colonies (EEC) in haematological disorders. Blood Rev 1:133-140, 1987

    Hess G, Rose P, Gamm H, et al: Molecular analysis of the erythropoietin receptor system in patients with polycythaemia vera. Br J Haematol 88:794-802, 1994

    Asimakopoulos FA, Hinshelwood S, Gilbert JGR, et al: The gene encoding hematopoietic cell phosphatase (Shp-1) is structurally and transcriptionally intact in polycythemia vera. Oncogene 14:1215-1222, 1997

    Komura E, Chagraoui H, Mansat de Mas V, et al: Spontaneous STAT5 activation induces growth factor independence in idiopathic myelofibrosis: Possible relationship with FKBP51 overexpression. Exp Hematol 31:622-630, 2003

    Bock O, Neusch M, Busche G, et al: Constitutive expression of the FK506 binding protein 51 (FKBP51) in bone marrow cells and megakaryocytes derived from idiopathic myelofibrosis and non-neoplastic haematopoiesis. Eur J Haematol 72:239-244, 2004

    Briard D, Brouty-Boye D, Giron-Michel J, et al: Impaired NK cell differentiation of blood-derived CD34+ progenitors from patients with myeloid metaplasia with myelofibrosis. Clin Immunol 106:201-212, 2003

    Steelman LS, Pohnert SC, Shelton JG, et al: JAK/STAT, Raf/MEK/ERK, PI3K/Akt and BCR-ABL in cell cycle progression and leukemogenesis. Leukemia 18:189-218, 2004

    Boudny V, Kovarik J: JAK/STAT signaling pathways and cancer: Janus kinases/signal transducers and activators of transcription. Neoplasma 49:349-355, 2002

    Steunou V, Le Bousse-Kerdiles MC, Colin-Micouin A, et al: Altered transcription of the stem cell leukemia gene in myelofibrosis with myeloid metaplasia. Leukemia 17:1998-2006, 2003

    Harrison CN, Gale RE, Pezella F, et al: Platelet c-mpl expression is dysregulated in patients with essential thrombocythaemia but this is not of diagnostic value. Br J Haematol 107:139-147, 1999

    Yoon SY, Li CY, Tefferi A: Megakaryocyte c-Mpl expression in chronic myeloproliferative disorders and the myelodysplastic syndrome: Immunoperoxidase staining patterns and clinical correlates. Eur J Haematol 65:170-174, 2000

    Moliterno AR, Hankins WD, Spivak JL: Impaired expression of the thrombopoietin receptor by platelets from patients with polycythemia vera. N Engl J Med 338:572-580, 1998

    Horikawa Y, Matsumura I, Hashimoto K, et al: Markedly reduced expression of platelet C-Mpl receptor in essential thrombocythemia. Blood 90:4031-4038, 1997

    Elliot MA, Yoon SY, Kao P, et al: Simultaneous measurement of serum thrombopoietin and expression of megakaryocyte c-Mpl with clinical and laboratory correlates for myelofibrosis with myeloid metaplasia. Eur J Haematol 68:175-179, 2002

    Tefferi A: The pathogenesis of chronic myeloproliferative diseases. Int J Hematol 73:170-176, 2001

    Castro-Malaspina H, Gay RE, Jhanwar SC, et al: Characteristics of bone marrow fibroblast colony-forming cells (CFU-F) and their progeny in patients with myeloproliferative disorders. Blood 59:1046-1054, 1982

    Castro-Malaspina H, Rabellino EM, Yen A, et al: Human megakaryocyte stimulation of proliferation of bone marrow fibroblasts. Blood 57:781-787, 1981

    Rameshwar P, Denny TN, Stein D, et al: Monocyte adhesion in patients with bone marrow fibrosis is required for the production of fibrogenic cytokines: Potential role for interleukin-1 and TGF-beta. J Immunol 153:2819-2830, 1994

    Terui T, Niitsu Y, Mahara K, et al: The production of transforming growth factor-beta in acute megakaryoblastic leukemia and its possible implications in myelofibrosis. Blood 75:1540-1548, 1990

    Martyre MC, Romquin N, Le Bousse-Kerdiles MC, et al: Transforming growth factor-beta and megakaryocytes in the pathogenesis of idiopathic myelofibrosis. Br J Haematol 88:9-16, 1994

