Chronic myelogenous leukemias (CMLs)


Summary

The chronic myelogenous leukemias (CMLs) include BCR rearrangement-positive CML, chronic myelomonocytic leukemia, juvenile myelomonocytic leukemia, chronic neutrophilic leukemia, chronic eosinophilic leukemia, chronic basophilic leukemia, and possibly chronic monocytic leukemia. The term chronic, in contrast to acute, once had prognostic implications. However, although the terms remain useful for nosology, they no longer reflect an invariable difference in prognosis. For example, acute myelogenous leukemia in children and young adults has higher remission and cure rates than juvenile or chronic myelomonocytic leukemia in children or adults, respectively. BCR rearrangement-positive CML presents with anemia, exaggerated granulocytosis, a large proportion of myelocytes and mature neutrophils, absolute basophilia, normal or elevated platelet counts, and, frequently, splenomegaly. The marrow is hypercellular, and marrow cells contain the Philadelphia (Ph) chromosome in approximately 90 percent of cases by cytogenetic analysis. A rearrangement of the BCR gene on chromosome 22 is present in approximately 96 percent of cases by molecular diagnostic analysis. The disease usually responds to imatinib mesylate, a specific tyrosine kinase inhibitor, and median survival has been extended significantly. Allogeneic stem cell transplantation can cure the disease, especially if the transplantation is applied early in the chronic phase. The effect of stem cell transplantation is related in part to a robust graft-versus-leukemia effect, engendered by donor T lymphocytes. The chronic phase usually is followed by an accelerated phase that often terminates in acute leukemia (blast crisis), at which point therapy with imatinib mesylate and other agents may induce a remission in a proportion of patients, but median survival is measured in months. Blast crisis results in a myelogenous leukemic phenotype in 75 percent of cases and a lymphoblastic leukemic phenotype in approximately 25 percent of cases. Ph-chromosome–positive acute myeloblastic leukemia (AML) may appear de novo in approximately 1 percent of cases of AML, and Ph-chromosome–positive acute lymphocytic leukemia (ALL) may occur de novo in approximately 20 percent of cases of adult ALL and approximately 5 percent of childhood ALL cases. In Ph-chromosome–positive ALL, the translocation between chromosomes 9 and 22 results in the fusion gene encoding a mutant tyrosine kinase oncoprotein that may be identical in size to that in classic CML (210 kDa) in approximately one-third of cases. A smaller mutant tyrosine kinase (190 kDa) is encoded in approximately two-thirds of cases. In children, the cells in approximately 90 percent of cases contain a 190-kDa mutant tyrosine kinase. These acute leukemias may reflect (1) the presentation of CML in acute blastic transformation without a preceding chronic phase or (2) de novo cases resulting from a BCR-ABL mutation occurring in a different hematopoietic cell from the event in CML or with as yet unidentified modifying gene alterations. Chronic myelomonocytic leukemia has variable presenting features. Anemia may be accompanied by mildly or moderately elevated leukocyte counts; an elevated total monocyte count; a low, normal, or elevated platelet count; and sometimes splenomegaly. Although cytogenetic abnormalities may be present, there is no specific genetic marker of the disease. In a very small proportion of cases, a translocation involving the platelet-derived growth factor receptor-beta (PDGFR-) gene is associated with eosinophilia and is responsive to imatinib mesylate. Juvenile myelomonocytic leukemia occurs in infancy or very early childhood. Anemia, thrombocytopenia, and leukocytosis with monocytosis are usual. The disease is refractory to treatment and, even with current maximal therapy and stem cell rescue, cures are uncommon. Chronic neutrophilic leukemia presents with mild anemia and exaggerated neutrophilia, with very few immature cells in the blood. Splenomegaly is common. The disease usually occurs after age 60 years and is refractory to current treatment approaches. Chronic and juvenile myelomonocytic leukemia and chronic neutrophilic leukemia have a propensity to evolve into acute myelogenous leukemia. Prior to that evolution, morbidity and mortality are related to infection, hemorrhage, and complicating medical conditions. Chronic eosinophilic leukemia represents the major subset of the hypereosinophilic syndrome. It is a clonal disorder with a striking absolute eosinophilia, often neurologic and cardiac manifestations secondary to toxic effects of eosinophil granules, and sometimes a translocation involving the platelet-derived growth factor receptor-alpha (PDGFR-) gene that encodes a mutant tyrosine kinase, imparting sensitivity to imatinib mesylate.

Acronyms and Abbreviations

Acronyms and abbreviations that appear in this chapter include: AGP, 1-acid glycoprotein; ALL, acute lymphocytic leukemia; BCR, breakpoint cluster region; CCyR, complete cytogenetic remission; CFU-GM, colony-forming unit–granulocyte-monocyte; CHR, complete hematologic response; CLL, chronic lymphocytic leukemia; CML, chronic myelogenous leukemia; CMML, chronic myelomonocytic leukemia; DLI, donor lymphocyte infusion; FISH, fluorescence in situ hybridization; G-CSF, granulocyte colony-stimulating factor; GM-CSF, granulocyte-monocyte colony-stimulating factor; GRB2, growth factor receptor-bound protein-2; GTP, guanosine triphosphate; GTPase, guanosine triphosphatase; GVHD, graft-versus-host disease; HLA, human leukocyte antigen; HPRT, hypoxanthine phosphoribosyltransferase; HR, hematologic response; hsp, heat shock protein; HUMARA, human androgen receptor assay; IFN, interferon; IL, interleukin; JAK, Janus-associated kinase; LTC-IC, long-term culture-initiating cell; MCP, monocyte chemotactic protein; MCyR, major cytogenetic response; mCyR, minor cytogenetic response; MDS, myelodysplastic syndrome; MIP, macrophage inflammatory protein; MMR, major molecular response; mRNA, messenger RNA; NF-B, nuclear factor-B; NF1, neurofibromatosis tumor suppressor gene; NK, natural killer; NOD, nonobese diabetic; PCR, polymerase chain reaction; PDGF, platelet-derived growth factor; PDGFR, platelet-derived growth factor receptor; PHR, partial hematologic response; Ph, Philadelphia chromosome; PI3K, phosphatidylinositol 3′-kinase; Rb, retinoblastoma; RT-PCR, reverse transcriptase polymerase chain reaction; SCID, severe combined immunodeficiency; STAT, signal transducer and activator of transcription; TBI, total-body irradiation; TdT, terminal deoxynucleotidyl transferase; TGF, transforming growth factor; VEGF, vascular endothelial growth factor; WT, Wilms tumor.

Definition and History

Chronic myelogenous leukemia (CML) is a pluripotential stem cell disease characterized by anemia, extreme blood granulocytosis and granulocytic immaturity, basophilia, often thrombocytosis, and splenomegaly. The hematopoietic cells contain a reciprocal translocation between chromosomes 9 and 22 in more than 95 percent of patients, which leads to an overtly foreshortened long arm of one of the chromosome pair 22 (i.e., 22, 22q–) referred to as the Philadelphia (Ph) chromosome. A rearrangement of the breakpoint cluster gene on the long arm of chromosome 22 defines this form of CML and is present even in the 10 percent of patients without an overt 22q abnormality by Giemsa banding. The natural history of the disease is to undergo clonal evolution into an accelerated phase and/or a rapidly progressive phase resembling acute leukemia, which is refractory to therapy.

In 1845, Bennett1 in Scotland and Virchow2 in Germany described patients with splenic enlargement, severe anemia, and enormous concentrations of leukocytes in their blood at autopsy. Bennett initially favored an extreme pyemia as the explanation, but Virchow argued against suppuration as a cause. Additional cases were reported by Craige3 and others, and in 1847 Virchow4 introduced the designation weisses Blut and leukämie (leukemia). In 1878, Neumann5 proposed that the marrow not only was the site of normal blood cell production, but also was the site from which leukemia originated and used the term myelogene (myelogenous) leukemia. Subsequent observations amplified the clinical and laboratory features of the disease, but few fundamental insights were gained until the discovery by Nowell and Hungerford,6 who reported in 1960 that two patients with the disease had an apparent loss of the long arm of chromosome 21 or 22, an abnormality that was quickly confirmed7–9 and designated the Philadelphia chromosome.7 This observation led to a new approach to diagnosis, a marker to study the pathogenesis of the disease, and a focus for future studies of the molecular pathology of the disease. The availability of banding techniques to define the fine structure of chromosomes10,11 led to the discovery by Rowley12 that the apparent lost chromosomal material on chromosome 22 was part of a reciprocal translocation between chromosomes 9 and 22. The discovery that the cellular oncogene ABL on chromosome number 9 and a segment of chromosome 22, the breakpoint cluster region (BCR), fuse as a result of the translocation provided a basis for the study of the molecular cause of the disease.13,14 The appreciation that the fusion gene encoded a constitutively active tyrosine kinase (BCR-ABL) that was capable of inducing the disease in mice established the fusion gene product as the proximate cause of the malignant transformation. The search for, identification of, and clinical development of a small molecule inhibitor of the mutant tyrosine kinase has provided a specific agent, imatinib mesylate, with which to inhibit the molecule that incites the disease.15 Several more potent congeners have also been synthesized (see “Etiology and Pathogenesis” below). Thomas and colleagues established that allogeneic hematopoietic stem cell transplantation could cure the disease.16

EpidemiologyCML accounts for approximately 15 percent of all cases of leukemia, or approximately 5000 new cases per year in the United States. The age-adjusted incidence rate in the United States is approximately 2.0 per 100,000 persons for men and approximately 1.1 per 100,000 persons for women. The incidence around the world varies by a factor of approximately twofold. The lowest incidence is in Sweden and China (approximately 0.7 per 100,000 persons), and the highest incidence is in Switzerland and the United States (approximately 1.5 per 100,000 persons).17 The age-specific incidence rate for CML in the United States increases logarithmically with age, from approximately 0.2 per 100,000 persons younger than 20 years to a rate of approximately 10.0 per 100,000 octogenarians per year (Fig. 90–1). Although CML occurs in children and adolescents, less than 10 percent of all cases occur in subjects between 1 and 20 years old. CML represents approximately 3 percent of all childhood leukemias. Multiple occurrences of CML in families are rare. There is no concordance of the disease between identical twins. Analytical epidemiologic evidence for a familial predisposition in CML was not found in a Swedish database.18

Figure 90–1.
Incidence of chronic myelogenous leukemia by age. Note the exponential increase in incidence with age from about teenagers to octogenarians. Rare cases occur in younger children but too few to generate an incidence rate.

Etiology and Pathogenesis

Environmental Leukemogens

Exposure to very high doses of ionizing radiation can increase the occurrence of CML above the expected frequency in comparable populations. Three major populations—the Japanese exposed to the radiation released by the atomic bomb detonations at Nagasaki and Hiroshima,19 British patients with ankylosing spondylitis treated with spine irradiation,20,21 and women with uterine cervical carcinoma who received radiation therapy22—had a frequency of CML (as well as acute leukemia) significantly above the frequency expected in comparable unexposed groups. The median latent period was approximately 4 years in irradiated spondylitics, among whom approximately 20 percent of the leukemia cases were CML; 9 years in the uterine cervical cancer patients, of whom approximately 30 percent had CML; and 11 years in the Japanese survivors of the atomic bombs, of whom approximately 30 percent of the leukemia patients had CML.23 Chemical leukemogens, such as benzene and alkylating agents, are not causative agents of CML, although they are well established to produce a dose-dependent increase in acute myelogenous leukemia.24

Origin from a Hematopoietic Stem Cell Clone

CML results from the malignant transformation of a single multipotential hematopoietic cell. The disease is acquired (somatic mutation), given that the identical twin of patients with CML and the offspring of mothers with the disease neither carry the Ph chromosome nor develop the disease.25 The origin of CML from a single multipotential hematopoietic cell is supported by the following lines of evidence:

  1. Involvement of erythropoiesis, neutrophilopoiesis, eosinophilopoiesis, basophilopoiesis, monocytopoiesis, and thrombopoiesis in chronic phase CML26
  2. Presence of the Ph chromosome (22q–) in erythroblasts; neutrophilic, eosinophilic, and basophilic granulocytes; macrophages; and megakaryocytes27
  3. Presence of a single glucose-6-phosphate dehydrogenase isoenzyme in red cells, neutrophils, eosinophils, basophils, monocytes, and platelets, but not in fibroblasts or other somatic cells in women with CML who are heterozygotes for isoenzymes A and B28–30
  4. Presence of the Ph translocation only on a structurally anomalous chromosome 9 or 22 of each chromosome pair in every cell analyzed in occasional patients with a structurally dissimilar 9 or 22 chromosome within the pair31–33
  5. Presence of the Ph chromosome in one but not the other cell lineage of patients who are a mosaic for sex chromosomes, as in Turner syndrome (45X/46XX)34 and Klinefelter syndrome (46XY/47XXY)35
  6. Molecular studies showing variation in the breakpoint of chromosome 22 among different patients with CML but precisely the same breakpoint among cells within a single patient with CML36,37
  7. Combined DNA hybridization-methylation analysis of women who have restriction fragment length polymorphisms at the X-linked locus for hypoxanthine phosphoribosyltransferase (HPRT), which enables distinction of the two alleles of the HPRT gene in heterozygous females, coupled with methylation-sensitive restriction-enzyme cleavage patterns, which permits delineation of whether cells contain either the maternally derived or the paternally derived copy of the gene38

The foregoing observations place the parent cell of the clone at least at the level of the hematopoietic stem cell.

The Chronic Myelogenous Leukemia Stem Cell

Acquisition of the BCR-ABL fusion gene as a result of the t(9;22) (q34;q11.2) in a single multipotential hematopoietic cell results in the CML stem cell, necessary for the initiation and maintenance of the chronic phase of CML.39,40 The phenotype of the CML stem cell is not fully defined but they are among the CD34+CD33– fraction of CML cells. A large proportion of CML stem cells are in the Go phase of the cell cycle and are resistant to therapy with BCR-ABL inhibitors. These cells represent a pool for the regrowth of the tumor, if suppressive therapy is interrupted. The acquisition of genetic and epigenetic events in a derivative BCR-ABL–positive cell can result in evolution to accelerated phase and blastic transformation41 (see “Accelerated Phase and Blast Crisis of CML” below).

Pluripotential versus Hematopoietic Stem Cell Lesion

Some patients in chronic phase CML have lymphocytes that are derived from the primordial malignant cell. Evidence for this finding includes the following: A single isoenzyme for glucose-6-phosphate dehydrogenase has been found in some T and B lymphocytes in women with CML who are heterozygous for isoenzymes A and B;42 blood cells from patients with CML induced to proliferate with Epstein-Barr virus (presumptive B lymphocytes) are of the same glucose-6-phosphate dehydrogenase isoenzyme type, have cytoplasmic immunoglobulin heavy and light chains, and contain the Ph chromosome;43 blood lymphocytes stimulated with B lymphocyte mitogens contain the Ph chromosome;44,45 purified B lymphocytes from the blood in chronic phase CML contain an abnormal, elongated phosphoprotein coded for by the chimeric gene resulting from the t(9;22);46 and fluorescence in situ hybridization (FISH) has detected the BCR-ABL fusion gene in approximately 25 percent of B lymphocytes in some, but not all, patients in chronic phase.47,48 These findings suggest that B lymphocytes are derived from the malignant clone, placing the lesion closer to, if not in, the pluripotential stem cell.42–46 Almost all studies find that the B lymphocyte pool is a mosaic, containing both Ph-chromosome– and BCR-ABL–positive cells and Ph-chromosome– or BCR-ABL–negative cells. Results of studies examining the derivation of T lymphocytes from the malignant clone are more ambiguous but indicate that T lymphocytes are derived from the malignant clone in some, but not most, patients.42,44,49–58 Natural killer (NK) cells isolated from patients with chronic phase CML do not contain the BCR-ABL fusion gene.59 It is possible that myelopoiesis is invariably clonal and lymphopoiesis is an unpredictable mosaic derived largely from normal residual stem cells. This conclusion is supported by the finding that progenitors of T, B, and NK lymphocytes contain the Ph chromosome and BCR-ABL, but most B-cell and all T-cell progenitors derived from the leukemic clone undergo apoptosis, leaving unaffected cells in the blood.60–63

The cell in which the mutation occurs may be even more primitive in that some endothelial cells generated in vitro express the BCR-ABL fusion gene, as do some cells in the patient’s vascular endothelium.64

Etiologic Role of the pH Chromosome

Early studies indicated that the Ph chromosome may appear after the initial leukemogenic event.65–68 Patients with CML have developed the Ph chromosome during the course of the disease, have experienced periods of the disease when the Ph chromosome disappeared,69 or have had Ph-chromosome–positive and Ph-chromosome–negative cells concurrently.70–74

Nearly all, if not all, patients with CML have an abnormality of chromosome 22 at a molecular level (BCR rearrangement). Thus, earlier studies indicating an absence of a Ph chromosome were not a valid measure of the normality of chromosome 22. The molecular abnormality in CML involving the ABL gene on chromosome 9 and the BCR gene on chromosome 22 has been established as being the proximate cause of the chronic phase of the disease (see “Molecular Pathology” below).

Coexistence of Normal Stem Cells

Most, if not all, patients with CML have hematopoietic stem cells that, after treatment75–77 or culture in vitro,78–80 use of special cell isolation techniques,81,82 or use of cell transfer to nonobese diabetic (NOD)/severe combined immunodeficiency (SCID) mice83 do not have the Ph chromosome84,85 or the BCR-ABL fusion gene.86–90 The switch to Ph-chromosome–negative cells in vitro is associated with a loss of monoclonal glucose-6-phosphate dehydrogenase isoenzyme patterns, indicating the persistence and reemergence of normal polyclonal hematopoiesis rather than reversion to a Ph-chromosome–negative clone.91 In confirmation, BCR-ABL+, CD34+, human leukocyte antigen (HLA)-DR– cells isolated from women with early phase CML are polyclonal using the human androgen receptor assay (HUMARA) to assess X chromosome inactivation patterns.92 Very primitive hematopoietic cells, the long-term culture-initiating cells (LTC-ICs), are present in Ph-chromosome–negative cytapheresis samples collected during early recovery after chemotherapy for CML.93 These LTC-ICs are most commonly present when samples are collected within 3 months of diagnosis.94 Variable levels of BCR-ABL–negative progenitors are found in the CD34+DR– population, but low levels are found in the CD34+CD38– population.90,95 Preprogenitors for the CD34+DR– cells are predominantly BCR-ABL–negative in both marrow and blood at diagnosis.96 However, some cells with surface marker characteristics of very primitive normal hematopoietic cells do express the BCR-ABL gene.97 Both normal and leukemic SCID-repopulating cells coexist in the marrow and blood from CML patients in chronic phase, whereas only leukemic SCID-repopulating cells are detected in blast crisis.98,99

Progenitor Cell Characteristics

Progenitor Cell Dysfunction

The leukemic transformation resulting from the BCR-ABL fusion oncogene is maintained by a relatively small number of BCR-ABL stem cells that favor differentiation over self-renewal.100 This predisposition to differentiation and progenitor cell expansion is mediated by an autocrine interleukin (IL)-3–granulocyte colony-stimulating factor (G-CSF) loop.100 The earliest progenitors have the capacity to undergo marked expansion of erythroid, granulocytic, and megakaryocytic cell populations, and have a decreased sensitivity to regulation.100–102 This expansion is especially dramatic in the more mature progenitor cell compartment.100,103 The proliferative capacity of individual granulocytic progenitors is decreased compared to normal cells. Thus, the progenitor cell population in marrow and blood expands proportionately more than the increase in granulopoiesis.104,105 Moreover, the progenitors have buoyant density that is lighter than that of their normal counterparts but similar to that of hepatic fetal granulopoietic progenitors, suggesting an oncofetal pattern.104 The marked expansion of the total blood granulocyte pool results from a total expansion of granulopoiesis,103,106 with a minor contribution from prolonged intravascular circulation time.107 BCR-ABL reduces growth factor dependence of progenitor cells.

Erythroid progenitors are expanded, erythroid precursor maturation is blocked at the basophilic erythroblast stage, and the extent of erythropoiesis is inversely proportional to the total white cell count.108

Progenitor Cell Characterization

Phenotypic differences of stem and progenitor cells in CML patients compared to normal subjects have been identified.109 For example, a greater proportion of the circulating leukemic colony-forming unit–granulocyte-monocytes (CFU-GMs) express high levels of the adhesion receptor CD44110 and low levels of L-selectin111 in contrast to normal cells. Leukemic CD34+ cells overexpress the P glycoprotein that determines the multidrug resistance phenotype.112

BCR-ABL–positive progenitors survive less well in long-term culture than do their normal counterparts. Leukemic CFU-GM colonies, unlike normal colonies, decrease in long-term cultures that are deficient in KIT ligand,113 whereas their proliferation is favored in the presence of KIT ligand.114 Macrophage inflammatory protein (MIP)-1 does not inhibit growth factor-mediated proliferation of CD34+ cells from CML patients, as it does CD34+ cells from normal subjects, even though the MIP-1 receptor is expressed.115 Another chemokine, monocyte chemotactic protein (MCP)-1, unlike MIP-1, is an endogenous chemokine that cooperates with transforming growth factor beta (TGF-) to inhibit the cycling of primitive normal, but not CML, progenitors in long-term human marrow cultures.116 Leukemic progenitors are less sensitive than normal progenitors to the antiproliferative effects of TGF-.117

Effects of BCR-ABL on Cell Adhesion

Primitive progenitors and blast colony-forming cells from patients with CML have decreased adherence to marrow stromal cells.118,119 This defect is normalized if stromal cells are treated with interferon (IFN)-.119,120 As a result, BCR-ABL–negative progenitors are enriched in the adherent fraction of circulating CD34+ cells in chronic phase CML patients. The most primitive BCR-ABL–positive cells in the blood of patients with CML differ from their normal counterparts. They are increased in frequency and are activated, such that signals that block cell mitosis are bypassed.121

Ph-chromosome–positive colony-forming cells adhere less to fibronectin (and to marrow stroma) than do their normal counterparts. Adhesion is fostered as a result of restoration of cooperation between activated 1 integrins and the altered epitopes of CD44.122–124 CML granulocytes have reduced and altered binding to P-selectin because of modification in the CD15 antigens.125 BCR-ABL–induced defects in integrin function may underlie the abnormal circulation and proliferation of progenitors126,127 because growth signaling can occur through the fibronectin receptor.128 IFN- restores normal integrin-mediated inhibition of hematopoietic progenitor proliferation by the marrow microenvironment.129 There are conflicting data regarding the effects of tyrosine kinase inhibitor effects on adhesion of CML cells to stroma.130,131

BCR-ABL–encoded fusion protein p210BCR–ABL binds to actin, and several cytoskeletal proteins are thereby phosphorylated. The p210BCR–ABL interacts with actin filaments through an actin-binding domain. BCR-ABL transfection is associated with increased spontaneous motility, membrane ruffling, formation of long actin extensions (filopodia), and accelerated rate of protrusion and retraction of pseudopodia on fibronectin-coated surfaces. IFN- treatment slowly converts the abnormal motility phenotype of BCR-ABL–transformed cells toward normal.132 Integrins regulate the c-ABL–encoded tyrosine kinase activity and its cytoplasmic nuclear transport.133 The p210BCR–ABL abrogates the anchorage requirement but not the growth factor requirement for proliferation.134

In normal cells exposed to IL-3, paxillin tyrosine residues are phosphorylated. In cells transformed by p210BCR–ABL, the tyrosines of paxillin, vinculin, p125FAK, talin, and tensin are constitutively phosphorylated. Pseudopodia enriched in focal adhesion proteins134,135 are present in cells expressing p210BCR–ABL.