    Martyre MC, Magdelenat H, Bryckaert MC, et al: Increased intraplatelet levels of platelet-derived growth factor and transforming growth factor-beta in patients with myelofibrosis with myeloid metaplasia. Br J Haematol 77:80-86, 1991

    Le Bousse-Kerdiles MC, Chevillard S, Charpentier A, et al: Differential expression of transforming growth factor-beta, basic fibroblast growth factor, and their receptors in CD34+ hematopoietic progenitor cells from patients with myelofibrosis and myeloid metaplasia. Blood 88:4534-4546, 1996

    Le Bousse-Kerdiles MC, Martyre MC, French Irno IM: Involvement of the fibrogenic cytokines, TGF-beta and bFGF, in the pathogenesis of idiopathic myelofibrosis. Pathol Biol (Paris) 49:153-157, 2001

    Martyre MC, Le Bousse-Kerdiles MC, Romquin N, et al: Elevated levels of basic fibroblast growth factor in megakaryocytes and platelets from patients with idiopathic myelofibrosis. Br J Haematol 97:441-448, 1997

    Di Raimondo F, Azzaro MP, Palumbo GA, et al: Elevated vascular endothelial growth factor (VEGF) serum levels in idiopathic myelofibrosis. Leukemia 15:976-980, 2001

    Wang JC, Novetsky A, Chen C, et al: Plasma matrix metalloproteinase and tissue inhibitor of metalloproteinase in patients with agnogenic myeloid metaplasia or idiopathic primary myelofibrosis. Br J Haematol 119:709-712, 2002

    Jensen MK, Holten-Andersen MN, Riisbro R, et al: Elevated plasma levels of TIMP-1 correlate with plasma suPAR/uPA in patients with chronic myeloproliferative disorders. Eur J Haematol 71:377-384, 2003

    Katoh O, Kimura A, Itoh T, et al: Platelet derived growth factor messenger RNA is increased in bone marrow megakaryocytes in patients with myeloproliferative disorders. Am J Hematol 35:145-150, 1990

    Schmitt A, Jouault H, Guichard J, et al: Pathologic interaction between megakaryocytes and polymorphonuclear leukocytes in myelofibrosis. Blood 96:1342-1347, 2000

    Centurione L, Di Baldassarre A, Zingariello M, et al: Increased and pathological emperipolesis of neutrophils within megakaryocytes associated with marrow fibrosis in GATA-1low mice. Blood 104:3573-3580, 2004

    Pelus LM, Bian H, King AG, et al: Neutrophil-derived MMP-9 mediates synergistic mobilization of hematopoietic stem and progenitor cells by the combination of G-CSF and the chemokines GRO-beta/CXCL2 and GRO-betaT/CXCL2-delta-4. Blood 103:110-119, 2004

    Barosi G, Viarengo G, Pecci A, et al: Diagnostic and clinical relevance of the number of circulating CD34(+) cells in myelofibrosis with myeloid metaplasia. Blood 98:3249-3255, 2001

    Xu M, Bruno E, Chao J, et al: The constitutive mobilization of CD34+ cells into the peripheral blood in idiopathic myelofibrosis may be due to the action of a number of proteases. Blood 105:4508-4515, 2005

    Chagraoui H, Komura E, Tulliez M, et al: Prominent role of TGF-beta 1 in thrombopoietin-induced myelofibrosis in mice. Blood 100:3495-3503, 2002

    Yanagida M, Ide Y, Imai A, et al: The role of transforming growth factor-beta in PEG-rHuMGDF-induced reversible myelofibrosis in rats. Br J Haematol 99:739-745, 1997

    Yan XQ, Lacey D, Fletcher F, et al: Chronic exposure to retroviral vector encoded MGDF (mpl-ligand) induces lineage-specific growth and differentiation of megakaryocytes in mice. Blood 86:4025-4033, 1995

    Yan XQ, Lacey D, Hill D, et al: A model of myelofibrosis and osteosclerosis in mice induced by overexpressing thrombopoietin (mpl ligand): Reversal of disease by bone marrow transplantation. Blood 88:402-409, 1996

    Villeval JL, Cohensolal K, Tulliez M, et al: High thrombopoietin production by hematopoietic cells induces a fatal myeloproliferative syndrome in mice. Blood 90:4369-4383, 1997