The sum of evidence suggests that defects in adhesion (contact and anchoring) of CML primitive cells remove them from their controlling signals normally received from microenvironmental cells via cytokine messages. These signals retain the balance among cell survival, cell death, cell proliferation, and cell differentiation. Inappropriate phosphorylation of cytoskeletal proteins, possibly independent of tyrosine kinase, is thought to be the key factor in disturbed integrin function of CML cells.

Molecular Pathology

pH Chromosome

The genic disturbance became evident with the knowledge that CML was derived from a primitive cell containing a 22q– abnormality.6,11 The abnormal chromosome contained only 60 percent of the DNA in other G-group chromosomes.136 Cytogenetic analysis indicated the G-group chromosome involved was different from the extra G-group chromosome in Down syndrome, which had been assigned number 21. Thus, the former was assigned number 22—even though it proved to be slightly longer than the chromosome involved in Down syndrome.11,137 The Paris Conference on Nomenclature decided not to undo the concept that Down syndrome is trisomy 21 and assigned the Ph chromosome and its normal counterpart, 22.138 Using quinacrine (Q) and Giemsa (G) banding, Rowley12 reported in 1973 that the material missing from chromosome 22 was not lost (deleted) from the cell, but was translocated to the distal portion of the long arm of chromosome 9. The amount of material translocated to chromosome 9 was approximately equivalent to that lost from 22, and the translocation was predicted to be balanced.12 Moreover, the breaks were localized to band 34 on the long arm of 9 and band 11 on the long arm of 22. Therefore, the classic Ph chromosome is t(9;22)(q34;q11), abbreviated t(Ph) (Fig. 90–2). The Ph chromosome can develop on either the maternal or the paternal member of the pair.139

Figure 90–2.
Schematic of normal chromosome 9 showing the ABL gene between band q34 and qter of chromosome 22, which has the BCR and SIS genes between band q11 and qter. The t(9;22) is shown on the right. The ABL from chromosome 9 is transposed to the chromosome 22 M-bcr sequences, and the terminal portion of chromosome 22 is transposed to the long arm of chromosome 9. The 22q– is the Ph chromosome. bcr, breakpoint cluster region; c-SiS, cellular homologue of the viral simian sarcoma virus-transforming gene; IGL, gene for immunoglobulin light chains.(From De Klein A: Oncogene activation by chromosomal rearrangement in chronic myelocytic leukemia. Mutat Res 186:161, 1987, with permission.)

Mutation of ABL and BCR Genes

Mutations of the ABL gene on chromosome 9 and of the BCR gene on chromosome 22 are central to the development of CML (Fig. 90–3).140–142

Figure 90–3.
Schematic of the normal ABL and BCR genes and of the BCR-ABL fusion transcripts. In the upper panel of the diagram, the possible breakpoint positions in ABL are marked by vertical arrows. Note the position immediately upstream of the ABL locus of the 8604Met gene. The BCR gene contains 25 exons, including first (e1) and second (e2) exons. The positions of the three breakpoint cluster regions, m-bcr, M-bcr, and –bcr, are shown. The lower panel of the figure shows the structure of the BCR-ABL messenger RNA fusion transcripts. Breakpoints in –bcr result in BCR-ABL transcripts with an e19a2 junction. The associated number designates the exon (location) at which the break occurs in each gene.(From Verschraegen CF, Kantarjian HM, Hirsch-Ginsberg C, et al,179 with permission.)

In 1982, the human cellular homologue ABL of the transforming sequence of the Abelson murine leukemia virus was localized to human chromosome 9.143 In 1983, ABL was shown to be on the segment of chromosome 9 that is translocated to chromosome 22144 by demonstrating reaction to hybridization probes for ABL only in somatic cell hybrids of human CML cells containing 22q– but not those containing 9q+. v-abl is the viral oncogenic homologue of the normal cellular ABL gene. This gene (v-abl) can induce malignant transformation of cells in culture and can induce leukemia in susceptible mice.145

The ABL gene is rearranged and amplified in cell lines from patients with CML.146 Cell lines and fresh isolates of CML cells contain an abnormal, elongated 8-kb RNA transcript,147–150 which is transcribed from the new chimeric gene produced by the fusion of the 5′ portion of the BCR gene left on chromosome 22 with the 3′ portion of the ABL gene translocated from chromosome 9144 (Fig. 90–4). The fusion messenger RNA (mRNA) leads to the translation of a unique tyrosine phosphoprotein kinase of 210 kDa (p210BCR–ABL), which can phosphorylate tyrosine residues on cellular proteins similar to the action of the v-abl protein product.151–155 The anomalous tyrosine kinase is difficult to identify in chronic phase cells because of inhibitors in granulocytes;155 molecular variants reflect variations in the breakpoint on chromosome 22.156

Figure 90–4.
Molecular effects of the Ph chromosome translocation t(9;22)(q34;q11). The upper panel shows the physically joined 5′ BCR and the 3′ ABL regions on chromosome 22. The exons are solid (from chromosome 22, BCR) and hatched (from chromosome 9, ABL). The middle panel depicts transcription of chimeric messenger RNA. The lower panel shows the translated fusion protein with the amino-terminus derived from the BCR of chromosome 22 and the carboxy-terminus from the ABL of chromosome 9.(From De Klein A: Oncogene activation by chromosomal rearrangement in chronic myelocytic leukemia. Mutat Res 186:161, 1987, with permission.)

The ABL locus contains at least two alleles, one having a 500-bp deletion.157 In normal cells, the ABL protooncogene codes for a tyrosine kinase of molecular weight 145,000, which is translated only in trace quantities and lacks any in vitro kinase activity.152 The fusion product expressed by the BCR-ABL gene is hypothesized to lead to malignant transformation because of the abnormally regulated enzymatic activity of the chimeric tyrosine protein kinase.153,154,158,159 Construction of BCR-ABL fusion genes indicated that BCR sequences could also activate a microfilament-binding function, but the tyrosine kinase and microfilament-binding functions were not linked. Nevertheless, tyrosine kinase modification of actin filament function has been proposed as a step in leukemogenesis.160

p210Bcr–Abl Fusion Protein

The breakpoints on chromosome 9 are not narrowly clustered, ranging from approximately 15 to more than 40 kb upstream from the most proximate region (first exon) of the ABL gene.143,144,161 The breakpoints on chromosome 22 occur over a very short, approximately 5 to 6 kb, stretch of DNA referred to as the breakpoint cluster region (M-bcr),162,163 which is part of a much longer breakpoint cluster region gene, BCR164,165 (see Fig. 90–4). Three main breakpoint cluster regions have been characterized on chromosome 22: major (M-bcr), minor (m-bcr), and micro (-bcr). The three different breakpoints result in a p210, p190, and p230 fusion protein, respectively (see Fig. 90–3). The overwhelming majority of CML patients have a BCR-ABL fusion gene that encodes a fusion protein of 210 kDa (p210BCR–ABL), for which mRNA transcripts have e14a2 or a e13a2 fusion junction (see Fig. 90–3).166 The “e” represents the BCR exon and “a” the ABL exon sites involved in the translocation. A BCR-ABL with an e1a2 type of junction has been identified in approximately 50 percent of the Ph chromosome–positive acute lymphoblastic leukemia cases and results in the production of a BCR-ABL protein of 190 kDa (p190BCR–ABL). Almost all CML cases at diagnosis that encode a p210BCR–ABL also express BCR-ABL transcripts for p190.167 The biologic or clinical significance of these dual transcripts is not known. Transgenic mice expressing p210BCR–ABL develop acute lymphoblastic leukemia in the founder mice, but all transgenic progeny have a myeloproliferative disorder resembling CML.168

The BCR gene encodes a 160-kDa serine-threonine kinase, which, when it oligomerizes, autophosphorylates and transphosphorylates several protein substrates.169 Aberrant methylation of the M-bcr in CML occurs.166 The first exon sequences of the BCR gene potentiate the tyrosine kinase of ABL when they fuse as a result of the translocation.170 The central portion of BCR has homology to DBL, a gene involved in the control of cell division after the S phase of the cell cycle. The C-terminus of BCR has a guanosine triphosphatase (GTPase)-activating protein for p21rac, a member of the RAS family of guanosine triphosphate (GTP)-binding proteins.171 A reciprocal hybrid gene ABL-BCR is formed on chromosome 9q+ when BCR-ABL fuses on chromosome 22. The ABL-BCR fusion gene actively transcribes in most patients with CML.172

Variations in breakpoints involving smaller stretches of chromosome 9 and rearrangements outside the M-bcr of chromosome 22 can occur.37 In a few cases of CML with no evident elongation of chromosome 9, molecular probes have shown that ABL still is translocated to chromosome 22.173 In occasional patients with Ph-chromosome–positive CML, the break in chromosome 22 is outside the M-bcr, and transcription of a fusion RNA of the usual type fails or a fusion RNA is transcribed that does not hybridize with the classic M-bcr complementary DNA (cDNA) probe.174

In cases in which the Ph chromosome is not found, BCR-ABL still may be located on chromosome 9 (a masked Ph chromosome).175 The BCR gene can recombine with genomically distinct sites on band 11q13 in complex translocations in a region rich in Alu repeat elements.176 ETV6/ABL fusion genes have also been found in BCR-ABL–negative CML.177

The BCR breakpoint site has been examined as a factor in disease prognosis. Some studies have shown no correlation between CML chronicity and breakpoint site, although thrombocytosis may be more common with 3′ breakpoint sites and basophilia with 5′ breakpoint sites.178 No difference in response to IFN- therapy was noted, and survival was not significantly different, although patients with 3′ deletions tended to have shorter survival.179 Others have observed a better response to IFN- in patients with a 3′ rearrangement, which is being examined with imatinib mesylate therapy.180

CML patients with m-bcr breakpoints develop a blast crisis with monocytosis and an absence of splenomegaly and basophilia.181 The p230 (e19a2 RNA junction) encoded by –bcr is rarely expressed but has been associated with neutrophilic CML or thrombocytosis (see “Special Clinical Features” below). Other rare breakpoints have been described.182 For example, a case with a 12-bp insert between BCR1 and ABL1 resulted in a BCR-ABL–negative (false-negative), Ph-chromosome–positive CML with thrombocythemia.183 Another novel BCR-ABL fusion gene (e6a2) in a patient with Ph-chromosome–negative CML encoded an oncoprotein of 185 kDa.184 Typical CML also has been associated with an e19a2 junction BCR-ABL transcript.185

Experimental support for the hypothesis that p210BCR–ABL tyrosine phosphoprotein kinase is transforming is provided by a retroviral gene transfer system that permits expression of the protein. Mouse marrow cells transfected with BCR-ABL develop clonal outgrowths of immature cells expressing the p210BCR–ABL tyrosine kinase. Some clones progress to a malignant phenotype, can be transplanted, and can induce tumors in syngeneic mice.186 Similar studies suggest that the p210BCR–ABL can transform 3T3 murine fibroblasts if the gag gene sequence from a helper virus cooperates.187 The BCR-ABL gene from a retroviral vector has been expressed in an IL-3–dependent cell line. Clones derived from the infected line transform over months to IL-3 independency, are capable of increased proliferation, and develop chromosomal abnormalities.188

A series of mouse models in which the BCR-ABL was used to induce leukemogenesis have been described.189–197 Lethally irradiated mice have been reconstituted with marrow enriched for cycling stem cells infected with a BCR-ABL–bearing retrovirus. Fatal diseases with abnormal accumulations of macrophagic, erythroid, mast, and lymphoid cells develop.188 Classic CML did not occur, and complete transformation was not documented. The cell lines from spleen and marrow from mice with a BCR-ABL retrovirus infection were predominantly mast cells; however, in some cases these cell lines spontaneously switched to either erythroid and megakaryocytic, erythroid, or granulocytic lineages displaying maturation. They were transplantable (transformed) and contained the same proviral inserts as the original mast cell line.198 Murine marrow also has been infected with a retrovirus encoding p210BCR–ABL and transplanted into irradiated syngeneic recipients.189 Although several types of hematologic malignancies developed, a syndrome mimicking human CML also occurred. Mice transgenic for a p190BCR–ABL develop an acute lymphocytic leukemia (ALL) lymphoma syndrome190 that resembles human Ph-chromosome–positive ALL. When a p210BCR–ABL transcript is introduced into a mouse germ line (one-cell fertilized eggs), the p210 founder and progeny transgenic animals developed leukemia of B or T lymphoid or of myeloid origin after a relatively long latency period. In contrast, p190 transgenic mice exclusively developed leukemia of B-cell origin, with a relatively short period of latency. This finding was believed to be consistent with the apparent indolent nature of human CML during the chronic phase.191 When transgenic mice express p210BCR–ABL, the transgenes develop ALL, whereas the progeny develop a myeloproliferative disorder.192

Mouse models remain important for exploring the pathogenesis of the acute and chronic BCR-ABL–mediated leukemias in vivo and in examining the potential effects of new drugs targeted at BCR-ABL.199

BCR-ABL in Healthy Subjects

BCR-ABL fusion genes can be found in the leukocytes of some normal individuals using a two-step reverse transcriptase polymerase chain reaction assay. Thus, although BCR-ABL may be expressed relatively frequently at very low levels in hematopoietic cells, only infrequently do the cells acquire the additional changes necessary to produce leukemia. This may be a dosage effect.200

BCR-ABL and Signal Transduction

The tyrosine phosphoprotein kinase activity of p210BCR–ABL has been causally linked to the development of Ph-chromosome–positive leukemia in man.201–212 p210BCR–ABL is, unlike the ABL protein that is located principally in the nucleus, located in the cytoplasm making it accessible to a large number of interactions, especially components of signal transduction pathways.205,206,213 It binds and/or phosphorylates more than 20 cellular proteins in its role as an oncoprotein.206 A subunit of phosphatidylinositol 3′-kinase (PI3K) associates with p210BCR–ABL; this interaction is required for the proliferation of BCR-ABL–dependent cell lines and primary CML cells. Wortmannin, a nonspecific inhibitor of the p110 subunit of the kinase, inhibits growth of these cells.207

The pathways and interactions invoked by BCR-ABL acting on mitogen-activated protein kinases are multiple and complex.214,215

An RAF-encoded serine-threonine kinase activity is regulated by p210BCR–ABL. Downregulation of RAF expression inhibits both BCR-ABL–dependent growth of CML cells and growth factor–dependent proliferation of normal hematopoietic progenitors.208

The efficiency of cell transformation by BCR-ABL is affected by an adaptor protein that can relate tyrosine kinase signals to RAS. This involves growth factor receptor-bound protein-2 (GRB2). p210BCR–ABL also activates multiple alternative pathways of RAS.209 PI3K is constitutively activated by BCR-ABL, generates inositol lipids, and is dysregulated by the downregulation by BCR-ABL of polyinositol phosphate tumor suppressors, such as PTEN and SHIP1.213 Figure 90–5 demonstrates interaction of p210BCR–ABL with various mediators of signal transduction.

Figure 90–5.
Major intracellular signaling events associated with BCR/ABL. Constitutive activation of ABL protein tyrosine kinase (PTK) induces phosphorylation of the tyrosine moiety of various substrates, including autophosphorylation of BCR/ABL and complex formation of BCR/ABL with adaptor proteins. This process subsequently activates multiple intracellular signaling pathways, including RAS activation and phosphatidylinositol 3′-kinase (PI3-K) activation pathways. BCR/ABL also activates the c-MYC pathway, which involves ABL-SH2 domain. BCR/ABL inhibits apoptosis, possibly in part through upregulation of Bcl-2, and alters cellular adhesive properties, possibly by interacting with focal adhesion proteins and the actin cytomatrix. Broken lines indicate hypothetical pathways. ERK, extracellular signal-regulated kinase; FAK, focal adhesion kinase; JNK, Jun N-terminal kinase; MEKK, MEK kinase; Sos, Son-of-sevenless; STAT, signal transducer and activator of transcription.(From Gotoh A, Broxmeyer HE: The function of BCR/ABL and related proto-oncogenes. Curr Opin Hematol 4:3, 1997, with permission.)

Reactive oxygen species are increased in BCR-ABL–transformed cells and may act as a second messenger to modulate enzymes regulated by the redox equilibrium. An increase in these reactive oxygen products is postulated to play a role in the acquisition of additional mutations as a result of production of reactive oxygen species through the chronic phase, contributing to the progression to accelerated phase.213,216

The adaptor molecule CRKL is a major in vivo substrate for p210BCR–ABL, and it acts to relate p210BCR–ABL to downstream effectors. CRKL is a linker protein that has homology to the v-crk oncogene product. Antibodies to CRKL can immunoprecipitate paxillin. Paxillin is a focal adhesion protein210 that is phosphorylated by p210BCR–ABL. The p210BCR–ABL may be physically linked to paxillin by CRKL. CRKL binds to CBL, an oncogene product that induces B cell and myeloid leukemias in mice.211 The Src homology 3 domains of CRKL do not bind to CBL, but they do bind BCR-ABL. Therefore, CRKL mediates the oncogenic signal of BCR-ABL to CBL. The p120CBL and the adaptor proteins CRKL and c-CRK also link c-abl, p190BCR–ABL, and p210BCR–ABL to the PI3K pathway.212 The p120CBL also coprecipitates with the p85 subunit of PI3K, CRKL, and c-CRK. The p210BCR–ABL may, therefore, induce the formation of multimeric complexes of signaling proteins.217 These complexes contain paxillin and talin and may explain some of the adhesive defects of CML cells.218

Hef2 also binds to CRKL in leukemic tissues of p190BCR–ABL transgenic mice. Hef2 is involved in the integrin signaling pathway219 and encodes a protein that accelerates GTP hydrolysis of RAS-encoded proteins and neurofibromin. The latter negatively regulates granulocyte-monocyte colony-stimulating factor (GM-CSF) signaling through RAS in hematopoietic cells.220 P62DOK, a constitutively tyrosine-phosphorylated, p120RAS GAP-associated protein, which is rapidly tyrosine phosphorylated upon activation of the c-kit receptor,221 is also associated with ABL.222

Nuclear factor (NF)-B activation is also required for p210BCR–ABL-mediated transformation.223 Expression of p210BCR–ABL leads to activation of NF-B–dependent transcription via nuclear translocation.224

Cell lines that express p210BCR–ABL also demonstrate constitutive activation of Janus-associated kinases (JAKs) and signal transducers and activators of transcription (STATs), usually STAT5.225 STAT5 is also activated in primary mouse marrow cells acutely transformed by the BCR-ABL226; p210BCR–ABL coimmunoprecipitates with and constitutively phosphorylates the common subunit of the IL-3 and GM-CSF receptors and JAK2.227 Both ABL and BCR are also multifunctional regulators of the GTP-binding protein family Rho228,229 and the growth factor-binding protein GRB2, which links tyrosine kinases to RAS and forms a complex with BCR-ABL and the nucleotide exchange factor Sos that leads to activation of RAS.230

The p210BCR–ABL also activates Jun kinase and requires Jun for transformation.231 In some CML cell lines, p210BCR–ABL is associated with the retinoblastoma (Rb) protein.232 Loss of the neurofibromatosis (NF1) tumor-suppressor gene, a RAS GTPase-activating protein, also is sufficient to produce a myeloproliferative syndrome in mice akin to human CML resulting from RAS-mediated hypersensitivity to GM-CSF.233

Effects of BCR-ABL on Apoptosis

Whether p210BCR–ABL influences the expansion of the malignant clone in CML by inhibiting apoptosis is uncertain. In one study, the survival of normal and CML progenitors was the same after in vitro incubation in serum-deprived conditions and after treatment with X-irradiation or glucocorticoids.234 p210BCR–ABL inhibits apoptosis by delaying the G2/M transition of the cell cycle after DNA damage.235 The p210BCR–ABL also may exert an antiapoptotic effect in factor-dependent hematopoietic cells.236,237

p210BCR–ABL does not prevent apoptotic death induced by human NK or lymphokine-activated killer cells directed against CML or normal cells.238 In accelerated and blast phases, apoptosis rates were lower in CML neutrophils. G-CSF and GM-CSF considerably decreased the rate of apoptosis in CML neutrophils.239

Telomere Length

Patients with CML present with a somewhat shortened mean telomere length in granulocytic cells but not blood T lymphocytes at diagnosis, but considerable overlap exists in the distribution of telomere length with healthy individuals.240–242 The rate of shortening of telomere length during the chronic phase is correlated with a more rapid onset of accelerated phase.240,242 Telomerase reverse transcriptase (TERT) is the catalytic subunit, expression of which is closely correlated with telomerase activity. In CML CD34+ cells containing BCR-ABL, the expression of TERT is significantly lower than in normal CD34+ cells, consistent with accelerated shortening of telomeres in CML cells.243 A further significant decrease in telomere length occurs in the accelerated phase of CML. Telomerase activity is increased in the accelerated phase.244 When therapy permits restoration of Ph-negative cells in the blood, these cells have telomere length comparable to that in matched healthy controls.245

Clinical Features

Signs and Symptoms Chronic myelogenous leukemias (CMLs)

In the 70 percent of patients who are symptomatic at diagnosis, the most frequent complaints include easy fatigability, loss of sense of well-being, decreased tolerance to exertion, anorexia, abdominal discomfort, early satiety (related to splenic enlargement), weight loss, and excessive sweating.246–248 The symptoms are vague, nonspecific, and gradual in onset (weeks to months). A physical examination may detect pallor and splenomegaly. The latter was present in approximately 90 percent of patients at diagnosis, but with medical care being sought earlier, the presence of splenomegaly at the time of diagnosis is decreasing in frequency.247 Sternal tenderness, especially the lower portion, is common; occasionally, patients notice it themselves.

Uncommon presenting symptoms include those of dramatic hypermetabolism (night sweats, heat intolerance, weight loss) simulating thyrotoxicosis; acute gouty arthritis, presumably related in part to hyperuricemia; priapism, tinnitus, or stupor from the leukostasis associated with greatly exaggerated blood leukocyte count elevations249–251; left upper quadrant and left shoulder pain as a consequence of splenic infarction and perisplenitis; vasopressin-responsive diabetes insipidus252,253; and acne urticata associated with hyperhistaminemia.254 Acute febrile neutrophilic dermatosis (Sweet syndrome), a perivascular infiltrate of neutrophils in the dermis, can occur. In the latter situation, fever accompanied by painful maculonodular violaceous lesions on the trunk, arms, legs, and face are characteristic.255,256 Spontaneous rupture of the spleen is a rare event.257,258 Digital necrosis has been reported as a rare paraneoplastic event.259,260

In an increasing proportion of patients, the disease is discovered, coincidentally, when blood cell counts are measured at a periodic medical examination.