    Vannucchi AM, Bianchi L, Cellai C, et al: Development of myelofibrosis in mice genetically impaired for GATA-1 expression (GATA-1(low) mice). Blood 100:1123-1132, 2002

    Kaushansky K: Thrombopoietin: The primary regulator of platelet production. Blood 86:419-431, 1995

    Vyas P, Ault K, Jackson CW, et al: Consequences of GATA-1 deficiency in megakaryocytes and platelets. Blood 93:2867-2875, 1999

    Zhou W, Toombs CF, Zou T, et al: Transgenic mice overexpressing human C-Mpl ligand exhibit chronic thrombocytosis and display enhanced recovery from 5-fluorouracil or antiplatelet serum treatment. Blood 89:1551-1559, 1997

    Kakumitsu H, Kamezaki K, Shimoda K, et al: Transgenic mice overexpressing murine thrombopoietin develop myelofibrosis and osteosclerosis. Leuk Res 29:761-769, 2005

    Frey BM, Rafii S, Teterson M, et al: Adenovector-mediated expression of human thrombopoietin cDNA in immune-compromised mice: Insights into the pathophysiology of osteomyelofibrosis. J Immunol 160:691-699, 1998

    Abina MA, Tulliez M, Lacout C, et al: Major effects of TPO delivered by a single injection of a recombinant adenovirus on prevention of septicemia and anemia associated with myelosuppression in mice: Risk of sustained expression inducing myelofibrosis due to immunosuppression. Gene Ther 5:497-506, 1998

    Ulich TR, Delcastillo J, Senaldi G, et al: Systemic hematologic effects of PEG-rHuMGDF-induced megakaryocyte hyperplasia in mice. Blood 87:5006-5015, 1996

    Shehata M, Schwarzmeier JD, Hilgarth M, et al: TGF-beta1 induces bone marrow reticulin fibrosis in hairy cell leukemia. J Clin Invest 113:676-685, 2004

    Chagraoui H, Tulliez M, Smayra T, et al: Stimulation of osteoprotegerin production is responsible for osteosclerosis in mice overexpressing TPO. Blood 101:2983-2989, 2003

    Vannucchi A, Bianchi L, Laurenzana A, et al: In vivo treatment with thrombopoietin corrects the thrombocytopenia and does not accelerate the progression of myelofibrosis in GATA-1low mice. Exp Hematol 31:186-187, 2003 (abstr)

    Vannucchi A, Bianchi L, Paoletti F, et al: Impaired GATA-1 expression and myelofibrosis in an animal model. Pathol Biol 52:275-279, 2004

    Tefferi A, Elliot MA, Yoon SY, et al: Clinical and bone marrow effects of interferon alfa therapy in myelofibrosis with myeloid metaplasia. Blood 97:1896, 2001

    Tefferi A, Silverstein MN, Li CY: 2-Chlorodeoxyadenosine treatment after splenectomy in patients who have myelofibrosis with myeloid metaplasia. Br J Haematol 99:352-357, 1997

    Faoro LN, Tefferi A, Mesa RA: Long-term analysis of the palliative benefit of 2-chlorodeoxyadenosine for myelofibrosis with myeloid metaplasia. Eur J Haematol 74:117-120, 2005

    Yoon SY, Li CY, Mesa RA, et al: Bone marrow effects of anagrelide therapy in patients with myelofibrosis with myeloid metaplasia. Br J Haematol 106:682-688, 1999

    Tefferi A, Silverstein MN, Plumhoff EA, et al: Suramin toxicity and efficacy in agnogenic myeloid metaplasia. J Natl Cancer Inst 85:1520-1522, 1993

    Mesa RA, Tefferi A, Elliott MA, et al: A phase II trial of pirfenidone (5-methyl-1-phenyl-2-[1H]-pyridone), a novel anti-fibrosing agent, in myelofibrosis with myeloid metaplasia. Br J Haematol 114:111-113, 2001

    Tefferi A, Mesa RA, Gray LA, et al: Phase 2 trial of imatinib mesylate in myelofibrosis with myeloid metaplasia. Blood 99:3854-3856, 2002