Laboratory Findings

Blood

The presumptive diagnosis of CML can be made from the results of the blood cell counts and examination of the blood film.26,246,247 The blood hemoglobin concentration is decreased in most patients at the time of diagnosis. Red cells usually are only slightly altered, with an increase in variation from small to large size and only occasional misshapen (elliptical or irregular) erythrocytes. Small numbers of nucleated red cells are commonly present. The reticulocyte count is normal or slightly elevated, but clinically significant hemolysis is rare.246,261,262 Rare cases of mild erythrocytosis263,264 or erythroid aplasia265,266 have been documented.

The total leukocyte count is always elevated at the time of diagnosis and is nearly always greater than 25,000/L (25 x 109/L); at least half the patients have total white counts greater than 100,000/L (100 x 109/L) (Fig. 90–6).26,246,247 The total leukocyte count rises progressively in untreated patients. Rare patients may have dramatic cyclic variations in white cell counts as much as an order of magnitude with cycle intervals of approximately 60 days.267,268 Granulocytes at all stages of development are present in the blood and are generally normal in appearance (Fig. 90–7). The mean blast cell prevalence is approximately 3 percent but can range from 0 to 10 percent; progranulocyte prevalence is approximately 4 percent; myelocytes, metamyelocytes, and bands account for approximately 40 percent; and segmented neutrophils account for approximately 35 percent of total leukocytes (Table 90–1). Often, there is a “myelocyte bulge” in which the differential count shows an exaggerated proportion of myelocytes compared to the proportion observed in normal persons. Hypersegmented neutrophils are commonly present.

Table 90–1. White Blood Cell Differential Count at the Time of Diagnosis in 90 Cases of pH-Chromosome–Positive Chronic Myelogenous Leukemia
Percent of Total Leukocytes (Mean Values)
Myeloblasts 3
Promyelocytes 4
Myelocytes 12
Metamyelocytes 7
Band forms 14
Segmented forms 38
Basophils 3
Eosinophils 2
Nucleated red cells 0.5
Monocytes 8
Lymphocytes 8
NOTE: In these 90 patients, the mean hematocrit was 31 mL/dL, mean total white cell count was 160 x 109/L, and mean platelet count was 442 x 109/L at the time of diagnosis.SOURCE: Hematology Unit, University of Rochester Medical Center.

Neutrophil alkaline phosphatase activity is low or absent in more than 90 percent of patients with CML.269–271 The mRNA for alkaline phosphatase is undetectable in neutrophils of patients with CML.272 The activity increases toward or to normal in the presence of intense inflammation or infection and when the total leukocytic count is decreased to or near normal with treatment.271,273 CML neutrophils regain alkaline phosphatase activity after infusion into leukopenic recipients, suggesting the effect of regulators or factors extrinsic to the neutrophils.274 In vitro, a monocyte-derived soluble mediator is capable of inducing increased alkaline phosphatase activity in neutrophils from CML patients.275 Neutrophil alkaline phosphate is decreased sporadically in a variety of disorders and conditions,276 but is decreased markedly and consistently in paroxysmal nocturnal hemoglobinuria,276 in hypophosphatasia,277 in approximately one-fourth of patients with idiopathic myelofibrosis, and in patients using androgens. Neutrophil alkaline phosphatase is increased in polycythemia vera, in 25 percent of patients with idiopathic myelofibrosis, in pregnant women, and in subjects with inflammatory disorders or infections. With the advent of specific markers, BCR-ABL in CML and JAK2 mutations in polycythemia, leukocyte alkaline phosphatase is of limited diagnostic use.

The proportion of eosinophils usually is not increased, but the absolute eosinophil count nearly always is increased. Rarely, eosinophils are so prominent that they dominate the granulocytic cells and lead to the designation Ph-positive eosinophilic CML. An absolute increase in the basophil concentration is present in almost all patients, and this finding can be useful in preliminary consideration of the differential diagnosis.26,278 Basophilic progenitor cells are increased in the blood.279 The proportion of basophils usually is not greater than 10 to 15 percent during the chronic phase but may, in rare patients, represent 30 to 80 percent of the total leukocyte count during chronic phase and lead to the designation of Ph-chromosome–positive basophilic CML.280 Flow cytometry using anti-CD203c provides very accurate assessment of the basophil frequency. Basophils may be hypogranulated or have an immature phenotype and may be left uncounted in an optical differential count. Anti-CD203c recognizes these cells as basophils.281 Granules of basophils in patients with CML, unlike normal basophils, contain mast cell -tryptase.281,282 Granulocytes containing both eosinophilic and basophilic granules (mixed granulation) are commonly present.283

The total absolute lymphocyte count is increased (mean: approximately 15 x 109/L) in patients with CML at the time of diagnosis284 as a result of the balanced increase in T-helper and T-suppressor cells.285 B lymphocytes are not increased.288 T lymphocytes also are increased in the spleen.286 NK cell activity is defective in CML patients as a result of decreased maturation of these cells in vivo287,288 and a decrease in the absolute number of circulating NK cells in patients with CML. The latter change can perhaps be related to increased apoptosis.289 The CD56 bright subset of NK cells is particularly decreased. These cells are reduced more as CML progresses, and they respond less to stimuli that recruit clonogenic NK cells compared to NK cells from normal subjects.290

The platelet count is elevated in approximately 50 percent of patients at the time of diagnosis and is normal in most of the rest.291 The median value in patients at diagnosis is approximately 400,000 cells/L (400 x 109 cells/L). The platelet count may increase during the course of the chronic phase. Platelet counts greater than 1,000,000/L (1000 x 109/L) are not unusual, and platelet counts as high as 5,000,000 to 7,000,000/L (5000–7000 x 109/L) have occurred. Thrombohemorrhagic complications of thrombocytosis are infrequent. Occasionally, the platelet count may be below normal at the time of diagnosis, but this finding usually signals an impending progression to the accelerated phase of the disease (see “Accelerated Phase and Blast Crisis of CML” below).

Functional abnormalities of neutrophils (adhesion, emigration, phagocytosis) are mild; are compensated for by high neutrophil concentrations; and do not predispose patients in chronic phase to infections by either the usual or opportunistic organisms.292–294 Platelet dysfunction can occur but is not associated with spontaneous or exaggerated bleeding. A decrease in the second wave of epinephrine-induced platelet aggregation is the most common abnormality and is associated with a deficiency of adenine nucleotides in the storage pool.295,296

Marrow

Morphology

The marrow is markedly hypercellular, and hematopoietic tissue takes up 75 to 90 percent of the marrow volume, with fat markedly reduced (see Fig. 90–7).297,298 Granulopoiesis is dominant, with a granulocytic-to-erythroid ratio between 10:1 and 30:1, rather than the normal 2:1 to 4:1. Erythropoiesis usually is decreased, and megakaryocytes are normal or increased in number. Eosinophils and basophils may be increased, usually in proportion to their increase in the blood. Mitotic figures are increased in number. Uncommonly a juxtamembrane domain mutant of KIT coincides with BCR-ABL in CML.299 Rare reports of marrow mastocytosis have been explained by a KIT mutation as an additional genetic abnormality or by dual clones in the marrow.300,301 Macrophages that mimic Gaucher cells in appearance are sometimes seen. This finding is a result of the inability of normal cellular glucocerebrosidase activity to degrade the increased glucocerebroside load associated with markedly increased cell turnover.302 Macrophages also can become engorged with lipids, which, when oxidized and polymerized, yield ceroid pigment. This pigment imparts a granular and bluish cast to the cells after polychrome staining; such cells have been referred to as sea-blue histiocytes.302

Collagen type III (reticulin fibrosis), which takes the silver impregnation stain, is commonly increased at the time of diagnosis in nearly half the patients,303 and is correlated with the proportion of megakaryocytes in the marrow.304,305 Increased fibrosis also is correlated with larger spleen size, more severe anemia, and a higher proportion of marrow and blood blast cells.

The marrows of CML patients have a mean doubling of microvessel density compared to healthy controls and have more angiogenesis in marrow than other forms of leukemia.306–308 This increased marrow vascularity decreases to normal after treatment.309

The marrow cells of approximately 50 percent of patients express cancer testis antigens, especially those encoded by HAGE genes.310

Progenitor Cell Growth

Cells that form colonies of neutrophils and macrophages or eosinophils (CFUs) are increased in the marrow and blood. The increase in CFUs in marrow is approximately 20-fold normal and in blood approximately 500-fold normal. The CFUs are of lighter buoyant density than those in normal marrow.95 More primitive progenitors that can initiate long-term cultures of hematopoiesis also are markedly increased.311 Spontaneous blood-derived granulocyte-macrophage colony growth is common, although CFUs also respond to growth factor stimulation.105

Cytogenetics

The marrow and nucleated blood cells of more than 90 percent of patients with clinical and laboratory signs that fall within the criteria for the diagnosis of CML contain the Ph chromosome (22q–) as measured by G-banding, and virtually all patients have the t(9;22)(q34;q11)(BCR-ABL) by fluorescence in situ hybridization. The Ph chromosome is present in all blood cell lineages (erythroblasts, granulocytes, monocytes, megakaryocytes, T- and B-cell progenitors) but is not present in the majority of blood B lymphocytes or in most T lymphocytes.49,51 Approximately 70 percent of patients in the chronic phase have the classic Ph chromosome in their cells.312 The remaining 20 percent also have a missing Y chromosome [t(Ph),–Y]; an additional C-group chromosome, usually number 8 [t(Ph),+8]; an additional chromosome 22q– but without the 9q+ [t(Ph), 22q–]; or t(Ph) plus either another stable translocation or another minor clone.75 These variations have not been shown to affect the duration of the chronic phase. Deletion of the Y chromosome occurs in approximately 10 percent of healthy men older than 60 years.313,314

Variant Ph chromosome translocations occur in approximately 5 percent of subjects with CML and involve complex rearrangements (three chromosomes), and every chromosome except the Y chromosome can be involved.315–319 The Ph chromosome, that is, 22q–, is present, but the gross exchange of chromosomal material involves a chromosome other than 9 (simple variant) or involves exchange of material among chromosomes 9 and 22 and a third or more chromosomes (complex variant; see Fig. 90–8). High-resolution techniques have indicated that 9q34-qter is transposed to 22q11 in simple and in complex translocations.320,321 Thus, the fusion of 9q34 with 22q11 seems to occur in the cells of most patients with CML.323 Complex translocations involving chromosome 3 have been notable.322–324 In rare cases, a reciprocal translocation with a chromosome other than 9 to chromosome 22 is larger than usual, and the posttranslocation shortening of the long arms of 22 is not apparent. This circumstance has been referred to as a masked Ph chromosome or masked translocation because the 22q– is not evident by microscopic examination,325,326 although t(9;22) may occur as judged by banding techniques or molecular probes.327Approximately 10 percent of patients have a deletion of the derivative 9 chromosome adjacent to the chromosome breakpoint. Although this deletion is thought to be an important factor in resistance to drug effects with IFN therapy, it does not appear to be significant with the use of imatinib.214

Molecular Probes

In a small proportion of patients with a clinical disease analogous to CML, cytogenetic studies do not disclose a classic, variant, or masked Ph chromosome. In these cases, use of a panel of restriction enzymes and Southern blot analyses with a molecular probe for the breakpoint cluster region on chromosome 22 nearly always detects rearrangement of fragments. This finding has led to the conclusion that almost all cases of CML have an abnormality of the long arm of chromosome number 22 (BCR rearrangement).328–332 Ph-chromosome–negative CML cells with BCR rearrangement can express p210BCR–ABL, and such patients have a clinical course similar to Ph-chromosome–positive CML.328,333–336

The ability to identify the molecular consequences of the t(9;22), that is, BCR rearrangement, mRNA transcripts of the mutant fusion gene, and p210BCR–ABL, has resulted in diagnostic tests supplementary to cytogenetic analysis.332 These tests include Southern blot analysis of BCR rearrangement,334–338 polymerase chain reaction (PCR) amplification of the abnormal mRNA,339 and a less complex variation on the latter, a hybridization protection assay.340

PCR can achieve a sensitivity of one positive cell in approximately 500,000 to 1 million cells. This extreme sensitivity requires special care in analysis and the inclusion of negative controls.341–344 Immunodiagnosis of CML by identification of p210BCR–ABL also is possible. This tumor-specific protein for CML is unique, based on the amino acids at the junction between the ABL and BCR sequences. Oligopeptides corresponding to the junctional amino acids have been synthesized and used as antigens345–348 to develop specific antibodies to p210BCR–ABL.

A multicolor FISH method to detect the BCR-ABL fusion in patients with CML is a rapid and sensitive alternative to Southern blot and PCR-dependent methods.349 For diagnostic purposes, FISH is simple, accurate, and sensitive, and can detect the various molecular fusions (e.g., e13a2, e14a2, e1a2).350–354 Interphase FISH is faster and more sensitive than cytogenetics in identifying the Ph chromosome. If the concentration of CML cells is very low, interphase FISH may not detect BCR-ABL, so it has limited use for detecting minimal residual disease.355 Hypermetaphase FISH allows analysis of up to 500 metaphases per sample in 1 day. Several factors influence the false-positive and false-negative rates of FISH identification of BCR-ABL, including definition of a fusion signal, nuclear size, and the genomic position of the ABL breakpoint.356 Double BCR-ABL fusion signals (double-fusion [D]-FISH) have been proposed as being more accurate than the fusion signal used in dual color (single-fusion) S-FISH, because in the latter case a small percentage of the normal BCR and ABL signals overlap.357

The frequency of cytogenetic analysis can be reduced if patients are monitored by molecular methods such as quantitative Southern blotting, FISH, quantitative Western blotting, or competitive reverse transcriptase (RT)-PCR. Molecular analyses can be performed on blood samples and therefore are much easier to use than cytogenetic analysis of marrow cell metaphases. Quantitative RT-PCR is the method of choice for monitoring patients for residual disease or reappearance of disease after marrow transplantation and for following response to tyrosine kinase inhibitors once routine cytogenetics and FISH are negative for the Philadelphia chromosome. Competitive PCR can detect reappearance of or increasing levels of RNA BCR-ABL transcripts prior to clinical relapse in patients after transplantation.358–360

Chemical Abnormalities

Uric Acid

An increased production of uric acid with hyperuricemia and hyperuricosuria occurs in untreated CML.361 Uric acid excretion often is two to three times normal in patients with CML. If aggressive therapy leads to rapid cell lysis, excretion of the additional purine load may produce urinary tract blockage from uric acid precipitates. Formation of urinary urate stones is common in patients with CML, and some patients with latent gout may develop acute gouty arthritis or uric acid nephropathy.362 The likelihood of complications from urate overproduction is greatly increased by starvation, acidosis, renal disease, or diuretic drug therapy.

Serum Vitamin B12-Binding Proteins and Vitamin B12

Neutrophils contain vitamin B12-binding proteins, including transcobalamin I and III (synonym: R-type B12-binding protein or cobalophilin).363–366 Patients with myeloproliferative diseases have an increased serum level of B12-binding capacity, and the source of the protein is principally mature neutrophilic granulocytes.363,364 The increase in transcobalamin level and the resultant increase in vitamin B12 concentration are particularly notable in CML, although any increase in the number of neutrophilic granulocytes, as in leukemoid reactions, can be accompanied by an increase in serum B12-binding protein levels and vitamin B12 concentration.366 The serum B12 level in CML patients is increased on average to more than 10 times normal.367 The increase is proportional to the total leukocyte count in untreated patients and falls toward normal levels with treatment, although increased B12 levels commonly persist even after the white cell count is lowered to near normal with therapy.

Pernicious anemia and CML may rarely coexist. In this situation, the tissues are vitamin B12 deficient, but the serum vitamin B12 level may be normal because of the elevated level of transcobalamin I, a binder with a very high affinity for vitamin B12.367

Whole Blood Histamine

Mean histamine levels are markedly increased in patients in chronic phase (median: ~5000 ng/mL) compared to healthy individuals (median: ~50 ng/mL); and, this elevation is correlated with the blood basophil count.368 Cases of exaggerated basophilia and disabling pruritus, urticaria, and gastric hyperacidity have occurred, associated with enormous increases (several hundred-fold) of blood histamine concentration.369,370

Serum Lactic Dehydrogenase, Potassium, Calcium, and Cholesterol

The level of serum lactic acid dehydrogenase (LDH) is elevated in CML.371 Pseudohyperkalemia resulting from the release of potassium from white cells during clotting372 and spurious hypoxemia or pseudohypoglycemia from in vitro utilization of oxygen or glucose by granulocytes can occur. Hypercalcemia373 or hypokalemia374 has occurred during the chronic phase of the disease, but such complications are very rare until the disorder transforms to acute leukemia. Elevated serum and urinary lysozyme levels are features of leukemia with greater monocytic components and are not features of CML.375 Serum cholesterol is decreased in patients with CML.376,377

Serum Angiogenic Factors

Angiogenin, endoglin (CD105), vascular endothelial growth factor (VEGF), -fibroblast growth factor, and hepatocyte growth factor are increased strikingly in the serum of CML patients.307,308,378,379

Special Clinical FeaturesBCR-ABL–Positive ThrombocythemiaEither of two syndromes—thrombocythemia with the Ph chromosome and BCR-ABL rearrangement or thrombocythemia without a Ph chromosome but with the BCR-ABL rearrangement—may precede the overt signs of CML or its accelerated phase.380–386 In general, the disease closely mimics classic thrombocythemia initially: marked platelet elevation, extreme megakaryocytic hyperplasia, normal or mildly elevated white cell count, no or very slight myeloid immaturity in the blood, and minimal anemia. Minor bleeding, such as epistaxis, erythromelalgia, or signs of thrombosis, such as cerebral or limb ischemia, are occasionally present.387 In some cases, the absolute basophil count is mildly elevated. Using immunostaining, Ph-positive thrombocythemia is proposed to be distinctive from Ph-negative thrombocythemia by small megakaryocytes in the former and by large clusters of megakaryocytes in the latter.387 However, this distinction was not found in other studies,388 has to be validated, and is difficult to use as a discriminator. In two studies, approximately 5 percent of patients with apparent essential thrombocythemia had a Ph chromosome.382,389 In another study, 2 of 121 patients with essential thrombocythemia had BCR-ABL transcripts, and 1 of these patients also had a Ph chromosome in the marrow cells,390 whereas in a different study, 4 of 32 patients with thrombocythemia had low levels of BCR-ABL transcripts in blood cells.391 Approximately 1 in 20 patients with CML present with the features of essential thrombocythemia.383,384 Evolution to blast crisis may occur.381,392,393 Thus, the frequency of Ph-chromosome–negative, BCR-ABL–positive thrombocythemia ranges from approximately 1.6 to 13 percent of patients, which may reflect in part the range of sensitivity of the detection method.389–391,394,395Neutrophilic CMLA rare variant of BCR-ABL–positive CML has been described in which the elevated white cell count is composed principally of mature neutrophils.396,397 The white cell count is lower on average (30,000–50,000/L) at the time of diagnosis than is the case with classic CML (median, 100,000–150,000/L). Moreover, patients with neutrophilic CML usually do not have basophilia, notable myeloid immaturity in the blood, prominent splenomegaly, or low leukocyte alkaline phosphatase scores. The cells of these patients have the Ph chromosome but have an unusual BCR-ABL fusion gene in that the breakpoint in the BCR gene is between exons 19 and 20. This breakpoint location results in fusion of most of the BCR gene with ABL (e19a2 type BCR-ABL), which leads to a larger fusion protein (230 kDa) compared to the fusion protein in classic CML (210 kDa; see Fig. 90–3). This correlation between genotype and phenotype has not been observed in all cases.398 This variant usually has an indolent course, which may be the result of very low levels of mRNA for p230 and the undetectable or barely detectable p230 protein in cells.399

Minor-BCR Breakpoint–Positive CML

A small proportion of patients with BCR-ABL–positive CML have the breakpoint on the BCR gene in the first intron (m-bcr), resulting in a 190-kDa fusion protein instead of the classic 210-kDa protein observed in most patients with CML (see Fig. 90–3). The m-bcr molecular lesion is similar to that observed in approximately 60 percent of patients with BCR rearrangement-positive ALL. In patients with m-bcr CML, monocytes are more prominent, the white cell count is lower on average, and basophilia and splenomegaly are less prominent than in disease with classic BCR breakpoint (M-bcr). The few reported cases had a short interval before either myeloid or lymphoid blast transformation developed.400,401

Hyperleukocytosis

Approximately 15 percent of patients present with symptoms or signs referable to leukostasis as a result of the intravascular flow-impeding effects of white cell counts greater than 300,000/L (300 x 109/L).249 Hyperleukocytosis is more prevalent in children with Ph-chromosome–positive CML.250 The effects of total leukocyte counts from 300,000 to 800,000/L (300–800 x 109/L) include impaired circulation of the lung, central nervous system, special sensory organs, and penis, resulting in some combination of tachypnea, dyspnea, cyanosis, dizziness, slurred speech, delirium, stupor, visual blurring, diplopia, retinal vein distention, retinal hemorrhages, papilledema, tinnitus, impaired hearing, and priapism.251 In asymptomatic patients with hyperleukocytosis, initial treatment with hydration and hydroxyurea usually can be used to decrease the white cell count. Hydroxyurea treatment should be designed to accomplish a gradual decrease in white cell count over a few days so as to avoid the tumor lysis syndrome. If signs of hyperleukocytosis are present, hydration, leukapheresis, and hydroxyurea can be used simultaneously; hydroxyurea dose should be selected to avoid exaggerated tumor lysis.

Concurrence of Lymphoid Malignancies

CML has an association with lymphoproliferation that can take four principal forms. (1) Patients may develop CML years after irradiation treatment of non-Hodgkin or Hodgkin lymphoma. (2) Approximately one-third of CML patients enter the accelerated phase of the disease by evolution and dedifferentiation of the CML clone into one that supports lymphoblastic proliferation (acute lymphoblastic transformation). (3) Patients may have concurrent lymphoproliferative or plasmacytic malignancies and CML. Lymphoma or lymphoblastic leukemia,402–408 essential monoclonal gammopathy,409,410 myeloma,411–413 and Waldenström macroglobulinemia414 have occurred in association with CML. Several cases of CML emergence in patients with established chronic lymphocytic leukemia (CLL) have been reported.415–417 A few patients have presented with simultaneous occurrence of the two diseases.418,419 A single case of lymphocytic leukemoid reaction simulating CLL that regressed as CML emerged has been reported.420 In some cases, the CLL lymphocytes did not contain the Ph chromosome, whereas the CML cells did, suggesting the presence of two independent clonal disorders.415,416,421,422 In other cases, the Ph chromosome was present in the myeloid and lymphoid cells, indicating a common origin.419 (4) Patients may present with Ph-chromosome–positive acute lymphoblastic leukemia and, following chemotherapy-induced remission, develop the features of typical CML.420

Differential Diagnosis

Diseases Mimicking CML

The diagnosis of CML is made based on the characteristic granulocytosis, white cell differential count, increased absolute basophil count, and splenomegaly coupled with the presence of the Ph chromosome or its variants (90% of patients) or a BCR rearrangement on chromosome 22 (>95% of patients).

Patients with other chronic hematopoietic stem cell diseases, such as polycythemia vera, essential thrombocythemia, or primary myelofibrosis, only occasionally have closely overlapping features. For example, the total white cell count is greater than 30 x 109/L in more than 90 percent of patients with CML and increases inexorably over weeks or months of observation, whereas the total white cell count is less than 30 x 109/L in more than 90 percent of patients with the three other classic chronic clonal myeloid diseases and usually does not change significantly over months to years. Polycythemia vera is associated with increased red cell mass and hemoglobin concentration and displays clinical signs of plethora; CML does not have these features. Patients with primary myelofibrosis invariably have marked teardrop poikilocytes and other severe red cell shape, size, and chromicity changes, as well as prominent nucleated red cells in the blood; CML rarely has these features. Patients with essential thrombocythemia have a platelet count greater than 450,000/L (450 x 109/L) and usually only mild neutrophilia (<20,000/L); the slight neutrophilia distinguishes it from the proportion (~25%) of CML patients with platelet counts greater than 450,000/L (450 x 109/L), who at the time of diagnosis have white cell counts above 25,000/L. In addition, patients with the clinical features of polycythemia vera or primary myelofibrosis do not have the Ph chromosome or BCR rearrangement in their blood and marrow cells, except in extremely rare cases. A very small proportion of patients with apparent essential thrombocythemia have BCR-ABL transcripts in their marrow and blood cells, and occasionally a Ph chromosome and may represent an atypical initial phase of CML (see “BCR-ABL–Positive Thrombocythemia” above). The presence of a mutation in the JAK2 gene in approximately 95 percent of patients with polycythemia vera and in approximately 40 to 50 percent of patients with primary myelofibrosis or essential thrombocythemia by current assay techniques is a very useful distinguishing feature when present. More sensitive assays for JAK2 may make this marker even more useful in discrimination of these myeloproliferative diseases from CML.423

Increased awareness of the features of related disorders, such as chronic myelomonocytic leukemia (CMML) and chronic neutrophilic leukemia, and an appreciation that older patients are prone to atypical clonal myeloid diseases, have minimized the inappropriate diagnosis of Ph-chromosome–negative CML, which should be avoided unless the clinical features are characteristic of classic CML and a masked Ph chromosome or BCR rearrangement is not found.

Reactive leukocytosis can occur with absolute neutrophil counts of 30,000 to 100,000/L (30–100 x 109/L). Usually these leukemoid reactions occur in the setting of an overt inflammatory disease (e.g., pancreatitis), cancer (e.g., lung), or infection (e.g., pneumococcal pneumonia). If the incitant is not apparent, the absence of granulocytic immaturity, basophilia, splenomegaly, and decreased neutrophil alkaline phosphatase activity argue against CML. The absence of a cytogenetic or molecular abnormality in chromosome 22 virtually eliminates classic CML as a consideration.

The precise diagnosis of CML is helpful in estimating the patient’s prognosis, determining the potential response to tyrosine kinase inhibitors, and assessing the timing of special therapies, such as allogeneic hematopoietic stem cell transplantation.

pH-Chromosome–Positive Clonal Myeloid Diseases and Aplastic Anemia

The Ph chromosome has been found rarely in patients with apparent polycythemia vera,27,424 polycythemia vera that later evolves into Ph-chromosome–positive CML,425–427 idiopathic myelofibrosis,428,429 and myelodysplastic syndrome (MDS).430,431 Molecular studies to determine the presence of the BCR-ABL were not performed in cases reported before 1985. Primary (essential) thrombocythemia with a Ph chromosome and/or BCR-ABL rearrangement in blood cells was discussed earlier (see “Special Clinical Features” above). Rare cases of aplastic anemia have presented with BCR-ABL–positive cells or have evolved into BCR-ABL CML.432,433 The frequency of this association is uncertain because of the inability to have sufficient cells for a cytogenetic analysis during the period of aplasia and the unavailability in past years of FISH or PCR to test for the BCR-ABL gene in patients with this clinical presentation.

Chronic myelogenous leukemias (CMLs)Therapy

Hyperuricemia

Hyperuricemia and hyperuricosuria are frequent features of CML at diagnosis or in relapse.434 The need for treatment of hyperuricemia is a function of the elevated pretreatment serum uric acid concentration, blood white cell concentration, spleen size, and dose of chemotherapy planned. If these variables suggest a high risk for a significant amount of cell lysis, allopurinol 300 mg/day orally and adequate hydration to maintain a good urine flow should be instituted prior to therapy. Allopurinol is associated with a high frequency of allergic skin reactions and should be discontinued after the blood leukocyte count and spleen size have d ecreased and the risk of exaggerated cell lysis has passed. If hyperuricemia is extreme, usually over 9 mg/dL, alkalinization of urine can be achieved with sodium bicarbonate, and rasburicase can be administered.435 Rasburicase is a recombinant urate oxidase that converts uric acid to allantoin. Rasburicase, unlike allopurinol, reduces the uric acid pool very rapidly, does not result in the accumulation of xanthine or hypoxanthine, and does not require alkalinization of urine facilitating phosphate excretion.436 Although the manufacturer recommends a dose every day for 5 days, several reports have indicated that one injection will produce a rapid and sustained decrease in serum uric acid, significantly decreasing the cost of therapy.437 Another alternative is to use allopurinol for a few days after one injection of rasburicase. A dose of 0.2 mg/kg of ideal body weight of rasburicase intravenously has been used.438

Initial Cytoreduction Therapy

Imatinib mesylate (imatinib) is now used as initial therapy in almost all patients with CML presenting in the chronic phase. In cases where the white cell count is markedly elevated, hydroxyurea can be used prior to or in conjunction with imatinib. If rapid cytoreduction is required because of signs of the hyperleukocytic syndrome, leukapheresis and hydroxyurea often are combined.

Leukapheresis

Leukapheresis can control CML only temporarily. For this reason, it is rarely used in chronic phase CML and is useful in only two types of patients: the hyperleukocytic patient in whom rapid cytoreduction can reverse symptoms and signs of leukostasis (e.g., stupor, hypoxia, tinnitus, papilledema, priapism),249–251 and in the pregnant patient with CML who can be controlled by leukapheresis treatment without other therapy either during the early months of pregnancy when therapy poses a higher risk to the fetus or, in some cases, throughout the pregnancy.439,440 Because of the large body burden of leukocytes in marrow, blood, and spleen, and the high proliferative rate in CML, leukocyte reduction by apheresis is less efficient than in other types of leukemias.249,251 Leukapheresis reduces the burden of tumor cells subject to chemotherapeutically induced cytolysis and thus the production and the excretion of uric acid. In hyperleukocytic nonpregnant patients, leukapheresis is best used in conjunction with hydroxyurea to ensure rapid and optimal reduction in white cell count.

Hydroxyurea

Hydroxyurea 1 to 6 g/day orally, depending on the height of the white cell count, can be used to initiate elective therapy.441 Urgent treatment of extraordinary total white cell counts may require higher doses. The dose of hydroxyurea should be decreased as the total white cell count decreases and usually is given at 1 to 2 g/day when the total white cell count reaches 20,000/L (20 x 109/L). The drug should be temporarily discontinued if the white cell count drops below 5000/L (5 x 109/L). If hydroxyurea is being used in combination with imatinib, the hydroxyurea usually is tapered and discontinued once a hematologic response to imatinib is observed.

Anagrelide

Anagrelide can be used for platelet reduction in patients who present with elevated platelet counts. This agent acts directly to decrease megakaryocyte mass, and it can lead to a precipitous fall in platelet counts. In occasional patients who still have significant thrombocythemia after imatinib is initiated, the combination of imatinib and anagrelide is associated with a normalization of platelet counts.442

Tyrosine Kinase Inhibitor Therapy

Imatinib Mesylate

Patients with newly diagnosed chronic phase CML should be started on imatinib, 400 mg/day by mouth. Imatinib is easier to use, induces a higher frequency of hematologic remission, a higher frequency of complete cytogenetic remission, and greater suppression of the CML clone (molecular remission) than therapy with interferon (INF)-. The goal of imatinib therapy is to decrease the cells bearing the t(9;22) translocation (leukemic cells) to the lowest levels possible, under which conditions normal (polyclonal) hematopoiesis is restored. The efficacy of imatinib is judged by measuring the three benchmarks: hematologic response, cytogenetic response, and molecular response (Table 90–2).443,444 These are used to determine its maximal effect. The time to achieve a maximal effect is variable and can range from months to years. Thus, as long as a patient is having a continued reduction in the size of the leukemic clone as judged by cytogenetic or PCR measurements, the drug is continued at 400 mg/day. If the patient stops responding before a complete cytogenetic remission or complete molecular remission is achieved, the dose can be increased to 600 mg/day or to 800 mg/day (400 mg every 12 hours), if tolerated. About two-thirds of patients who do not have a significant hematologic response or who relapse while receiving imatinib at a dose of 400 mg/day achieve a complete or partial hematologic response with higher doses, but few cytogenetic responses occur.445 Some patients without a cytogenetic response can enter a partial or complete cytogenetic response with higher doses of imatinib. Unfortunately, the responses to higher doses of imatinib in patients lacking a hematologic or cytogenetic response at 400 mg/day usually are transient.446,447

Table 90–2. Criteria for Extent of Imatinib Treatment Response
Hematologic response White cell count <10 x 109/L, platelet count <450 x 109/L, no immature myeloid cells in the blood, and disappearance of all signs and symptoms related to leukemia (including palpable splenomegaly) lasting for at least 4 weeks.
Major cytogenetic response Less than 35% of cells containing the Ph chromosome by cytogenetic analysis of marrow cells.
Complete cytogenetic response No cells containing the Ph chromosome by cytogenetic analysis of marrow cells.
Major molecular response Blood cell BCR-ABL/ABL ratio <0.05% (3-log reduction in PCR signal from mean pretreatment baseline value).
Complete molecular response Blood cell BCR-ABL levels undetectable (usually by nested RT-PCR method).

Patients with newly diagnosed chronic phase CML treated with imatinib, 800 mg/day, administered in two 400 mg doses every 12 hours had a frequency of 90 percent complete cytogenetic responses and 96 percent had at least a major cytogenetic response. At a median of 15 months, no patients had progressed and 63 percent showed blood BCR-ABL/ABL percentage ratios of less than 0.05 percent. Twenty-eight percent of patients had undetectable BCR-ABL blood levels.448 Despite these reports the current starting dose is customarily 400 mg/day, balancing both effectiveness and tolerability in newly diagnosed patients. Moreover, the more rapid response with higher doses of imatinib may not translate into a better long-term survival.

Doses of imatinib lower than 400 mg/day result in fewer complete cytogenetic responses and a shorter duration of complete cytogenetic response. Patients who are older and who have lower body weight may only tolerate a lower dose but they are less likely to achieve a complete cytogenetic response.449 If however, a patient is on a lower dose (e.g., 300 mg/day) for a special reason (body size or tolerance level) and achieves a complete hematologic and cytogenetic response within 12 months of onset of therapy, acceptable outcomes without excess toxicity may result.450

After 5 years of experience with imatinib, the proportion of patients achieving a complete molecular response continues to increase. Imatinib has been shown to be safe and well tolerated in the majority of patients during this period of observation.451 Some patients have been studied for up to 7 years; the proportion with an undetectable BCR-ABL level in blood cells increased from 7 percent at 36 months to 52 percent at 84 months. During this time a major molecular response was lost in approximately 25 percent of patients with a detectable blood cell BCR-ABL signal. No patients with an undetectable blood cell BCR-ABL signal lost their major molecular response status after a median followup of 33 months.452

At 5 years, of the patients treated, the cumulative incidence of complete cytogenetic response was reached in more than 75 percent and of major molecular response was reached in more than 50 percent. The estimated overall survival and progression-free survival was approximately 80 percent of patients treated with imatinib at 400 mg/day for that period. By 5 years, 25 percent had discontinued imatinib treatment because of an unsatisfactory response or toxicity. The 5-year probability of remaining in major cytogenetic response while still receiving imatinib was approximately 60 percent. Achieving a complete cytogenetic response correlated with progression-free survival, but achieving a major molecular response conferred no further survival benefit.453

Use of Imatinib in Patients with Variant Chromosomal Translocations or Breakpoints

Patients with variant Ph chromosome translocations have a similar prognosis to that of patients with classic Ph chromosome translocations who are treated with imatinib.454 (See Fig. 90–3 for a diagram of breakpoints.) Patients with the e13a2 (formerly designated b2a2) p210BCR-ABL translocation respond well to imatinib, with similar rates of complete cytogenetic remission.455 The e13a2 transcript may be more sensitive to imatinib than the e14a2 (formerly designated b3a2) transcript.456 In a patient with both e1a2 and e14a2 fusion transcripts, only the p210 e14a2 transcript disappeared, whereas the e1a2 transcript persisted during progression to blast phase. No mutation in the kinase domain of ABL was found.457 This finding indicates that different clones in an individual patient may have a different sensitivity to imatinib.

Response to Imatinib in Children and Older Patients

More than 80 percent of children with chronic phase CML who are treated with imatinib, 260 to 570 mg/m2, enter a complete cytogenetic remission. Weight gain is the most common side effect of imatinib.458 In patients who were older than age 60 years, similar cytogenetic response rates and survival rates were noted as in younger patients in the late chronic phase who were treated concurrently, suggesting that age is not usually a factor in response.459,460

Side Effects and Special Treatment Considerations

Imatinib is relatively well tolerated. Most adverse effects are manageable and seldom require permanent cessation of therapy. Reduction to subtherapeutic doses is not recommended; it is better to interrupt therapy for a time.461

Myelosuppression is common in CML patients, especially at treatment onset when the CML clone accounts for most of blood cells. Dose reduction to less than 300 mg/day is not advisable for myelosuppression. The drug should be stopped until blood counts recover. G-CSF and GM-CSF can prevent or treat neutropenia.462,463 Platelet transfusion may be used for severe thrombocytopenia. Patients with imatinib-induced chronic cytopenias have inferior responses.464 Myelosuppression is an independent adverse factor for achieving cytogenetic responses with imatinib.465 Severe irreversible marrow aplasia after imatinib exposure has been reported.466

The main side effects noted with imatinib include fatigue, edema, nausea, diarrhea, muscle cramps, and rash.467 Elevated hepatic transaminases can occur. Mild transaminase elevations often respond to glucocorticoid use.468 Hepatotoxicity is uncommon, occurring in approximately 3 percent of patients, usually within 6 months of onset of imatinib use. Acute liver failure has been described.469 The severe periorbital edema occasionally observed is postulated to be an effect on platelet-derived growth factor receptor (PDGFR) and KIT expressed by dermal dendrocytes. Surgical decompression of severe edema rarely has been required.470 Although no effects on spermatogenesis have been reported, women of child-bearing age are at risk of teratogenic effects on a fetus.470

Uncommon side effects include splenic rupture,471 cerebral edema and visual disturbances resulting from retinal edema,472 varicella-zoster infection,473 gynecomastia,474 immune-mediated hemolytic anemia,475 severe muscle edema,476 severe fluid retention,477 interstitial lung disease,478,479 and panniculitis.480 Hypophosphatemia481 and altered bone and mineral metabolism have occurred.482,483

Cutaneous reactions with imatinib therapy occur in approximately 15 percent of patients.484 Except for severe reactions (approximately 5% of patients), such as Stevens-Johnson syndrome, exfoliative dermatitis, and erythema multiforme, cutaneous reactions rarely require permanent discontinuation of therapy. With milder reactions, concomitant glucocorticoid therapy or brief discontinuation of imatinib with gradual reintroduction at a lower dose and then a gradual increase in dose can be accomplished.485,486 With very mild cases, concurrent treatment with antihistamine or other symptomatic therapy may be successful. Oral desensitization regimens have been described that allow some patients to continue imatinib therapy.487 Sweet syndrome with CML cell infiltration has been reported at the time of molecular remission.488 Pityriasis rosea,489 palmoplantar hyperkeratosis,490 and oral and cutaneous lichenoid reactions,491 have also been described. Hair repigmentation492 and hypopigmentation of the skin,493 probably related to the inhibition of the KIT receptor tyrosine kinase by imatinib, have been reported. Imatinib has been proposed as a therapy for vitiligo.494

Other Effects of Imatinib

Imatinib has been found to cause regression of marrow fibrosis.495 One study found that the extent of marrow fibrosis in CML is not a prognostic factor with imatinib therapy,496 whereas another study observed that although imatinib reverses marrow fibrosis in patients with CML, it does not change the unfavorable prognosis associated with fibrosis.497

Imatinib reverses exaggerated VEGF secretion in patients with CML,498 and it may reverse exaggerated marrow angiogenesis.499 It can reduce marrow cellularity and normalize morphologic features regardless of cytogenetic response.

Pharmacokinetic Considerations during Imatinib Therapy

Mean plasma trough concentration of imatinib and its metabolite CGP74588 obtained at about 1 month (presumptive steady state) was 979 ± 530 ng/mL. The rate of complete cytogenetic response and major molecular response was higher within the highest quartiles of imatinib trough levels.500 Some therapists suggest that imatinib plasma levels be checked in cases of suboptimal response in order to adjust the dose.501 Comedications and population covariates such as body weight and white cell count had no or minimal effect on imatinib clearance.502 Patients with CML on hemodialysis have been successfully treated with imatinib.503 Therapy interruptions and nonadherence with oral imatinib usage are common, and patient education and close monitoring are important to ensure compliance.504

No significant clinical responses to imatinib have been noted in patients with acute myelogenous leukemia (AML), MDS, Ph-chromosome–negative CML, or CMML without PDGFR or KIT mutations.505

Defining a Response to Imatinib

Table 90–2 contains definitions of hematologic, cytogenetic, and molecular responses. The median BCR-ABL levels for imatinib-treated patients can continue to decrease over at least 5 years. Table 90–3 lists the approximate milestones expected of patients treated with 400 mg/day of imatinib.444 There is variation in an individual patient’s time of maximal response. Consequently, if a patient has not met those precise milestones but shows a continued decrease in the proportion of Ph-chromosome–positive cells on cytogenetic examination of marrow, or if in a complete cytogenetic remission, a continued decrease in the level of the PCR signal for the BCR-ABL, imatinib should be continued. Loss of response is defined as loss of a complete hematologic or complete cytogenetic response, an increase of 30 or more percent in the number of Ph-chromosome–positive metaphases examined at 3-month intervals, development of new cytogenetic abnormalities, or an increase in the BCR-ABL/ABL ratio of one log or more on serial RT-PCR testing or into the range associated with metaphase positivity. Because of variability in PCR testing, these changes should be confirmed within 1 month. Patients who have 100 percent Ph-chromosome–positive cells after 6 months of therapy have a minimal chance of later achieving a major or complete cytogenetic response and may be offered allogeneic stem cell transplantation, if applicable.444,506

Table 90–3. Guidelines for Response to Imatinab Mesylate443,444
Time of Observation (months) Disease Response
Unsatisfactory Suboptimal Response Optimal Response
3 No HR PHR CHR
6 No mCyR mCyR MCyR
12 No MCyR MCyR CCyR
18 No CCyR CCyR MMR
CHR, complete hematologic response; CCyR, complete cytogenetic response; HR, hematologic response; mCyR, minor cytogenetic response; MCyR, major cytogenetic response; MMR, major molecular response; PHR, partial hematologic response.NOTE: Response is defined in Table 90–2. These data are applicable to therapy with imatinib, 400 mg/day, as initial therapy in chronic phase. Unsatisfactory or suboptimal implies need to consider change in treatment approach, as appropriate for that patient. Usually this change is an increase in the dose of imatinib, a shift to a second-generation tyrosine kinase inhibitor, or allogeneic stem cell transplantation, if eligible. These guidelines are approximate in that a patient showing continued response to imatinib can be continued on that therapy until a response plateau has been reached, at which time the response can be evaluated using the benchmarks noted. See text for further details.

Stopping Imatinib Therapy

Discontinuation of imatinib in 12 patients who had undetectable disease for at least 2 years resulted in 6 patients having a molecular relapse within 1 to 5 months (imatinib was reintroduced with a response) and 6 others remaining in molecular complete remission for a median of 18 months.507 There are numerous anecdotes of patients relapsing when imatinib was stopped. In patients with intolerable side effects on imatinib, the dose may be reduced in some cases without the loss of a complete molecular response.508 In occasional patients in whom imatinib is stopped, a cytogenetic response of up to 15 months has persisted.509–512 Because early, quiescent Ph-chromosome–positive cells (CD34+Lin–) are insensitive to imatinib in vitro,513 at present it is advisable to maintain treatment indefinitely until the criteria for cessation, if any, can be established in clinical trials.

Use of Imatinib in Pregnancy

Imatinib is possibly teratogenic. Normal newborns have been delivered by patients who conceived and ingested imatinib during early pregnancy.514–517 In 125 women exposed to imatinib during pregnancy, 50 percent delivered normal infants, and 25 percent underwent elective terminations, three of the latter following the identification of fetal abnormalities. Twelve other infants had abnormalities.518 The majority of patients who discontinue imatinib during pregnancy lose their complete hematologic remission and their cytogenetic responses.519 One fetal fatality during pregnancy as a result of a meningocele has been reported. Males treated with imatinib have fathered healthy infants.520 Current recommendations are to practice contraception during imatinib treatment or if pregnant at the onset of the disease, to consider IFN treatment until delivery.517,521 Imatinib does appear in breast milk.522

Secondary Chromosomal Changes with Imatinib Mesylate

Clonal abnormalities in cells lacking a detectable Ph chromosome or BCR-ABL rearrangements have been detected in patients undergoing imatinib therapy who previously were treated with IFN-.523,524 These cytogenetic changes were noted in 7 patients at a median of 13 months of imatinib therapy, and trisomy 8 was the most frequent abnormality. All of these patients had major cytogenetic responses to imatinib.523 In some patients, clonal evolution may be related to imatinib resistance.525 Clonal abnormalities may be present in up to 10 percent of patients taking imatinib.526 Some of these cases may be associated with an MDS, especially in those patients with previous exposure to cytarabine and idarubicin. The antiproliferative effect of imatinib allows restoration of a polyclonal hematopoiesis in complete cytogenetic remission, which might favor the manifestation of a Ph-chromosome–negative disorder.527 Some investigators have found that, with the possible exception of +8, +Ph, and i(17), additional chromosomal abnormalities at diagnosis are not associated with an inferior outcome.528,529 In contrast, another group found that development of trisomy 8 in patients taking imatinib, while associated with pancytopenia, did not result in signs of disease progression. In a series of 34 CML patients who developed Ph-chromosome–negative clones while taking imatinib, the most common abnormalities were trisomy 8 and monosomy 7. In 11 of these patients, no archival evidence of these clones was present before imatinib therapy was initiated, and none of the patients developed myelodysplasia.530 In patients treated at diagnosis with imatinib, 9 percent developed chromosomal abnormalities in Ph-negative metaphases. These appeared at a median of 18 months, and the most common abnormalities were –Y and +8. Most were temporary and had disappeared within 5 months. Only one patient with –7 progressed to AML.531 Cytogenetic clonal evolution may not be an important impediment to achieving a major or complete cytogenetic response with imatinib, but it is an independent poor prognostic factor for survival of patients in chronic and accelerated phases of CML.532 Imatinib therapy may overcome the poor prognostic significance of derivative chromosome 9 in CML.533

Development of Imatinib Mesylate Resistance

The development of resistance to imatinib is not surprising.534–538 Its specificity and “snug fit” into the ABL-kinase pocket provide the ideal circumstance for resistance.539 Some cases demonstrate primary resistance to imatinib, and gene profiling has demonstrated differential expression of about 46 genes in responders compared to nonresponders.540 Even in patients with complete cytogenetic response, malignant progenitors at the LTC-IC stage persist. Chronic phase CML stem cells are resistant to imatinib and are genetically unstable.541 These cells have a high level of BCR-ABL transcription, and they are thought to express transporter proteins that result in abnormal imatinib flux.542 Mathematical models suggest that imatinib rapidly eliminates differentiated leukemic progenitors, but does not deplete leukemic stem cells. Such models predict the probability of developing resistant mutations and can estimate the time that resistance will emerge.543 Intermittent administration of G-CSF exposure may promote elimination of CML CD34+ cells.544,545 Imatinib upregulates CXCR4 expression, and thus might promote survival of quiescent CML stem cells by enhancing their interaction with marrow stroma.546

Several potential mechanisms of resistance include BCR-ABL amplification in the presence of imatinib,547–549 P-glycoprotein–mediated drug efflux,550,551 altered drug metabolism,538 acquisition of BCR-ABL–independent signaling characteristics,549 and point mutations in the ABL kinase domain that alter imatinib binding. There is evidence that each of these mechanisms of resistance may have clinical relevance.