    Mesa RA, Elliott MA, Schroeder G, et al: Durable responses to thalidomide-based drug therapy for myelofibrosis with myeloid metaplasia. Mayo Clin Proc 79:883-889, 2004

    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 101:2534-2541, 2003

    Tefferi A, Elliot MA: Serious myeloproliferative reactions associated with the use of thalidomide in myelofibrosis with myeloid metaplasia. Blood 96:4007, 2000

    Elliott MA, Mesa RA, Li CY, et al: Thalidomide treatment in myelofibrosis with myeloid metaplasia. Br J Haematol 117:288-296, 2002

    Steensma DP, Mesa RA, Li CY, et al: Etanercept, a soluble tumor necrosis factor receptor, palliates constitutional symptoms in patients with myelofibrosis with myeloid metaplasia: Results of a pilot study. Blood 99:2252-2254, 2002

    Dredge K, Marriott JB, Macdonald CD, et al: Novel thalidomide analogues display anti-angiogenic activity independently of immunomodulatory effects. Br J Cancer 87:1166-1172, 2002

    Corral LG, Haslett PA, Muller GW, et al: Differential cytokine modulation and T cell activation by two distinct classes of thalidomide analogues that are potent inhibitors of TNF- alpha. J Immunol 163:380-386, 1999

    List A, Kurtin S, Roe DJ, et al: Efficacy of lenalidomide in myelodysplastic syndromes. N Engl J Med 352:549-557, 2005

    Zhang B, Prendergast GC, Fenton RG: Farnesyltransferase inhibitors reverse Ras-mediated inhibition of Fas gene expression. Cancer Res 62:450-458, 2002

    Cortes J, Albitar M, Thomas D, et al: Efficacy of the farnesyl transferase inhibitor R115777 in chronic myeloid leukemia and other hematologic malignancies. Blood 101:1692-1697, 2003

    Adams J: The development of proteasome inhibitors as anticancer drugs. Cancer Cell 5:417-421, 2004

    Giles FJ, Cooper MA, Silverman L, et al: Phase II study of SU5416–a small-molecule, vascular endothelial growth factor tyrosine-kinase receptor inhibitor–in patients with refractory myeloproliferative diseases. Cancer 97:1920-1928, 2003

    Smolich BD, Yuen HA, West KA, et al: The antiangiogenic protein kinase inhibitors SU5416 and SU6668 inhibit the SCF receptor (c-kit) in a human myeloid leukemia cell line and in acute myeloid leukemia blasts. Blood 97:1413-1421, 2001

    Giles FJ, List AF, Roboz GJ, et al: Phase I/II study of PTK787/ZK 222584 (PTK/ZK), a novel, oral VEGF-receptor inhibitor, in patients with myelofibrosis with myeloid metaplasia. Blood 102:922a–923a, 2003

    Butzal M, Loges S, Schweizer M, et al: Rapamycin inhibits proliferation and differentiation of human endothelial progenitor cells in vitro. Exp Cell Res 300:65-71, 2004

    Shegogue D, Trojanowska M: Mammalian target of rapamycin positively regulates collagen type I production via a phosphatidylinositol 3-kinase-independent pathway. J Biol Chem 279:23166-23175, 2004

    Gomez-Cambronero J: Rapamycin inhibits GM-CSF-induced neutrophil migration. FEBS Lett 550:94-100, 2003

    Hu X, Zuckerman KS: Transforming growth factor: Signal transduction pathways, cell cycle mediation, and effects on hematopoiesis. J Hematother Stem Cell Res 10:67-74, 2001

    Chujo S, Shirasaki F, Kawara S, et al: Connective tissue growth factor causes persistent proalpha2(I) collagen gene expression induced by transforming growth factor-beta in a mouse fibrosis model. J Cell Physiol 203:447-456, 2005

    Leask A, Holmes A, Black CM, et al: Connective tissue growth factor gene regulation: Requirements for its induction by transforming growth factor-beta 2 in fibroblasts. J Biol Chem 278:13008-13015, 2003

    Mageto Y, Flaherty K, Brown K, et al: Safety and tolerability of human monoclonal antibody FG-3019, anti-connective tissue growth factor, in patients with idiopathic pulmonary fibrosis. Chest 126:773Sa, 2004 (abstr)