Expression of the OCT-1 cellular transporter, which mediates drug influx, is thought to be important for imatinib but not dasatinib effectiveness.553,554 Many CML patients who have a suboptimal response to imatinib have low OCT-1 activity but this can be overcome with higher doses of imatinib or use of dasatinib.554,554 CML CD34+ cells overexpress the drug transporter ABCG2, but imatinib mesylate is not a substrate for this protein.555

Amplified gene expression and increased BCR-ABL protein expression are often reported in resistant patients. Duplication of the Ph-chromosome and isodicentric chromosomes are a possible mechanism of resistance to imatinib.556,557

Mutations in the ABL kinase domain may predate imatinib treatment,558 and several BCR-ABL kinase domain mutants associated with imatinib resistance remain sensitive to the drug, suggesting a need for characterization before a resistant phenotype can be attributed to the given mutation.559 The mutant clone does not always have a proliferative advantage.560 Some of these mutations may lie outside the kinase domain, and more than 40 such mutations have been described. Screening early phase CML patients for mutations before the start of imatinib therapy is not cost-effective because of their low incidence, but in patients with evidence of an increase in CML cells while on imatinib, mutation searches are indicated.561 BCR-ABL kinase domain point mutations are rare in those who have had good cytogenetic responses to imatinib, and when detected in that setting, their presence does not always predict relapse.562 Mutations in the ABL portion of the BCR-ABL oncogene are present in approximately 40 percent of patients who do not achieve a hematologic or cytogenetic response to imatinib. ABL mutations were found in those patients with both primary and acquired resistance. Amino acid substitutions in seven residues accounted for 85 percent of all mutations associated with resistance.563 The mutations most associated with resistance are Thr315ILe, Gly250Glu, Glu255Lys, and Thr253His substitutions. Few of the described mutations directly affect imatinib binding.537,564 Mutations in the ABL–ATP phosphate-binding loop (P-loop) are most closely associated with a poor prognosis,565 and these P-loop mutations predict for disease progression. Overall survival is worse for P-loop and for T315I mutations but not significantly different for other mutations.566

Second-generation BCR-ABL inhibitors (see “Dasatinib and Nilotinib” below) are able to overcome imatinib-resistant mutants, with the exception of the T315I mutations. This mutation results in steric hindrance which precludes access of some inhibitors to the ATP-binding pocket of the ABL kinase domain.567 Based on the crystal structure, the kinase domain of ABL T315I can be predicted by Aurora kinase inhibitors. The inhibitor binds in an active conformation of the kinase domain in the ATP-binding pocket.568 In a series of 27 patients with T315I mutation, survival was dependent on stage of disease, with many of the chronic phase patients described as having an indolent course.569 Agents such as IFN- and homoharringtonine have been proposed as salvage therapy for those with the T315I mutation.570

In some cases of resistance associated with imatinib, other signal pathways independent of BCR-ABL may become important in cell proliferation.571 These include heat shock protein 70,572 survivin,573 LYN kinase,574 SRC,575 and GRB2.576

Dose escalation, combination therapy, and treatment interruption have been proposed as means to overcome drug resistance.577 Combination therapy from the outset578 also has been proposed to prevent development of resistance. Treatment interruption to stop clonal selection of resistant cells has been proposed.575 Gene expression profiles may be useful to predict the clinical effectiveness of imatinib for CML treatment, thereby allowing individualized therapy from the outset.578 In patients with relapse or resistance, alternative approaches include increasing the dose of imatinib or switching to dasatinib or nilotinib.579

Dasatinib and Nilotinib

Dasatinib and nilotinib, two second-generation tyrosine kinase inhibitors, are used in cases of imatinib resistance or intolerance. Trials are underway to ascertain whether use of these agents at time of diagnosis will improve response rates and obviate later resistance.

Dasatinib is 325-fold more potent than imatinib and responses occur among all ABL mutant genotypes with the exception of T315I.580 As a dual inhibitor of SRC and ABL kinases, dasatinib is able to bind to BCR-ABL with less stringent conformational requirements.581 Dasatinib,100 mg/day, is administered in chronic phase CML.582,583 The major cytogenetic response rate is approximately 50 percent, and molecular responses occur as well.584 In patients resistant to imatinib, dasatinib, 140 mg/day (70 mg q12h), resulted in a higher proportion of major cytogenetic responses, complete cytogenetic responses, and major molecular responses than did 800 mg/day (400 mg q12h) of imatinib. Treatment failure was decreased and progression-free survival was improved with dasatinib.585 Unlike imatinib, dasatinib penetrates the blood–brain barrier.586

The major adverse event with dasatinib use is cytopenia. In addition to hematologic toxicity, fluid retention, diarrhea, and skin rash also can occur. Dasatinib may be more effective in the presence of F359I ABL mutation as compared with imatinib and nilotinib.587 Unlike the case with imatinib, dasatinib cellular uptake is not affected by OCT-1 activity, which is a substrate of the efflux proteins, ABCB1 and ABCG2.588 Resistance to dasatinib is often found with point mutations at residue 315 or 317.589

Nilotinib is an orally bioavailable, ATP-competitive inhibitor of BCR-ABL. It is FDA approved for patients in chronic phase CML resistant or who are intolerant to imatinib. It is approximately 30 times more potent than imatinib.590 Like imatinib, it does not induce apoptosis in CD34+ CML cells.591 A dose of 400 mg every 12 hours induced approximately 40 percent of patients who were resistant to or intolerant of imatinib into a major cytogenetic response, and approximately 30 percent into a complete cytogenetic response, with all resistance-inducing ABL mutations except T315I.

Adverse effects included neutropenia and minimal other side effects such as hyperbilirubinemia and hypophosphatemia.592 Nilotinib can cause electrocardiographic QT interval prolongation, so caution is required with concurrent medications that can prolong the QT interval.593 There is preclinical data to show that despite binding at the same site in the same target kinase, use of imatinib and nilotinib in combination may have additive or synergistic effects as BCR-ABL inhibitors.594

Combined Therapy

Agents that have been proposed for use in combination to improve response rates or to overcome resistance to imatinib have included IFN-, cytarabine, daunorubicin, homoharringtonine, multiagent chemotherapy, arsenic trioxide, and decitabine, with some supporting in vitro data.595–600 Combining imatinib with chemotherapeutic agents is more myelosuppressive, and final effects on response rates and survival have yet to be determined.601 Trials examining combinations of imatinib and either dasatinib or nilotinib are underway.602

Other Combinations Proposed to Overcome Imatinib Resistance

Several inhibitors of other signal transduction mediators involved in the downstream effects of BCR-ABL have been proposed for use in imatinib-resistant CML. These inhibitors include the JAK2 inhibitor AG490,603 SRC kinase inhibitors, mTOR (mammalian target of rapamycin) inhibitors, such as rapamycin,604 the proteasome inhibitor bortezomib,605,606 histone deacetylators,607,608 PI3K or MEK (mitogen-activated kinase) inhibitors, such as wortmannin and LY294002,609 and inhibitors of prenylation of RAS-related proteins downstream of BCR-ABL. These agents include the bisphosphonate zolendronate610 and farnesyltransferase inhibitors.611,612 The farnesyltransferase inhibitors SCH66336 and R115777 have shown some activity in CML.613–616 Imatinib resistance often is associated with restored activation of the BCR-ABL signal transduction pathway, suggesting that BCR-ABL remains a valid target to overcome resistance in these cases.617 BCR-ABL point mutations isolated from patients with imatinib-resistant CML remain sensitive to inhibitors of the BCR-ABL chaperone heat shock protein (hsp) 90, such as geldanamycin.618 Many of these agents have not yet entered clinical trials. Some are being used in conjunction with imatinib in resistant cases.

Disease Prognosis and Monitoring during Imatinib Therapy

Treatment failure should lead to alterations in therapeutic strategy.619 For patients treated initially with imatinib, BCR-ABL expression in cytogenetic responders and nonresponders was similar. BCR-ABL expression became significantly different 3 months after treatment and became increasingly different between responders and nonresponders with continued therapy at 6, 9, and 12 months.620

One mode of monitoring patients undergoing imatinib therapy is to measure blood counts at least once per month and to obtain marrow samples every 6 months until a complete cytogenetic remission is obtained.621 Thereafter, marrow samples are obtained yearly to monitor for other clonal abnormalities. Quantitative RT-PCR is performed every 3 months on blood or marrow. A one log increase in the level of BCR-ABL reactivity, confirmed on a repeat sample at least 1 month later, suggests a loss of response to treatment. In patients who do not have a complete hematologic response at 3 months, or a major cytogenetic response after 6 to 12 months, other therapeutic options are considered.622 The molecular response after 2 to 3 months of therapy is a strong predictor of clinical and cytogenetic response.623 Sequencing the BCR-ABL kinase domain can reveal emergence of resistant clones and is useful if there is an insufficient initial response to imatinib (see Table 90–3) or any sign of loss of response, such as relapse to Ph-positive status, a 1-log increase in BCR/ABL transcript ratio, or loss of a major molecular response (MMR).444 In patients receiving second-generation tyrosine kinase inhibitors, those who have no cytogenetic response at 3 to 6 months should be considered for allogeneic transplantation or switched to an alternative therapy in a clinical trial. After 12 months, those with a major cytogenetic response had a significant survival advantage over those with lesser responses.624

Summary of Imatinib Effects

Imatinib was first used experimentally for CML treatment in June 1998. Although it has completely altered the treatment approach to CML, its use has raised several questions. Studies require long-term followup of survival, but the degree of cytogenetic response, and degree of molecular response can be used as surrogate endpoints.625,626 The durability of cytogenetic and molecular responses in the face of persistent minimal residual disease during imatinib therapy require further followup of larger numbers of patients, especially because more than 95 percent of cases have molecular evidence of disease at 2 years.626

The success of imatinib therapy has diminished the use of allogeneic stem cell transplantation in chronic phase CML.627 Imatinib trials show a 95 percent progression-free survival at 24 months, but patients usually have evidence of ongoing molecular disease. Because stem cell transplantation is associated with high toxicity and mortality rates, deciding when and for whom to use this modality while responses to imatinib are ongoing can be difficult. For patients who lose or never achieve an imatinib-induced major cytogenic response and for whom an acceptable donor is available, allogeneic stem cell transplantation should be considered.628 Another therapeutic issue that has arisen is whether to use dasatinib or nilotinib before allogeneic stem cell transplantation in those in whom imatinib response has been absent or suboptimal.629 Some therapists would first increase the dose of imatinib, and if unsuccessful try dasatinib or nilotinib, and only if those steps were unsuccessful consider allogeneic transplantation.

Interferon

Prior to the approval of imatinib mesylate for upfront therapy in CML, INF- was often used as initial therapy. A complete cytogenetic response with IFN- was uncommon (13%), but 10-year survival rates in responders were approximately 70 percent.630 Cytogenetic responses to IFN- were stable and durable.631 Approximately 50 percent of complete responders become long-term survivors. Common toxicities of INF- use include fatigue, low-grade fever, weight loss, liver function test abnormalities, hematologic changes, and neuropsychiatric symptoms. Most studies have shown no benefit of high-dose IFN- compared with low-dose IFN- for chronic phase CML (5 million units/m2 per day vs. 3 million units/m2 five times per week).632 Low-dose IFN- minimizes toxicity and cost. Pegylated IFN-, which has a longer half-life, can be administered as a once-per-week injection at 6 mcg/kg per week.633 Dose-limiting toxicities are neurotoxicity, thrombocytopenia, fatigue, and liver dysfunction. In later studies, 4.5 mcg/kg per week was proposed as the optimal dose because of the toxicity at higher doses. A single weekly dose of 450 mcg pegylated IFN- has also compared favorably to the standard IFN- dose in terms of toxicity and response rates.634 Pegylated IFN- plus low-dose cytosine arabinoside administered weekly is effective but has significant toxicity in patients with CML.635

Overall survival is improved in imatinib-treated patients compared with patients treated with IFN- or IFN- plus cytarabine.636 Nevertheless, among all patients who attained a major or complete cytogenetic response at 12 months, the survival rate was comparable in either case. IFN- has also been proposed as an immune stimulant to consolidate imatinib remissions because additive effects have been noted.637,638 Conversely, those treated initially with INF- who achieve a complete cytogenetic response have an improved molecular response with imatinib.639,640 Some patients intolerant to a tyrosine kinase inhibitor may be treated successfully with INF-.

Use of Other Chemotherapeutic Agents in Chronic Phase

Hydroxyurea

The major side effect of hydroxyurea is an extension of its pharmacologic effect, that is, reversible suppression of hematopoiesis, often with megaloblastic erythropoiesis. The median survival of patients with CML treated with hydroxyurea alone is approximately 5 years. Studies with high-dose hydroxyurea indicate that marrow metaphase cells in some patients lose the Ph chromosome either partially or completely after such therapy.641 The drug may be very useful in patients of advanced age, in patients with comorbid conditions, and in patients in whom imatinib and IFN cannot be tolerated or are ineffective. Hydroxyurea often is used for initial cytoreduction. Chronic use of hydroxyurea is associated with leg ulcers.642

Cytarabine

IFN-2b combined with cytarabine (20 mg/m2 per day for 10 days per month) in the chronic phase was associated with a greater proportion of major cytogenetic response at 12 months after randomization and with greater survival prolongation than was IFN alone.643 Toxicities with these drug combinations were greater, and this combination has been replaced by tyrosine kinase inhibitor therapy.

Busulfan

Once the mainstay of treatment for the chronic phase, busulfan usage now is rare.644 It is used primarily as part of the preparative regimen for allografting or autografting. It may be used occasionally in older patients who do not tolerate tyrosine kinase inhibitors.

Homoharringtonine

Homoharringtonine, a plant alkaloid, can induce responses, including cytogenetic responses, in patients in the late chronic phase.645

Other Cytotoxic Agents

Intensive multidrug regimens have been used in an attempt to eradicate the Ph-chromosome–positive clone and have lead to prolongation of remission or cure of the disease. This approach has not significantly increased survival.646

Other Potential Therapeutic Agents in CML

The farnesyltransferase inhibitors lonafarnib and tipifarnib have been combined with imatinib and have activity after imatinib failure.647,648 The hypomethylation agent decitabine has activity in imatinib refractory CML.649 Berbamine, a natural small molecular compound, has in vitro activity against primary CML cells.650 Adaphostin, a tyrphostin, inhibits CML cell growth, including those cell populations resistant to imatinib by inducing oxidative stress.651 INNO-406, a dual BCR-ABL/LYN inhibitor, suppresses the growth of CML cells in the central nervous system where imatinib has limited penetration.652 This agent also enhances autophagy in CML cells.653 Agents that disrupt autophagy when combined with histone deacetylase inhibitors such as suberoylanilide hydroxamic acid (SAHA) are able to overcome imatinib resistance in preclinical models.654 The SRC-ABL inhibitor, SKI-606 (bosutinib), is able to overcome most resistance-mediating ABL mutations, except T315I, and it has entered clinical trials.655 Several third-generation inhibitors are being developed for inhibition of the T315I mutation.656 MicroRNA technology may eventually play a role in CML treatment,657 and synthetic BCR-ABL siRNA (small interfering ribonucleic acid) has been used in a patient with resistant CML, postallografting with inhibition of BCR-ABL noted.658

Ribozymes targeting BCR-ABL mRNA have been used as CML treatment,659,660 and these approaches probably will have the most utility for in vitro purging of CML marrow cells before autotransplantation.661,662

Immunotherapy

Several antigens have been proposed as targets of immune therapy for CML. These antigens include BCR-ABL itself, PR1, Wilms tumor protein-1 (WT1), minor histocompatibility antigens, CML-66, CML-28, and survivin.663,664 Other targets are VEGF and hsp90.665 A BCR-ABL fusion peptide, used as a vaccine, can elicit a specific T-cell immune response.666,667 CML-derived dendritic cells can process and present endogenous BCR-ABL fusion proteins to CD4+ T lymphocytes in an HLA class II-restricted antigen presentation.668 NM23-H2, an HLA-A32 restricted tumor associated antigen aberrantly expressed in tumors such as CML, can generate reactive T cells after transplantation.669 Immunization with GM-CSF–producing tumor vaccines to enhance a vaccine antitumor effect is being studied in CML.670 Numerous peptides from the BCR-ABL fusion region have been identified as vaccine candidates.671 Peptides from the e14a2 BCR-ABL junction have been examined in vaccine trials, elicit T-cell responses, and demonstrate molecular responses of a delayed nature in those who have had a major cytogenetic response to imatinib. Randomized trials with these vaccines have not yet been reported.672

Radiotherapy

Splenic irradiation may be useful occasionally in subjects who have entered the accelerated or advanced chronic phase and are troubled with extreme splenomegaly with splenic pain, perisplenitis, and encroachment of the spleen on the gastrointestinal tract.673 Splenic irradiation may palliate symptoms for a short time.674

Radiotherapy may be useful for extramedullary tumors, which may occur occasionally in bone or soft tissue during the late chronic or accelerated phase.

Splenectomy

Splenectomy does not prolong the chronic phase of CML, delay the onset of the accelerated phase, enhance sensitivity to standard or intensive chemotherapy, or prolong survival of patients.675 In carefully selected patients with symptomatic thrombocytopenia unresponsive to therapy, mechanical discomfort, hypercatabolic symptoms, and portal hypertension, splenectomy may be useful. Postoperative morbidity from infection, thrombosis, or hemorrhage has been high, with mortality rates up to 10 percent reported.676 Splenectomy performed before allografting has not been found to influence the severity of graft-versus-host disease (GVHD) or survival after allogeneic stem cell transplantation.677 Splenectomy may reverse poor graft function after allogeneic transplantation, but hyposplenism may trigger or worsen chronic extensive GVHD, leading to increased morbidity and mortality.678

Treatment of Chronic Phase CML during Pregnancy

Treatment of chronic phase CML during pregnancy is sometimes needed to prevent placental insufficiency from hyperleukocytosis. Imatinib use during pregnancy has the risk of a teratogenic effect.514–517 IFN can be used during pregnancy with minimum risk of teratogenicity. Eight patients treated with IFN from the first trimester have been described, and each of these pregnancies resulted in normal infants, except for one with mild neonatal thrombocytopenia. All infants had normal growth.680 Hydroxyurea may be useful during the second and third trimester but should be avoided in the first trimester.517,679 Leukapheresis in the first trimester (or longer) also can be used to avoid fetal drug exposure early in pregnancy (see “Leukapheresis” above). It is important to use imatinib after delivery to achieve the best outcome. Further observation may show it to be safe later in pregnancy. Although controversial, stopping and later restarting imatinib may not result in as favorable an outcome of therapy. 517

High-Dose Chemotherapy with Autologous Stem Cell Infusion -Chronic myelogenous leukemias (CMLs)

Since the availability of imatinib, autografting in CML is rarely used.681 Ph-chromosome–negative stem cells are present in most patients with CML at the time of diagnosis. Techniques that use these cells to reconstitute hematopoiesis after high-dose therapy have been developed.682 Ph-chromosome–negative progenitors can be mobilized with G-CSF and collected from the blood of patients who have responded to prior treatment with IFN or imatinib.683 Such cells also can be collected after recovery from chemotherapy regimens, such as after idarubicin and cytarabine, followed by G-CSF stimulation.682 G-CSF was used for at least 4 days while imatinib treatment was continued for stem cell mobilization in 58 patients with a complete cytogenetic response. The cells were collected in two cytapheresis procedures in 74 percent of patients, and the cells of 84 percent of those cytapheresis products were negative for the Ph chromosome.684

In another series, stem cells were mobilized in 32 patients in complete cytogenetic remission after imatinib, with uninterrupted imatinib therapy in 50 percent of patients and with imatinib temporarily withheld in approximately 50 percent. Blood levels of BCR-ABL were not changed by the use of G-CSF.685 In yet another series, 13 of 15 patients were successfully mobilized with G-CSF while receiving imatinib, and 28 percent of stem cell harvests were negative for BCR-ABL mRNA. No change in blood BCR-ABL transcript level was noted after stem cell mobilization as assessed by RT-PCR.686 No series of patients autografted with cells mobilized while they were receiving imatinib have been reported.687 Autografting might find a role in cases of imatinib resistance,688 or to reduce the level of residual disease in cases without a molecular response.689 Imatinib can be effective and safe in chronic phase CML patients who have previously undergone autografting,690 although increased hematologic toxicity occurs.

Allogeneic Stem Cell Transplantation -Chronic myelogenous leukemias (CMLs)

Until imatinib became available in 2000, allogeneic transplantation was used in most new patients with CML who were younger than 65 years of age and who had a suitable donor. The advent of imatinib treatment and the projected survival of patients with a complete cytogenetic remission has changed the indications for transplantation in CML.691,692 There has been a marked reduction in number of transplants performed for CML worldwide and a decline in the proportion performed in first chronic phase.693,694 Although no randomized trials of imatinib versus transplantation have been or are likely to be conducted, there is circumstantial evidence that survival is superior for populations of patients treated with drugs who have a complete cytogenetic remission as compared to transplantation.695 Allografting continues to play a prominent role in the treatment of patients with suboptimal imatinib responses, who are refractory or intolerant to tyrosine kinase inhibitors, and remains the optimal therapy in those who progress to accelerated phase or blast crisis.

Patients in the chronic phase of CML who are younger than 65 years and who have an identical twin,696 or a histocompatible sibling,697,698 or who are younger than 55 years with access to a histocompatible unrelated donor,699 can be transplanted after intensive therapy, usually with cyclophosphamide and fractionated total-body irradiation (TBI) or a combination of busulfan and cyclophosphamide. Busulfan can be administered as an intravenous preparation and as a single daily dose.700 When targeted steady-state busulfan levels are utilized, a 3-year survival rate of 86 percent and a disease-free survival rate of 78 percent with no age effect is achieved.701 With nonmyeloablative or “reduced-intensity” conditioning regimens, older patients and those with comorbidities can undergo successful allografting.

Myeloablative Allogeneic Transplants-Chronic myelogenous leukemias (CMLs)

Stem cell transplantation from HLA-compatible siblings results in engraftment and an actual or projected long-term survival in 45 to 70 percent of recipients.702–704 In patients older than age 50 years, survival rates are slightly less at 5 years. The risk of CML relapse is approximately 20 percent, with a plateau of relapse at 5 to 7 years. Transplanted T lymphocytes, especially if activated by a (mild) GVHD, may be an important factor in preventing leukemic relapse. This phenomenon, referred to as graft-versus-leukemia reaction, is thought to suppress the leukemic process through T-cell–mediated cytotoxicity.697 The relative benefit of marrow compared to mobilized blood stem cells as the source of the allograft has not been established.705,706 Mobilized blood stem cells engraft more rapidly but may be associated with more chronic GVHD. The majority of survivors have no evidence of residual leukemia.707

For younger patients who do not have a histocompatible sibling, an unrelated donor or a mismatched family member as a source of stem cells is feasible. The toxicity of this procedure is greater than that of an HLA-identical sibling donor transplant. Five-year disease-free survival is approximately 40 percent.708 Younger patients with cytomegalovirus-seronegative donors who are matched at the HLA-DRB1 allele by molecular methods fare better.709 When class I HLA genes are typed with molecular methods, an improvement in matching and better outcomes using unrelated donors are expected. When matched-unrelated donor and sibling donor transplants were compared, unrelated donor transplants had increased risk of graft failure and acute GVHD, but only a slightly poorer survival and disease-free survival. For patients who survived to 1 year, only a slightly inferior disease-free survival was observed.710 The rate of extensive chronic GVHD is up to 60 percent with unrelated donor transplants, but 63 percent disease-free survival in younger CML chronic phase patients has been reported. Cord blood stem cell transplantation from an unrelated donor has also been used in adults with CML.711

Pretransplantation imatinib is not associated with increased transplant-related morbidity or decreased survival, but those who are transplanted with suboptimal response to imatinib or loss of response to imatinib do poorer, probably related to a higher disease burden at the time of transplantation and more aggressive disease.712,713 Second-generation tyrosine kinase inhibitors do not increase transplant-related toxicity.714 Disease status after allografting can be monitored with cytogenetic studies, PCR, or FISH analysis. A positive PCR assay 3 months after allogeneic transplantation has not been found to correlate with an increased risk of relapse compared with PCR-negative patients. A positive assay at 6 months and beyond is associated with subsequent relapse. In one series, 42 percent of patients with a positive PCR assay at 6 to 12 months relapsed versus 3 percent with a negative assay.715 Paradoxically, patients who remain BCR-ABL positive more than 36 months after transplantation have little propensity for relapse.715 Serial quantitative RT-PCR analysis of blood specimens has been proposed to distinguish patients destined to relapse.716 Patients who remain in remission have undetectable, low, or falling BCR-ABL levels on sequential analysis. After 6 to 9 months, these levels are undetectable in most cases. Recognition of relapse at the molecular level may allow for early therapeutic intervention.

Killer immunoglobulin-like receptors (KIRs) are expressed by NK cells and subpopulations of T cells. NK clones from a single individual can vary substantially in the type of KIR molecules they express. The ligands for several of the inhibitory KIR have been shown to be subsets of HLA class I molecules. Missing KIR ligands in recipients lead to less relapse and increased GVHD based on NK alloreactivity.717 KIR ligand mismatch has also been found to be an important prognostic factor in achieving molecular responses after transplantation for CML.718 Increased frequency of Treg cells characterized as CD4+, CD25-high are associated with higher rates of relapse after allografting in CML.719

Nonmyeloablative Allogeneic Transplants- Chronic myelogenous leukemias (CMLs)

Nonablative regimens have been developed in an attempt to expand the indication for allogeneic transplantation to older patients. These regimens rely on immunosuppressive therapy to allow engraftment of cells that potentially will generate a graft-versus-leukemia effect. These procedures in general are associated with acceptable degrees of engraftment, less mortality, similar rates of GVHD, and possible durable effects on persistent or recurrent disease.720 Approximately 60 percent of patients, mostly in initial chronic phase (median age: 50 years) and transplanted with reduced-intensity conditioning regimens, had a 3-year survival and about one-third had a 3-year progression-free survival.721 Patients no longer in first chronic phase do not fare as well.722 Conditioning regimens include fludarabine and busulfan,723,724 low-dose TBI and fludarabine, and low-dose TBI and cyclophosphamide, but no prospective randomized trials comparing regimens or comparing ablative and nonmyeloablative transplant approaches have been conducted.723,725,726 In one case, imatinib given concurrently with nonmyeloablative stem cell transplantation did not compromise engraftment and resulted in a cytogenetic remission in a patient with CML in blast crisis.727

Use of Tyrosine Kinase Inhibitors after Stem Cell Transplant

In patients treated with stem cell transplantation before the wide availability of imatinib, complete cytogenetic remissions with imatinib treatment can occur if treated at relapse after allografting and after donor lymphocyte infusion (DLI) fails to give a response. Such remissions may include a molecular response.728 Complete responses after imatinib therapy have been noted in accelerated or blast phase CML persisting after stem cell transplantation.729 In another series, 45 percent of relapsed patients had a complete cytogenetic response of up to 28 months and without significant GVHD.730 In a series of 28 adults with relapse after allogeneic stem cell transplantation who then received imatinib, the response rate was 74 percent, and the complete cytogenetic remission rate was 35 percent. Five patients had recurrence of GVHD, and 13 had previous DLI infusions.728 In one series, imatinib was able to generate complete molecular remissions in 26 percent of chronic phase patients after allografting, with full donor chimerism usually observed.731 Prophylactic administration of imatinib after transplantation has been used for patients at high risk of relapse.732 Posttransplantation imatinib may also postpone the requirement for donor leukocyte infusions with its attendant risks of GVHD and marrow aplasia.733 Some have found that donor leukocyte infusions are superior to imatinib therapy in preventing relapse and increasing leukemia-free survival.734 Today, virtually all patients receive imatinib initially and receive transplantation only if refractory to or relapse after imatinib and a second-generation tyrosine kinase inhibitor.

Immunotherapy: Adoptive Cell Therapy for Posttransplantation Relapse

Substantial evidence indicates that the effectiveness of allografting in CML does not result solely from the eradication of the leukemic clone with high-dose chemoradiotherapy conditioning regimens, but also from adoptive immunotherapy provided by lymphocytes in the allograft, the graft-versus-leukemia effect (see “Myeloablative Allogeneic Transplants” above).735 This phenomenon has been recreated to produce a therapeutic response by infusing the lymphocytes from the stem cell donor after a relapse following allogeneic stem cell transplantation.736,737 The overall response rate to DLI is approximately 75 percent. The response rate is higher when this approach is used early after detecting a relapse by PCR,738 compared to use after a hematologic or cytogenetic relapse. Patients with a short interval between transplantation and DLI have a higher probability of response than patients with longer intervals. Responses are the same with related versus unrelated donors.739 Some patients show a very rapid decline of BCR-ABL transcript levels (<6 months after DLI), whereas other patients demonstrate PCR negativity only over a longer period.740 The responses to DLI can be durable.741 Molecular responses can occur in up to two-thirds of patients.742

The main toxicities of DLI have been the induction of GVHD and myelosuppression. Chronic GVHD can occur in up to 60 percent of cases.743 Attempts to diminish these toxicities have included use of CD8-depleted DLIs and infusion of smaller numbers of T cells.744,745 Lower initial cell dose is associated with less myelosuppression, the same response rate, better survival, and less DLI-related mortality, leading to suggestions that the initial dose should not exceed 0.2 x 108 mononuclear cells/kg.746 The initial doses should be lower when matched unrelated donors are used. Donor lymphocytes can also be transfected with vectors containing the herpes simplex virus genome in a replication defective form. If GVHD occurs, the lymphocytes can be eradicated with systemic ganciclovir treatment. The ultimate utility of such currently experimental approaches is still unmeasured.747 IFN after DLI may improve responses,748,749 and imatinib may synergize with DLI to foster rapid molecular responses after relapse.750 Methods for administering more specific immune effector cells have been sought, but these approaches are not in widespread clinical use.751

Course and Prognosis

Several large studies of treatment during the 1970s and early 1980s reported similar survival rates of patients with CML who were treated with standard chemotherapy, that is, busulfan or hydroxyurea, during the chronic phase.752–760 Median survival ranged from 39 to 47 months, the 5-year survival rate was approximately 25 to 35 percent of patients, and the 8-year survival rate was 8 to 17 percent of patients. A large randomized study comparing hydroxyurea to busulfan showed a significant prolongation of chronic phase with hydroxyurea644 and a further prolongation with IFN therapy. Only a small fraction of patients remained in the chronic phase from 10 to 25 years.761–768 The Surveillance, Epidemiology, and End Results Program of the National Cancer Institute gathers national statistics based on cancer registries in the United States. Table 90–4 lists the 5-year survival rates from these observations. Because these data are a mixture of the pre- and postimatinib therapy periods, they underestimate the 5-year life expectancy of patients with CML who are given imatinib treatment.

Table 90–4. Chronic Myelogenous Leukemia: 5-Year Relative Survival Rates (1996–2004)
Age (years) Percent of Patients
<45 77.2
45–54 76.5
55–64 62.3
65–74 38.7
>75 23.9
NOTE: These most recent data underestimate the effects of imatinib mesylate treatment between 2005 and the current date.SOURCE: Data from Surveillance, Epidemiology, End Results Cancer Statistics, 5-Year Survival Rates, Table 13-15, All Races and Sexes. National Cancer Institute, Washington, DC. Available at www.seer.cancer.gov.

At the time of diagnosis, the variables most closely associated with duration of chronic phase and thus survival are percent blasts in the blood, liver and spleen size, and total basophil plus eosinophil count. Using these variables in large numbers of patients, the population segregates into three risk groups: better risk, with a median survival of approximately 5.0 years; intermediate risk, with a median survival of 3.5 years; and poor risk, with a median survival of 2.5 years.769–771 In the better-risk group, 40 percent are alive at 7 years; in the poor-risk group, 10 percent or fewer are alive at 7 years. These figures are based on patients who were treated principally with busulfan. Treatment of chronic phase by hydroxyurea and then by IFN extended the median survival by approximately 18 months with the former, and by approximately 36 months with the latter drug in patients treated with these agents. Generalizations to all patients with CML based on these studies are not possible because many patients, especially those older than age 65 years, could not tolerate optimal doses of IFN. Thus, the results are for the select group of patients who remained on treatment with these agents. The median survival also has been dramatically improved by imatinib treatment. One projection indicated the median survival for all patients so treated will exceed 15 years.772 It is as yet uncertain how long patients on imatinib can remain in remission on average.691,773 In a 5-year followup of patients receiving imatinib for newly diagnosed CML, complete cytogenetic responses were seen in approximately 70 percent by 12 months and in nearly 90 percent by 60 months. In a major study, an estimated overall survival at 84 months of followup was 86 percent.774 Resistance rates decline with each passing year, and adverse effects have not emerged over time.775 Those who require 1 year or more of imatinib therapy to attain a complete cytogenetic remission have comparable rates of molecular response, progression-free, and overall survival to those who achieve cytogenetic remission sooner.776 In patients with failure to respond or intolerance to imatinib the estimated 3-year survival rate was approximately 70 percent for patients in chronic phase. Survival in chronic phase was better when subsequent therapy was nilotinib or dasatinib as compared to allogeneic hematopoietic stem cell transplantation or to others agents. This result was at a median followup of 2 years.777 Older patients have lower response rates when treated in late chronic phase, but if they attain a complete cytogenetic response, no difference was found in the level of molecular response.776

Studies linking the precise (3′) location of the breakpoint in the BCR gene with shortened duration of chronic phase778 have not been confirmed.779 Prior to the introduction of imatinib therapy, most patients died as a result of conversion from the chronic to the accelerated phase of the disease or blast crisis.780 It is too soon to determine the cause of death in patients treated with inhibitors of the BCR-ABL kinase. Very rare spontaneous cytogenetic, but not molecular, remissions of CML have been reported,781,782 but the disease may reappear in that case.783

Several other ancillary factors are associated with poor prognosis in CML, including marrow angiogenesis and marrow fibrosis784; telomere length shortening785; cellular VEGF expression786; large deletions at the t(9;22) breakpoint787; derivative chromosome 9 deletions788,789; increased marrow fiber content and reduction of medullary erythropoiesis790; higher CD7 expression by CD34-positive cells791; absence of cadherin 13 expression792; and persistence of malignant hematopoietic progenitors in CML in complete cytogenetic remission after imatinib.793 Deletion of the 5′ abl region on der(9), present in approximately 9 percent of patients with CML, occurs at the time of formation of the t(9;22) translocation and may be associated with a slightly worse prognosis.794 The prognosis may not be worse with imatinib therapy. The type of BCR breakpoint (5′ or 3′) does not affect the course of chronic phase CML.779,795 CML that occurs after treatment of other cancers appears to have comparable clinical and survival characteristics as de novo CML.796 Histamine levels during treatment with imatinib have been found to be of prognostic importance.797

Several prognostic scales have been proposed in CML, including the Sokal and Hasford systems for patients at the time of diagnosis and the European Bone Marrow Transplantation Consortium Risk Score for patients undergoing allogeneic stem cell transplantation,798 in which performance status is added to the five original variables (age, spleen size, blast cell count, basophil and eosinophil count, and platelet count [Hasford score]), which has been validated with good discrimination for survival.799 The Sokal score based on age, spleen size, platelet count, and percent myeloblasts was developed much earlier during the busulfan era of treatment and was less accurate in patients treated with IFN. A simple prognostic scale that includes donor type, stage of disease at time of transplantation, age of recipient, sex of donor and recipient, and interval between diagnosis and transplantation has been proposed to predict outcome of allogeneic stem cell transplantation.800 These prognostic scales require revalidation given the dramatic impact of conversion to universal imatinib therapy. There is evidence that a patient with chronic phase CML and a favorable Sokal Score at the time of diagnosis has a higher proportion of hematologic and cytogenetic responses than other patients.801 Low neutrophil count and poor cytogenetic response at 3 months of imatinib therapy may predict a poor overall outcome,802 but this observation requires validation.803 Cytogenetic response as a surrogate marker for survival appears to be useful in patients undergoing imatinib therapy.804

Detection of Minimal Residual Disease

Detection of minimal residual disease by molecular probes makes possible the identification of approximately 1 cell in 1,000,000 that is derived from the CML clone.805 Techniques used to monitor residual disease have been reviewed.806–808 PCR permits observation of regression or persistence of subclinical disease following therapy and of progression of subclinical disease prior to the disease becoming overt, and it is therefore critical for monitoring responses to CML treatment.809,810 The stable persistence of subclinical disease does not invariably predict early relapse.811,812

The risk of misinterpreting negative results of RT-PCR is increased when very small numbers of transcripts are present.806 The dilution threshold for reproducible amplification is 250,000 cells. mRNABCR–ABL can also be detected in single progenitor colonies after culture.813 A good correlation has been found between the proportion of Ph-chromosome–positive metaphase cells and levels of mRNABCR–ABL.814–816 Efforts are underway to standardize the technique for measuring and reporting real-time RT-PCR,817 and serial measurements are required for treatment decisions based on rises in transcript numbers.818 Some studies show that marrow values tend to be higher than blood in real-time RT-PCR assays, but both follow a similar trend during treatment.819 Interchanging these may lead to misinterpretation of disease status.819 In patients on imatinib, who have major molecular responses, confirmed by real-time quantitative PCR, no marrow cell cytogenetic abnormalities were found, indicating that patients with major molecular responses do not require regular marrow examinations for cytogenetics.

Through utilization of quantitative PCR, an increase of mRNABCR–ABL expression has been found to precede disease progression. This increase was detected up to 16 months before laboratory or clinical parameters showed phenotypic transformation of the malignant clone.820 The technique of detecting minimal residual disease is highly sensitive and is subject to false-positive reactions. Nested, competitive RT-PCR is more sensitive than RT-PCR, but RT-PCR, when normalized for the total ABL transcripts, can be used to monitor CML patients during therapy.821 Patients who achieve a major molecular remission (expressed as a 3-log reduction from median baseline value) at the time of achieving a complete cytogenetic response have been found to have longer cytogenetic remissions than those without this magnitude of molecular response.822 The achievement of a 2-log molecular response at the time of a complete cytogenetic response or a 3-log response anytime thereafter is an independent prognostic marker of progression-free survival.823 Other studies, however, show that patients who achieve complete cytogenetic response do not derive additional benefit from a complete molecular response.824

Interphase FISH may have a false-positive rate of 5 to 10 percent.825,826 FISH is not standardized, but a large number of cells can be rapidly analyzed (100–500). With D-FISH probes, which flank the breakpoints of the BCR and ABL genes, the false-positive rate is only approximately 0.2 percent.825 Fixation, specimen preparation, and hybridization conditions may account for differing false-positive ranges and scoring criteria.827 In CML patients treated with imatinib, FISH for BCR-ABL on interphase blood neutrophils, but not unselected white cells, correlates with marrow cytogenetics.828 FISH and RT-PCR can be useful complementary techniques,829 but FISH is generally not suitable for monitoring minimal residual disease.830

For patients who are undergoing allogeneic stem cell transplantation, the kinetics of minimal residual disease in either standard or nonmyeloablative transplants differ. BCR-ABL/ABL ratios were 0.2 percent with reduced-intensity transplants versus 0.01 percent in transplantation patients with traditional conditioning regimens in the first 3 months. By 12 months, however, 20 percent of patients who received standard transplants and 50 percent of patients who received reduced-intensity transplants had reached a level less than 0.01 percent, supporting the concept of different kinetics of disease eradication between the two transplantation modalities.831 Patients who relapse after allografting have reappearance and/or rising levels of BCR-ABL transcripts.832 Use of quantitative RT-PCR early (3–5 months) after stem cell transplantation can project long-term outcomes.833 When RT-PCR was negative, the 3-year risk of relapse was 16.7 percent; when RT-PCR was positive at a ratio of less than 0.02 percent, the relapse rate was 42.9 percent; and when RT-PCR was positive at a level greater than 0.02 percent, the relapse rate was 86.5 percent. Another group found that detection of blood BCR-ABL at 18 or more months after transplantation was associated with a highly significant risk of relapse and that patients who had a positive test result but failed to relapse generally had only one positive test result at a low copy number.834 Performance of qualitative PCR at regular intervals after allogeneic transplant (every 2–4 months in the first year and every 6 months thereafter) is appropriate. If the PCR results are persistently positive or become positive, quantitative PCR should be performed at monthly or shorter intervals. Molecular relapse is defined as a 10-fold increase of PCR positivity without any signs of cytogenetic relapse.835 Detection of increasing recipient chimerism by FISH for the male chromosome in sex-mismatched donor–recipient pairs or variable number of tandem repeats after allogeneic transplantation or after DLI infusion also is usually associated with a relapse.836,837

Imatinib therapy is associated with a rapid decrease in BCR-ABL transcript levels. Nested PCR transcript levels parallel cytogenetic response, and imatinib is superior to IFN or cytarabine in terms of the speed and degree of molecular responses, but residual disease is rarely eliminated.838 When quantitative RT-PCR is utilized for patients undergoing imatinib therapy, almost all patients have evidence of residual disease.839,840 Disadvantages of quantitative RT-PCR in monitoring imatinib response include lack of standardization, inability to detect clonal evolution, and current lack of widespread availability.825 The use of random pentadecamer primers may improve detection of BCR-ABL transcripts in CML,841 and flow cytometric assays of phosphotyrosine levels may be valuable for serial evaluation of disease response.842 Table 90–5 outlines suggested monitoring for chronic phase patients undergoing imatinib therapy.

Table 90–5. Guidelines for Monitoring of Patients in Chronic Phase Who Are Undergoing Imatinib Mesylate Therapy
1. At diagnosis, before starting therapy, obtain Giemsa-banding cytogenetics and measure BCR-ABL transcript numbers by quantitative PCR using marrow cells. If marrow cannot be obtained, use FISH on a blood specimen to confirm the diagnosis.
2. At 3, 6, 9, and 12 months after initiating therapy, perform FISH for t(9;22) on blood cells and quantitative PCR for BCR-ABL transcripts.
3. At 6 months obtain marrow cytogenetics for cells with Ph chromosome. Repeat every 6 months until complete cytogenic response. If there is a CCyR at 6 months, this does not need to be done at 12 months.
4. Once CCyR is obtained, monitor quantitative PCR on blood cells every 3 months. Continue yearly marrow examination of marrow cytogenetics to identify clonal evolution, if indicated.
5. These guidelines presume continued response to imatinib until complete cytogenetic response achieved. If this does not occur see text for approach.
CCyR, complete cytogenetic response; FISH, fluorescence in situ hybridization.SOURCE: Adapted in part from: www.nccn.org.

Accelerated Phase and Blast Crisis of CML

Definition

In all patients with chronic phase CML, the disease has the potential to evolve into a more aggressive, more symptomatic, and troublesome phase, which is poorly responsive to the therapy that formerly controlled the chronic phase. The failure of therapy to restore or maintain near-normal red cell and white cell counts, increased spleen size, increased numbers of marrow blasts and blood basophils, loss of the sense of well-being, and appearance of extramedullary tumors are the most consistent clinical hallmarks of the metamorphosis of the chronic to the accelerated phase of CML. The most objective findings are a blood blast percentage greater than 10, a platelet count less than 100,000/L (100 x 109/L), blood basophils greater than 20 percent, and new clonal cytogenetic abnormalities accompanying the Philadelphia chromosome.843

The terminology used has included accelerated phase, acute phase, acute transformation, or, in its most dramatic expression, blast crisis, but the metamorphosis, which can be acute, that is, myeloblastic or lymphoblastic crisis, often is more gradual and manifested by severe dysmorphic hematopoiesis, refractory splenomegaly, and extramedullary tumor masses, hence the preference for transformation or accelerated phase to describe this transition from a controllable to a poorly controlled malignancy. Blast crisis is the most severe manifestation of the accelerated phase and can occur abruptly or after a period of worsening disease. Blast crisis is in effect the evolution to overt acute leukemia.