    Uhl M, Aulwurm S, Wischhusen J, et al: SD-208, a novel transforming growth factor beta receptor I kinase inhibitor, inhibits growth and invasiveness and enhances immunogenicity of murine and human glioma cells in vitro and in vivo. Cancer Res 64:7954-7961, 2004

    Hjelmeland MD, Hjelmeland AB, Sathornsumetee S, et al: SB-431542, a small molecule transforming growth factor-beta-receptor antagonist, inhibits human glioma cell line proliferation and motility. Mol Cancer Ther 3:737-745, 2004

    Flanders KC: Smad3 as a mediator of the fibrotic response. Int J Exp Pathol 85:47-64, 2004

    Long J, Matsuura I, He D, et al: Repression of Smad transcriptional activity by PIASy, an inhibitor of activated STAT. Proc Natl Acad Sci U S A 100:9791-9796, 2003

    Ugo V, Marzac C, Teyssandier I, et al: Multiple signaling pathways are involved in erythropoietin-independent differentiation of erythroid progenitors in polycythemia vera. Exp Hematol 32:179-187, 2004

    Zupanska B, Szmigielski S, Litwin J, et al: Cytoenzymatic changes in the course of experimental osteomyelosclerosis in rats. Arch Immunol Ther Exp (Warsz) 14:356-362, 1966

    Stodtmeister R, Becker H, Cronkite EP, et al: Experimental bone marrow fibrosis in rats following sublethal whole-body x-ray irradiation and bone marrow transfusions as a model of human myelofibrosis [in German]. Schweiz Med Wochenschr 98:1671-1673, 1968

    Van Gorp LH, Swaen GJ: Myelofibrosis in rats experimentally infected with a murine leukaemia virus. J Pathol 97:235-240, 1969

    Takacsi-Nagy L, Juhasz J: Experimental myelosclerosis. Haematologia (Budap) 5:283-291, 1971

    Stodtmeister R, Fliedner TM: Morphological aspects of myelofibrosis, observed in rats following sublethal whole body irradiation and subsequent allogeneic bone marrow cell transfusion. Folia Haematol Int Mag Klin Morphol Blutforsch 100:23-50, 1973

    Hunstein W: Experimental myelofibrosis. Clin Haematol 4:457-478, 1975

    Ebbe S: Experimental and clinical megakaryocytopoiesis. Clin Haematol 8:371-394, 1979

    Ruddle NH, Li CB, Horne WC, et al: Mice transgenic for HTLV-I LTR-tax exhibit tax expression in bone, skeletal alterations, and high bone turnover. Virology 197:196-204, 1993

    Harrison CN, Gale RE, Wiestner AC, et al: The activating splice mutation in intron 3 of the thrombopoietin gene is not found in patients with non-familial essential thrombocythaemia. Br J Haematol 102:1341-1343, 1998

    Kiladjian JJ, Elkassar N, Hetet G, et al: Study of the thrombopoietin receptor in essential thrombocythemia. Leukemia 11:1821-1826, 1997

    Martyre MC, Steunou V, LeBousse-Kerdiles MC, et al: Lack of alteration in GATA-1 expression in CD34+ hematopoietic progenitors from patients with idiopathic myelofibrosis. Blood 101:5087-5088; author reply 5088-5089, 2003

    Gandini D, Allen AJ, Nash MJ, et al: No mutations in the gata-1 gene detected in patients with acquired essential thrombocythemia. Haematologica 89:613-615, 2004

    Fisher C, Steensma D, Janmohamed R, et al: Absence of mutations in the key megakaryocyte transcriptional regulator FOG-1 in patients with idiopathic myelofibrosis. Br J Haematol 126:750-752, 2004

    Najfeld V, Montella L, Scalise A, et al: Exploring polycythaemia vera with fluorescence in situ hybridization: Additional cryptic 9p is the most frequent abnormality detected. Br J Haematol 119:558-566, 2002

    Steensma DP, Tefferi A: Cytogenetic and molecular genetic aspects of essential thrombocythemia. Acta Haematol 108:55-65, 2002

    Pfeilstocker M, Reisner R, Nosslinger T, et al: Cross-validation of prognostic scores in myelodysplastic syndromes on 386 patients from a single institution confirms importance of cytogenetics. Br J Haematol 106:455-463, 1999(Ayalew Tefferi)