Pathogenesis

Effect of Tyrosine Kinase Inhibitors in Rate of Progression

The advent of tyrosine kinase inhibitor therapy for CML has resulted in a marked increase in the duration of a subclinical chronic phase, with normal blood counts and spleen size, often with the loss of identifiable Ph chromosome-bearing cells in blood and marrow, and sometimes with the loss of laboratory evidence of the BCR-ABL oncogene as judged by PCR. This therapeutic advance has greatly delayed the evolution to accelerated phase and blast crisis, but the risk for such a conversion exists since under experimental conditions CML stem cells do not undergo apoptosis when exposed to BCR-ABL tyrosine kinase inhibitors and BCR-ABL-positive cells return in virtually all patients if tyrosine kinase therapy is interrupted.

Blast Crisis Stem Cells

The onset of accelerated phase is thought to occur in a BCR-ABL-bearing granulocyte-monocyte progenitor. Experimental844,845 and theoretical846 evidence supports this concept. This locale for clonal evolution also could explain the reversion to chronic phase in some patients in whom the suppression of the advanced phase of the disease is achieved. Thus, during chronic phase there is the interplay between polyclonal normal hematopoietic stem cells and CML stem cells (see Chap. 85, Fig. 85–4) and in the acute phase a third stem cell pool (second cancer stem cell pool) is added that initiates and sustains accelerated phase and blast crisis.

Molecular and Genetic Alterations

The transformation of chronic phase CML to accelerated phase and then blast crisis or directly to blast crisis is thought to be the result of seven or more molecular processes: (1) maturation arrest, (2) failure of genome surveillance, (3) failure of adequate DNA repair, (4) development of a mutator phenotype, (5) telomere shortening, (6) loss of tumor-suppressor function, and (7) unknown factors.847,848

Progression of chronic phase to accelerated phase is marked by an increase in BCR-ABL expression.849,850 Superimposed on the increased transcription of mRNABCR–ABL are additional cytogenetic abnormalities that are added to the persistent Ph chromosome in approximately 50 to 65 percent of patients.843,850–853 In lymphoid blast crisis, in which the blast cells have a lymphocytic phenotype, acquisition of p16/ARF mutations occurs in approximately 50 percent of cases, and RB gene mutations occurs in approximately 20 percent of cases. In myeloid blast crisis, in which blast cells have a myeloid phenotype, approximately 25 percent of cases have cells containing a p53 mutation.854 The possible role of loss of p53 function in fostering transformation of a human chronic phase CML clone has been demonstrated in transgenic mice in which p53 function was abrogated.855 p51/p63 is a member of the p53 gene family that is not mutated in chronic phase but is mutated in approximately 8 percent of patients in blast crisis.856

Progression of the clone to a more malignant clone is reflected in a more disordered growth and maturation pattern of progenitor cells in culture, ultimately mimicking the growth failure of acute leukemia,851 and in increased morphologic and functional abnormalities of blood cells,857,858 eventuating in a block in maturation and replacement of blood and marrow by blast cells.

Approximately 65 percent of patients have cytogenetic abnormalities in addition to the Ph chromosome. A double Ph chromosome, trisomy 8, and isochromosome 17p are the secondary changes most commonly seen.854,859 Because the frequency of trisomy 8 was greater after treatment with busulfan compared to hydroxyurea, the frequency of secondary chromosomal changes may be quite different after imatinib mesylate therapy.854 Clonal instability has also been found in cases of lymphoid blast crisis. Clones distinct from those identified later may be detected before overt lymphoid transformation. Identification of these abortive clones suggests clonal instability before the onset of transformation, which might have prognostic value.860 FISH has been used to determine which cells have secondary cytogenetic abnormalities, and these cells often are not the blast cells. This finding suggests that some chromosomal abnormalities merely denote genomic instability.861 The abnormal mRNA and protein product p210BCR–ABL are present in the marrow and blood cells of patients who have transformed to acute leukemia.862–864

Although the breakpoint site on M-bcr was thought to be correlated with the time of the onset of the accelerated phase,865 subsequent studies have not indicated a correlation between length of chronic phase and the specific site of the BCR-ABL fusion.866 Rare cases have displayed deletion of the BCR-ABL fusion gene, loss of transcription of the message, and loss of expression of the p210 tyrosine kinase after transformation, the latter finding indicating the abnormal protein kinase may not always play a unique role in sustaining the acute state.867 In contrast, the frequent response, albeit temporary, to imatinib suggests that the mutant BCR-ABL product usually plays a role at this stage of the disease.

Numerous molecular changes identified in the cells of patients with acute transformation that might contribute to the increased malignant behavior of the CML clone, include activation of the N-RAS gene,868,869 rearrangement of the p53 gene,869–872 hypermethylation of the calcitonin gene,873 and methylation of the ABL1 gene.874 One report described p53 mutations in 17 percent of blast crisis patients. An association between the failure of CML cells to express the retinoblastoma 1 gene product and acute blast crisis with a megakaryoblastic phenotype has been reported.875 Homozygous deletions of the p16 tumor-suppressor gene are associated with lymphoid transformation of CML,876 but such deletions are not seen in the chronic phase and in myeloid blast crisis. p16 is also known as the cyclin-dependent kinase 4 inhibitor gene and is located on chromosome 9p21.877,878 This gene inhibits the kinase CDK-4, which regulates a cell-cycle checkpoint prior to commitment to DNA synthesis. The WT gene on chromosome 11p13 encodes a zinc finger motif-containing transcription factor found in CML patients only after progression to blast crisis.879 Overexpression of the EVI-1 gene has also been found in CML blast crisis.880,881 Microsatellite instability has not been found to be involved with progression to blast crisis.882BCL-2, c-MYC, and various other genes have also been implicated in the evolution of CML.883–886

Approximately 50 genes have been identified that could play a role in the progression to accelerated phase or blast crisis,847 including genes identified by expression profiling that are dysregulated in accelerated phase compared to chronic phase.887,888 These include the WNT–catenin and JunB pathways.

Clinical Features

Signs and Symptoms

The features that might signal the conversion of the chronic to the accelerated phase include unexplained fever, bone pain, weakness, night sweats, weight loss, loss of sense of well-being, arthralgia, and left upper quadrant pain related to splenic enlargement or infarcts. These features may occur weeks in advance of laboratory evidence of the accelerated phase. Localized or diffuse lymphadenopathy or enlarging masses in extralymphatic and extramedullary sites containing BCR-ABL–positive myeloblasts or lymphoblasts may develop. A poor response of blood cell counts and splenic enlargement despite previously effective therapy may be evident.843,854,889–891 Symptoms caused by histamine excess in basophilic crisis can be present.892

Several of these changes may occur in series or in parallel. The time of onset of transformation and the appearance of a blastic crisis and its clinical expression are unpredictable.

Laboratory Features

Blood Findings854,889–891

Anemia may worsen and be associated with increasing poikilocytosis, anisocytosis, and anisochromia. The number of nucleated red cells in the blood may increase. These red cell changes may be accentuated further if advancing marrow fibrosis is a feature of the disease.

The total leukocyte count may fall without treatment. The proportion of blasts increases to greater than 10 percent in blood and marrow in the accelerated phase and when blast crisis ensues represents 20 to 90 percent of the cells. The morphology of the blast cells may be lymphoid or myeloid. Myelocytes decrease in number. Hyposegmented neutrophils (Pelger-Huët cells) and other dysmorphic changes may become evident. Basophils increase and often represent 20 to 80 percent of the total blood leukocytes. A decrease of the platelet count to less than 100,000/L (<100 x 109/L) develops. Giant platelets, micromegakaryocytes, and megakaryocyte fragments may enter the blood. Decreased progenitor cell growth in culture is present, akin to that in acute leukemia.

Marrow Findings854,889–891

The marrow findings are widely variable. Marked dysmorphic changes in one, two, or three of the major cell lineages; an increase in blast count to greater than 10 percent; marrow morphology simulating subacute myelomonocytic leukemia; or, in the extreme, florid blastic transformation with blast counts greater than 30 percent can occur. Reticulin fibers may increase in prominence, and occasionally severe reticulin and collagen fibrosis develop. Additional clonal cytogenetic abnormalities develop in as many as half the patients in accelerated phase (see “Cytogenetic Studies” below).

Extramedullary Blast Crisis

A variety of symptoms or signs may occur as a result of the specific effects of new extramedullary blastic tumors, referred to as extramedullary blast crisis.893–896 Extramedullary blast crisis is the first manifestation of accelerated phase in approximately 10 percent of patients with CML. Lymph nodes,894,895 serosal surfaces,897,898 skin and soft tissue,893–896 breast,896,899 gastrointestinal or genitourinary tract,894,896 bone,894,896,900–903 and central nervous system894,904–906 are among the principal areas involved. Isolated or diffuse lymphadenopathy may occur. Bone involvement may lead to severe pain, tenderness, and pathologic fracture, and may be evident on imaging of the involved area. Central nervous system involvement usually is meningeal and may be preceded by headache, vomiting, stupor, cranial nerve palsies, and papilledema and is associated with an increase in cells, protein, and the presence of blasts in the spinal fluid.896,904–906

Appropriate histochemical and immunologic tests are required to determine if the extramedullary disease is composed of phenotypic myeloblasts or lymphoblasts. Because the tumor cells may have features of lymphoma cells, the terms myeloid or granulocytic sarcoma, chloroma, and myeloblastoma can be misnomers, and the term extramedullary blast crisis is used for this circumstance in CML.905,907–909 The lymphoblasts, like the myeloblasts, are Ph chromosome-positive. A combination of morphology, histochemistry (e.g., peroxidase, lysozyme), terminal deoxynucleotidyl transferase assay, and monoclonal antibodies specific for lymphoid or myeloid cells can be used to classify the extramedullary blast cells. Older reports (1930s–1960s) of concurrent lymphoma and CML probably were, in many cases, examples of extramedullary lymphoblast crisis in lymph nodes or other sites.

Marrow Blast Crisis

Approximately half of patients with CML enter the accelerated phase by developing acute leukemia. The onset of blast crisis can develop from days910–912 to decades after diagnosis of CML. The signs and symptoms may include fever, hemorrhage, bone pain, and lymphadenopathy.848,910–912 The morphology of the acute leukemia usually is myeloblastic or myelomonocytic.848,913 A substantial proportion of myeloid leukemia in this setting may not have myeloperoxidase demonstrable by cytochemistry.914 The proportion of cases classified as erythroblastic leukemia is approximately 10 percent, based on morphologic features,915 but may be as high as 20 percent if expression of glycophorin-A is used as the determinant.916 Occasional cases have megakaryoblastic transformation.875,917 These cases may be difficult to identify by light microscopy because the megakaryoblasts may be mistaken for lymphoid cells or undifferentiated blasts. Myelofibrosis is a feature of this variant. Antiplatelet glycoprotein antibodies and other monoclonal antiplatelet antibodies now are available as reagents to identify megakaryoblasts without the need for ultrastructural studies.917 Promyelocytic918–920 and eosinophilic921 blast crises also can occur. Basophilic leukemia is a known variant of CML.922 Patients with promyelocytic crisis often have t(15;17) in addition to the Ph chromosome, and some have presented with disseminated intravascular coagulation.923

CML may transform into acute lymphoblastic leukemia in approximately 30 percent of CML patients in blastic crisis.843,924–928 The lymphoid cells generally express terminal deoxynucleotidyl transferase (TdT)924,925 and are of the B cell lineage,928–930 as judged by antiimmunoglobulin staining. TdT is a DNA polymerase that adds deoxynucleoside monophosphates from triphosphate substrates to single-stranded DNA by end addition, differing in the latter respect from replicative polymerases.931 The enzyme is present in normal immature thymocytes and in the blast cells of nearly all patients with acute lymphoblastic leukemia. Rare patients have blasts with a T-lymphocyte phenotype.907,908,932,934 Some cases are biphenotypic; the blasts have both lymphoid and myeloid markers.913,935–937 Some cases may have myeloperoxidase activity in blast cells and express CD33 or CD13. Myeloid to lymphoid clonal succession following autologous transplantation in the second chronic phase has been described.938 Patients with lymphoid blast crisis seldom have an intermediate accelerated phase, have less splenomegaly and basophilia, and usually have a higher degree of marrow blast infiltration. With non–tyrosine-kinase-inhibitor therapy, remission rate and survival were somewhat longer in cases of lymphoid than in myeloid blast crisis.939

Cytogenetic Studies

Most large studies have shown seven recurrent changes in patients’ cells prior to, or during, the accelerated phase: trisomy 8 (33% of cases), additional 22q– (30% of cases), isochromosome 17 (20% of cases), trisomy 19 (12% of cases), loss of Y chromosome (8% of males), trisomy 21 (7% of cases), and monosomy 7 (5% of cases).940–943 In addition, a large number of other chromosome abnormalities have been described.944–948 In one study, 46 (63%) of 73 blast crisis patients had secondary cytogenetic abnormalities. These abnormalities were more common in myeloid blast crisis and were associated with shorter remission.860 The changes may be features of myeloid blast crisis compared to lymphoid crisis.941,946 Some abnormalities, such as inv16, are associated with early transformation to AML.942,946,949 A significant proportion (50%) of patients in the accelerated phase or blast crisis have no additional cytogenetic abnormalities beyond t(9;22)(q34;q11) after banding and multicolor FISH analysis.948 In cases where the blastic transformation is in extramedullary sites, such as lymph nodes or spleen, the additional cytogenetic abnormalities may be in the cells at those sites but not in cells in the blood or marrow.950

Treatment

Optimal treatment is allogeneic stem cell transplantation if the patient is eligible because of the patient’s age and donor availability. The role of stem cell transplantation is also evolving as tyrosine kinase inhibitor use in these phases of diseases becomes better defined.

Tyrosine Kinase Inhibitors in Accelerated and Blast Crisis

The initial dose of imatinib in accelerated phase is 600 mg/day.951 Imatinib, dasatinib, and nilotinib have been utilized as bridging therapies to permit allogeneic stem cell transplantation in accelerated phase.952 Dasatinib and nilotinib can achieve a better molecular response, and thus the role for and timing of transplantation in accelerated phase CML is being redefined. Combination therapies are being explored in the accelerated phase of disease, including dasatinib plus imatinib.953 Imatinib can be combined with mitoxantrone plus etoposide or cytarabine for patients in myeloid blast crisis.954 Imatinib has produced complete hematologic remissions in approximately 20 percent of patients.955,956 However, complete cytogenetic responses are uncommon. Central nervous system and other extramedullary blast crisis can occur during imatinib therapy for accelerated phase disease,957,958 and all types of blast crisis, including promyelocytic blast crisis,959 can occur during imatinib therapy. Compared to historical controls in which various combinations of chemotherapy were utilized, imatinib used alone results in comparable outcomes (6-month median survival of patients in blast crisis).960 Although dasatinib therapy can result in complete cytogenetic responses in 29 percent of blast crisis CML, and nilotinib can result in complete cytogenetic response in 27 percent of myeloid blast crisis and in 43 percent of lymphoid blast crisis, these responses are rarely durable, so in a patient of appropriate age with an acceptable donor, transplantation options should be considered with the second-line tyrosine kinase inhibitors utilized as a bridge to transplantation therapy.961,962

Chemotherapy

The treatment approach is predicated on the phenotype of the blast cells in CML patients with blast crisis and is rarely used in accelerated phase. In patients with myeloid phenotypes, the approach has been similar to that used for acute myelogenous leukemia: combinations of an anthracycline antibiotic, such as idarubicin or daunorubicin, with high-dose cytosine arabinoside and sometimes etoposide.963 Because this approach produces few remissions that are of short duration (median survival approximately 6 months), a variety of other drug combinations incorporating 5-azacytidine, busulfan, cladribine, fludarabine, clofarabine, high-dose cytosine, arabinoside, decitabine, etoposide, farnesyl transferase inhibitors, hydroxyurea, methotrexate, mitoxantrone, plicamycin, tiazofurin, or troxacitabine have been used, but with no significant improvement in outcome.960

In patients with lymphoid phenotypes, vincristine sulfate 1.4 mg/m2 (not to exceed 2 mg/dose) given intravenously once per week and prednisone 60 mg/m2 per day given orally are the mainstay of treatment. A minimum of two cycles of treatment (2 weeks) should be given to judge responsivity. Approximately one-third of patients with lymphoid blast transformation reenter the chronic phase after such treatment. However, because only about one-third of patients have lymphoid blasts, this number represents a remission rate of only approximately 10 percent of patients who enter blast crisis using this approach. Some relapsed patients become TdT-negative (myeloblastic relapse). However, even if relapsed patients remain TdT-positive, they are not likely to respond to a second treatment. Some therapists argue for a more intensive induction regimen for patients with lymphoblastic crisis, akin to regimens for de novo adult ALL or high-risk childhood ALL, and report somewhat better results: higher remission rates and longer remissions. The benefit of such an intensive approach has been small because remission durations have been modest. TdT-positive, CD10 (CALLA)-positive lymphoblasts may be the lymphoblast phenotype most responsive to vincristine and prednisone.927 mTOR inhibitors may be useful in lymphoid blast crisis.964

Allogeneic Stem Cell Transplantation Accelerated Phase and Blast Crisis of CML

Stem cell transplantation from an identical twin or sibling has been used in some patients after entry into the blastic phase. Occasional patients have had long-term survival. The 3-year survival rate is approximately 15 to 20 percent,965–967 unlike transplantation in the chronic phase, in which the 3-year survival rate is 50 to 60 percent. However, for patients who present in blast crisis, who develop blast crisis in the first year of the chronic phase, or who delay transplantation for other reasons, transplantation remains the best hope for long-term survival if a histocompatible donor is available.904,905 Relapse of accelerated phase after allogeneic stem cell transplantation has responded to infusion of donor cytotoxic T lymphocytes.968 The utilization of agents such as imatinib before allogeneic stem cell transplantation for patients in advanced phases of disease may favorably improve transplantation outcomes, especially when major cytogenetic response occurs before transplantation.969

Autologous Stem Cell Transplantation Accelerated Phase and Blast Crisis of CML

Autografting in the accelerated phase or blast crisis, either with stem cells collected during chronic phase or with mobilized Ph-chromosome–negative progenitor cells collected upon cell rebound after intensive chemotherapy, has resulted in apparent prolonged remission in some patients, but this procedure is rarely used in advanced disease because of the high rate of relapse.970 Whether Ph-chromosome–negative cells collected during imatinib therapy have the same potential is unknown.

Splenectomy

Splenectomy may be performed for palliation of painful splenic infarctions or hemorrhage. However, the complication rates are high, and the procedure performed in this setting should be avoided if possible.971

Course and Prognosis

The accelerated phase of CML generally is very poorly responsive or refractory to treatment and is a morbid state that can be fatal in weeks to months in all but a few patients who undergo a successful stem cell transplant from a histocompatible donor. Patients with myeloid blast crisis have a median survival of approximately 6 months, whereas patients with lymphoid blast crisis have a median survival of approximately 12 months.960–972 In the earliest study of imatinib mesylate in blast crisis, patients with myeloblastic crisis had a slightly longer median survival (about 5 months) than patients with lymphoid blast crisis plus Ph-chromosome–positive ALL (3 months). The results are poor in either case. The median survival after evidence of clonal evolution in patients in the chronic phase is approximately 15 months. A worse survival was seen with abnormalities of chromosome 17, other superimposed translocations, or a high percentage of abnormal metaphases.973 Severe cytopenias from repeated courses of cytotoxic therapy contribute to infections, hemorrhage, and organ dysfunction, especially liver and kidney dysfunction. Opportunistic infections with herpes viruses, cytomegalovirus, or fungi often supervene. The addition of imatinib mesylate therapy has made only a small difference in long-term outcome, although formal studies of combinations of chemotherapy and imatinib mesylate at a higher dose (600 mg/day) have not been completed, and the results of trials with second-generation tyrosine kinase inhibitors are anticipated.

Related Clonal Myeloid Diseases Without the BCR Rearrangement

Chronic Myelomonocytic Leukemia

This leukemia is part of the spectrum of clonal myeloid diseases that may have findings that simulate CML. In the past, when rigorous criteria for the diagnosis of CML were not applied, CMML was among a heterogenous group of related diseases that sometimes were referred to as Ph-chromosome–negative CML. These diseases share the feature of originating in the clonal expansion of a primitive multipotential hematopoietic cell.974 (Table 90–6)

Table 90–6. Types of Chronic Myelogenous Leukemia
Type of Chronic Myelogenous Leukemia Molecular Genetics Major Clinical Features Further Details
BCR rearrangement-positive chronic myelogenous leukemia >95% p210BCR-ABL; <5% p190 or p230 Splenomegaly in 80% of cases; WBC >25,000/L; blood blasts <5%; Ph chromosome in 90% of cases; BCR gene rearrangement in 100% of cases Page 1338
Chronic myelomonocytic leukemia Various cytogenetic abnormalities Anemia, monocytosis >1000/L; blood blasts <10%; increased plasma and urine lysozyme; BCR rearrangement absent; rare cases with PDGFR- mutation respond to imatinib Page 1356
Chronic eosinophilic leukemia Various cytogenetic abnormalities Blood eosinophil count >1500/L; cardiac and neurologic manifestations common; a proportion of cases have PDGFR– mutations and are responsive to imatinib mesylate Page 1358
Chronic basophilic leukemia Various cytogenetic abnormalities Only 5 cases reported; hemoglobin 6–13 g/dL; basophilia of 3.4–41 x 109/L; 2 of 5 cases with splenomegaly; very cellular marrow (>90%) in each case with mild increase in type III collagen, and megakaryocytic dysmorphia; increase in marrow mast cells in 3 of 5 cases Page 1359
Chronic monocytic leukemia Various cytogenetic changes Proportion of monocytes elevated; very rare form of leukemia Page 1359
Juvenile myelomonocytic leukemia Various cytogenetic changes Infants and children <4 years; eczematoid or maculopapular rash; anemia and thrombocytopenia; increased Hgb F in 70% of cases; neurofibromatosis in 10% of cases; abnormality of chromosome 7 (e.g., del 7, del 7q, etc.) in approximately 20% of patients; BCR rearrangement absent Page 1360
Chronic neutrophilic leukemia Various cytogenetic changes Segmented neutrophilia >20,000/L; splenomegaly >90% of cases; no blood blasts; platelets >100,000/L; 75% of cases have normal cytogenetics; BCR rearrangement absent Page 1361
BCR rearrangement-negative chronic myelogenous leukemia Various cytogenetic changes Clinical findings indistinguishable from BCR rearrangement-positive CML; Ph chromosome and BCR-ABL fusion gene absent Page 1362

Epidemiology

Most patients with CMML are older than age 50 years, and approximately 75 percent of patients are older than age 60 years at the time of diagnosis. The median age at diagnosis is approximately 70 years. Occasional cases have been reported in older children and younger adults. Men are affected somewhat more frequently than women (approximately 1.4:1).975,976

Clinical Findings

Signs and Symptoms

The onset usually is insidious, and weakness, infection, or exaggerated bleeding may bring patients to medical attention.975,976 Hepatomegaly and splenomegaly occur in approximately 50 percent of patients. Leukemia cutis occurs in a small proportion of patients and usually has a monocytic phenotype: CD45, CD68, and lysozyme positive by immunostaining.977 Immune manifestations, such as vasculitis, pyoderma gangrenosum, immune cytopenias, and connective tissue diseases, may occur in coincidence with CMML.978

Blood and Marrow Findings

The disease is characterized by anemia and blood monocytosis greater than 1000/L (1 x 109/L).979 The white cell count may be slightly decreased, normal, or moderately elevated. Occasional patients may have hyperleukocytosis with total white cell counts of 250,000 to 300,000/L (250–300 x 109/L) associated with respiratory insufficiency resulting from pulmonary leukostasis.980 Immature granulocytes may be present in the blood. Blood myeloblasts may be absent or, when present, do not exceed 10 percent of total white cells. Most patients have thrombocytopenia, but normal or elevated platelet counts may occur. Eosinophilia may be so prominent in occasional cases that the designation chronic eosinophilic leukemia may be appropriate.975,976,981

The marrow is hypercellular as a result of granulomonocytic hyperplasia; the dominant cells are early myelocytes. The proportion of progranulocytes is increased. Promonocytes also are increased in number. Distinction between poorly granulated myelocytes and promonocytes with primary granules can be difficult. Macronormoblasts and hypersegmented or hyposegmented, often bilobed (acquired Pelger-Huët anomaly), neutrophils are frequent. Despite thrombocytopenia, megakaryocytes usually are present in the marrow. Microvessel density is increased in the marrow, and myelomonocytic cells contain cytoplasmic mRNA for VEGF and membrane VEGF receptors.981,982 In vitro colony studies suggest that autocrine stimulation of cell growth by VEGF may occur. “Spontaneous” cluster/colony growth of granulocyte-monocyte colony-forming cells occurs in vitro. The spontaneous growth may result from autocrine or paracrine production of GM-CSF, based on anti–GM-CSF inhibition of colony growth.984

Cytogenetic Findings

Patients with CMML have an approximately 35 percent frequency of chromosomal abnormalities. Monosomy 7 and trisomy 8 are the most prevalent findings. Approximately 35 percent of patients have point mutations of the K-RAS or N-RAS gene. The RAS gene also may be involved in the transforming events. Abnormal methylation of p15INK4B is a common finding in CMML.985 Translocation between the gene PDGFR- on chromosome 5(q33) and four partner genes—TEL at 12(p13), HIP-1 at 7(q11.2), H4 at 10(q22), and Rabaptin-5 at 17(p13)—occur in a very small proportion of patients (approximately 3–4%).975,986–989 This mutation juxtaposes the gene encoding the PDGFR- with a partner gene, which results in the encoding of a mutant tyrosine kinase that is constitutively activated and sensitive to inhibition by imatinib mesylate990 (see “Treatment” below). The cases with PDGFR- translocations are more likely to be accompanied by eosinophilia than are cases with other cytogenetic abnormalities.

Serum and Urine Findings

Plasma and urine lysozyme concentrations nearly always are elevated. Plasma levels of VEGF, hepatocyte growth factor, and tumor necrosis factor alpha are elevated. Serum B12, 2-microglobulin, and LDH levels often are elevated.975,976

Treatment Chronic Myelomonocytic Leukemia

Treatment of most patients with CMML has been unsatisfactory, and remissions of any duration are uncommon. The age and performance status of the patient are considered in determining the intensity of treatment. Cytarabine, either standard or low-dose, etoposide, hydroxyurea, and other approaches used for the oligoblastic myelogenous leukemias have been attempted, but with little success (see Chap. 88). Decitabine and 5-azacytidine have been useful in a small proportion of patients.975,976 Unfortunately, although a particular approach may confer significant benefit in a small proportion of cases, determining which patients will respond is not possible, except by trial and error. An exception is the patient with a translocation involving PDGFR-, which itself occurs in only a small percentage of patients. In the case of PDGFR- fusion genes with several of the partner genes, imatinib mesylate 400 mg/day has resulted in normal blood counts, cytogenetic remissions, and, occasionally, molecular remission.985–987,991,992 These fortunate patients probably will benefit from this treatment, as evidenced by prolonged remissions and survival, compared to other drug options, but the number of patients and duration of followup do not permit quantitative estimates at this point. Allogeneic stem cell transplantation is an option for the small proportion of younger patients with an appropriate matched-related or unrelated donor.993

Course and Prognosis

Median survival in CMML is approximately 12 months, with a range from approximately 1 to more than 60 months. Approximately 20 percent of patients progress to frank AML. Arbitrary stratification of CMML into types 1 and 2 based on the height of the blast count has been proposed, but distinguishing patients by whether they have 8 or 12 percent blasts on a marrow examination is useless for patient care. It is well established that blast percentage usually is a significant correlate with outcome in any patient with a clonal myeloid disease, and this factor among several others should guide therapy. Clusters of prognostic variables have been used to stratify patients into risk groups for survival duration. In general, the severity of the anemia and the height of the blast percentage are the most important. Other variables that may confer a shorter life expectancy are height of the absolute lymphocyte count, high spontaneous rates of myelomonocytic colony growth, higher total leukocyte counts, higher LDH level, and larger spleen size.994–996 Unfortunately, at this time, unless the patient is a candidate for imatinib therapy or stem cell transplantation, long-term salutary therapeutic effects are uncommon.

Chronic Eosinophilic Leukemia

History and Definition

The recognition of eosinophilic lineage prominence in myelogenous leukemia dates to a case published in 1912.997 In 1968, the term hypereosinophilic syndrome was introduced to encompass a group of disorders with (1) prolonged exaggerated eosinophilia without an apparent cause, (2) frequent cardiac and neurologic tissue damage, (3) a poor or transient response to therapy, and (4) a progressive course and a high fatality rate. Shortly thereafter, Benvenisti and Ultmann998 presented five cases of eosinophilic leukemia and reviewed the literature regarding that phenotypic designation. In 1975, Chusid and colleagues999 described 14 cases of hypereosinophilic syndrome, highlighted the frequency of secondary cardiac and neurologic disorders, and suggested the existence of a continuum of manifestations. Because some cases had clonal cytogenetic abnormalities and hematologic findings compatible with a clonal myeloid disease, the presence of eosinophilic leukemia was suspected in this apparently heterogeneous group of patients.

The relationship of blood eosinophilia to clonal myeloid diseases is complex because the former can be reactive or represent acute eosinophilic leukemia, chronic eosinophilic leukemia, or eosinophilia associated with a different category of disease, such as BCR-ABL–positive CML, idiopathic myelofibrosis, oligoblastic leukemia (MDS), or mastocytosis.1000 Chronic eosinophilic leukemia is a BCR-ABL–negative, clonal myeloid disease with a striking eosinophilia in the blood and marrow, often with clonal cytogenetic abnormalities that have features including, when present, cytogenetic findings that usually distinguish chronic eosinophilic leukemia from other clonal myeloid diseases that may have an associated eosinophilia, such as CMML. The phenotype of the eosinophilic variant of CMML overlaps somewhat with that of chronic eosinophilic leukemia. However, the fusion gene associated with CMML involves the PDGFR- (see “Chronic Myelomonocytic Leukemia” above), whereas in chronic eosinophilic leukemia the cytogenetic findings are different and in some cases involve PDGFR-. This definition of chronic eosinophilic leukemia recognizes that, at the margins, classification may be arbitrary. Studies in cases with the FIP1L1-PDGFR- translocation were consistent with multilineage involvement, consistent with an origin in a pluripotential lymphohematopoietic cell.1001 Another report found multilineage involvement but with a presumptive lesion in a multipotential, not pluripotential, hematopoietic cell.1002

Signs and Symptoms

Fever, cough, weakness, easy fatigability, dyspnea, abdominal pain, maculopapular rash, cardiac symptoms and signs of heart failure, and a variety of neurologic manifestations ranging from peripheral neuropathy to cerebral encephalomalacia may occur, ranging from mild to severe in expression. Splenomegaly often is evident.

Laboratory Findings

Eosinophilia is a constant finding (see Chap. 62, Fig. 62–3). Anemia usually but not always is present at the time of presentation. The leukocyte count may be high-normal or more often elevated. Platelet counts often are normal or mildly decreased. The marrow shows myelocytic and eosinophilic hyperplasia and occasionally Charcot-Leyden crystals. Mast cells may be increased. Megakaryocytes usually are present but may appear dysmorphic. Reticulin fibrosis is common. Immunophenotyping and PCR does not show evidence of either a clonal T-cell population or T-cell-receptor rearrangement. Pulmonary function studies may provide evidence of fibrotic (restrictive) lung disease. Echocardiography may detect mural thrombi, thickening (fibrosis) of the ventricular wall, valvular dysfunction from papillary muscle, and chordae fibrosis. Magnetic resonance imaging can detect subendocardial fibrosis, thickening of ventricles, and markedly reduced ventricular lumen volume. Serum immunoglobulin (Ig) E, vitamin B12, and tryptase levels usually are elevated. Skin biopsy of lesions uncovers intense eosinophilic infiltrates. Neural or brain biopsy may disclose eosinophilic infiltrates, often perivascular, with microthrombi, axonal degeneration, and gliosis.

Cytogenetic Findings

A wide array of cytogenetic findings have been reported in cases of chronic eosinophilic leukemia.1003 Notable translocations include a high frequency of translocations involving chromosome 5, t(1;5), t(2;5), t(5;12), t(6;11), 8p11, trisomy 8, and numerous others infrequently. Chromosome 5 often is translocated at the site of the PDGFR- gene, and the phenotype usually is more compatible with CMML with eosinophilia. Chromosome 5 from band q31-35 contains several genes relevant to eosinophilopoiesis, including those encoding IL-5, IL-3, GM-CSF, and PDGFR-. A cryptic interstitial deletion on chromosome 4(q12;q12) results in the fusion gene FIL1L1-PDGFR- and in a phenotype of chronic eosinophilic leukemia, which is of particular note because, like the PDGFR- mutations in CMML with eosinophilia, of a near-universal response to treatment with imatinib mesylate.1003–1005

Serum Tryptase Level Elevation versus Normal Levels

The elevation of serum tryptase level (>11.5 ng/mL) has been used to distinguish a subset of patients who (1) are male, (2) have marrows that are intensely hypercellular with a higher proportion of immature eosinophils and with dysmorphic mast cells with a CD117–CD25+CD2– genotype and phenotype (distinguishing these cells from classic mastocytosis, which are CD117+CD25+CD2+), (3) have dramatically higher serum B12 and IgE levels, (4) are more prone to restrictive pulmonary disease and endomyocardial fibrosis, (5) have the FIP1L1-PDGFR- fusion gene, and (6) are responsive to imatinib mesylate.941 Patients with normal serum tryptase levels are more prone to obstructive pulmonary restrictive disease, eosinophilic dermatitis, and gastrointestinal complaints.

Differential Diagnosis

Eosinophilia can occur for many reasons (see Chap. 62). The first step is to identify signs that may point to a clonal myeloid disease. These signs include anemia, thrombocytopenia, splenomegaly, immature eosinophils in the marrow examination, evidence of dysmorphic cells in blood or marrow, for example, atypical megakaryocytes or dysmorphic mast cells, cardiac or pulmonary manifestations, which may occur secondary to chronic eosinophilic leukemia, and markedly elevated serum tryptase or vitamin B12 level. The former signs, especially in the aggregate, are highly suggestive, but the presence of a cytogenetic abnormality in myeloid cells is diagnostic of a clonal myeloid disease (leukemia). If the latter is not evident, PCR and/or flow cytometry to search for a clonal T lymphocyte abnormality should be performed. Whether the eosinophilic leukemia is typical or represents an eosinophilia with idiopathic myelofibrosis, CMML, or MDS is less important than if it has a mutation that is imatinib mesylate sensitive (e.g., PDGFR mutation).

Therapy

Patients (nearly always men) whose cells display a FIP1L1–PDGFR- have a very high probability of responding to imatinib mesylate at a dose of 100 to 400 mg/day.1004–1008 The tyrosine kinase activity of this fusion protein is two orders of magnitude more sensitive to imatinib than that of BCR-ABL. However, because not all patients taking 400 mg/day achieve a molecular remission, and that goal may be more likely to result in long-term remission, initial therapy remains at 400 mg/day and PCR monitoring is appropriate. Dose adjustment upward if molecular remission is not achieved can be considered. Unlike the case in CML, patients with chronic eosinophilic leukemia with significant side effects when taking imatinib mesylate, 400 mg/day, have a reasonable probability of having a good response at lower doses.1008 Responses to dasatinib and nilotinib have also been reported.1009

In patients with eosinophilic leukemia without an imatinib mesylate-sensitive mutation or in patients who become resistant to imatinib mesylate and unresponsive to second-generation tyrosine kinase inhibitor (e.g., dasatinib) and who are progressing, ablative or nonablative allogeneic stem cell transplantation can be considered if they are in an acceptable age range and have access to a matched-related or matched-unrelated donor.1010,1011

In imatinib mesylate-insensitive patients without the option of transplantation, empirical treatment with glucocorticoids, hydroxyurea, or anti-IL51012,1013 to decrease eosinophil counts and mute the progress of eosinophil-mediated cutaneous, cardiac, pulmonary, and neurologic tissue damage should be considered. These approaches may relieve symptoms for a time, but are temporizing if therapy with drugs that might be effective in inhibiting clonal expansion or evolution is not effective (e.g., cytarabine, anthracycline antibiotic, etoposide).

Course and Prognosis

If chronic eosinophilic leukemia is not imatinib sensitive, the long-term outlook is one of probable progressive cardiac and neurologic disability. Transformation to acute eosinophilic or myelogenous leukemia can occur. Allogeneic stem cell transplantation is potentially curative. In imatinib-sensitive cases, hematologic normalization, reversal of marrow fibrosis and mastocytosis, resolution of skin lesions, normalization of spleen size, and restoration of well-being occurs in the great preponderance of cases. Cardiac, neurologic, and pulmonary changes usually cannot be reversed but should be stabilized. The long-term outlook with imatinib mesylate therapy is uncertain. However, imatinib likely will decisively and dramatically improve survival compared to other prior therapy and should greatly improve the prognosis of patients with a molecular target for the drug.

Chronic Basophilic Leukemia

This type of clinical disorder, in which the patient has marrow and blood basophilia and other findings compatible with a clonal myeloid disease without evidence of the BCR-ABL translocation, is rare. Two reports of such a syndrome occurring in five patients have been published.1014,1015 The marrow was intensely hypercellular in the three major lineages. Dysmorphic megakaryocytes were evident. Basophilia in marrow and blood was striking, although eosinophilia also was evident in two patients and increased mast cells in three patients. The clinical effects of basophilic mediator release were evident in two patients. One patient evolved to AML; another recovered after allogeneic transplantation. The cases had similar findings, leading to the suggestion they represented Ph-negative chronic basophilic leukemia. In one case, a PRKG2-PDGFR- fusion gene was evident and the patient responded to imatinib mesylate.

Chronic Monocytic Leukemia

History

In 1937, Osgood1016 reviewed his experience with monocytic leukemia and included a case that probably represented the rare disorder chronic monocytic leukemia. In 1981, approximately 28 bona fide cases had been reported, 5 cases were added, and the characteristics were reviewed.1017

Clinical Findings

Patients range in age from 30 to 80 years. Males are affected more frequently than females. Fever, fatigue, and left upper quadrant pain are the most common complaints. Splenomegaly and hepatomegaly are nearly constant findings.1017–1019

Laboratory Findings

Anemia is mild. Anisocytosis and poikilocytosis usually are present. The leukocyte count usually is normal or low but can be elevated in a minority of patients. The percentage of monocytes is increased, but the absolute monocyte count often is normal, ranging from 300 to 1500/L (0.3–1.5 x 109/L), or mildly elevated. Occasional patients have more striking monocyte counts. The platelet count may be normal or decreased. Rare nucleated red cells may be present in the blood. The monocytes in the blood contain -naphthol acetate esterase, tartrate-sensitive acid phosphatase, fluoride-sensitive naphthol AS-D acetate esterase, and peroxidase as judged by histochemical tests. The marrow is cellular, often without an increase in monocytes. The Ph chromosome is absent. The leukemic cells are similar to mature monocytes with abundant cytoplasm. Erythrophagocytosis or thrombocytophagocytosis by monocytes may be seen.

The disease often is not recognized because the total white cell count, monocyte count, and number of marrow monocytes may not be elevated until the spleen is removed, usually for diagnostic purposes.1017 Following splenectomy, a gradual leukocytosis of 3000 to 100,000/L (3–100 x 109/L) may develop.1017 The absolute monocyte count increases dramatically, often from less than 1000/L (1 x 109/L) to as high as 75,000/L (75 x 109/L). The marrow may contain more than 50 percent mature monocytes following splenectomy.

The spleen is enlarged (300–2500 g). The red pulp is infiltrated with mononuclear cells, often obliterating sinus lumens. Erythrophagocytosis by the mononuclear cells frequently is evident. Liver biopsy may show a mononuclear infiltrate in the sinusoids. Although clinical lymph node enlargement is rare, lymph node biopsies show striking infiltration by leukemic monocytes.

Course, Prognosis, and Treatment

Median survival is approximately 25 months. Patients often die of septicemia1017–1019 or acute monocytic leukemia.1020,1021 Therapy has not been studied systematically, but neither intensive combination chemotherapy nor glucocorticoids have changed the course of the disease.

The World Health Organization Committee on the Lymphohematopoietic Tumors does not describe a disease called chronic monocytic leukemia and recent reports of cases are very rare. A few well-documented cases of chronic monocytosis, presumably clonal, evolving into myelogenous leukemia could also satisfy this designation (see Chap. 88).

Juvenile Myelomonocytic Leukemia

Epidemiology

Ph-chromosome–positive, adult-type CML occurring in children younger than age 15 years composes approximately 3 percent of childhood leukemias and approximately 10 percent of all cases of CML.1022 Although CML occurs in children of all ages, it is rare in children younger than age 5 years. With the exception of a propensity to present with higher total leukocyte counts and with leukostatic signs or symptoms, CML in children has the typical manifestations and course of the disorder seen in adults.

A disorder different from adult-type CML, designated juvenile myelomonocytic leukemia (juvenile CML), represents approximately 1.5 percent of childhood leukemias. It occurs most often in infants and children younger than age 4 years and is similar in some respects to adult subacute or chronic myelomonocytic leukemia because the two diseases share a prominent monocytic component in the leukemic cell population.1023–1026

Pathogenesis

This disorder is a clonal myeloid disease that originates in an early hematopoietic multipotential cell. Evidence indicates this cell may be pluripotential (myeloid-lymphoid) in some cases and myeloid in others.1027–1030 RAS mutations in hematopoietic cells are present in approximately 20 percent of patients.1031 Approximately 1 in 10 patients with juvenile myelomonocytic leukemia have mutations of NF1 and manifest type 1 neurofibromatosis. This frequency is approximately 400 times the expected occurrence in a comparable pediatric population.1032–1034 The linkage between neurofibromin, the protein encoded by the NF1 gene, guanosine triphosphatase activity proteins, and the activation state of RAS-encoded proteins has led to a postulated sequence of events that may be triggered by the extraordinarily heightened sensitivity of the colony-forming cells in the marrow and blood of infants with the disease to the proliferative effects of GM-CSF. The latter initiates signal transduction from the cell membrane to the nucleus via RAS protein activation.1035,1036 Mutations in the PTPN11 gene have been found in approximately one-third of children with juvenile CML, and the mutations in NF1, RAS, and PTPN11 usually do not coincide.1035,1036 However, they each may act through a common pathway. PTPN11 encodes SHP-2, a nonreceptor tyrosine kinase, which is an upstream regulator of RAS; thus, all three mutations can contribute to deregulation of RAS signaling. As an aside, children with Noonan syndrome, which is characterized by short stature, dysmorphic facies, skeletal abnormalities, and cardiac defects, have a germ-cell mutation of PTPN1. These children may have a transient disorder that closely mimics juvenile myelomonocytic leukemia.1036

Clinical Findings

Symptoms and Signs

Infants present with failure to thrive, and children present with malaise, fever, persistent infections, and exaggerated skin, oral, or nasal bleeding. Hepatomegaly can occur. Splenomegaly, sometimes massive, is present in almost all cases. Lymphadenopathy is frequent.1023–1026 More than half of the patients have eczematoid or maculopapular skin lesions1037 and xanthomatous lesions, and multiple café-au-lait spots (neurofibromatosis type 1) may occur.1024 The xanthomas may be the earliest signs of neurofibromatosis.1024,1025 Noonan syndrome (dysmorphic facies, short stature, heart disease, mental retardation, cryptorchidism, webbed neck, chest deformities, and bleeding diathesis) may coexist.1025

Laboratory Findings

Anemia, thrombocytopenia, and mild to moderate leukocytosis are common. The leukocyte count usually is greater than 10,000/L (10 x 109/L) with a median leukocyte count at diagnosis of about 35,000/L (35 x 109/L). The blood has an increased monocyte concentration of 1000 to 100,000/L (1–100 x 109/L), immature granulocytes including a small percentage of blast cells, and nucleated red cells. Fetal hemoglobin concentration is increased in approximately two-thirds of the patients. The marrow aspirate is hypercellular as a result of granulocytic hyperplasia; the number of erythroblasts and megakaryocytes usually are decreased. Monocytic cells are increased but may not be as striking as in the blood. Leukemic blast cells are present in modest proportions (<20%).

Cell culture of blood and marrow shows a striking preponderance of monocytic progenitors, even in the absence of overt monocytosis in the marrow.1038,1039 Granulocyte-monocyte colony-forming cells show a marked tendency to spontaneous growth if adherent (monocytic) cells are not depleted from culture.1039 The effect is mediated by a release of large quantities of GM-CSF by monocytes in culture.1040

Although clonal chromosome abnormalities have been found in some cases,1041 the cytogenetic abnormalities have no consistent pattern, and more than half of the patients have normal karyotypes. The BCR-ABL fusion gene is not present.1041–1043 The phenotype of monosomy 7 syndrome overlaps with juvenile myelomonocytic leukemia, and an abnormality of chromosome 7 (del 7, del 7q, others) is present in approximately one-fifth of patients.1023

Course, Prognosis, and Treatment

The median survival of patients with juvenile CML has been less than 2 years.1023,1024 Younger children (younger than 2 years) are more likely to have a protracted course.955 The disease has been refractory to most chemotherapy. In a study of 9 patients, 4 of whom were treated with a 5- or 6-drug intensive regimen, remissions were 11 to more than 27 months, compared with untreated or lightly treated patients, 4 of whom died within 7 months.1044 Even in the treated patients, complete suppression of the disease did not occur, and treatment protocols to induce and sustain remissions were lacking.1039 A program of cytosine arabinoside, etoposide, vincristine, and isotretinoin resulted in a highly favorable response in five children treated. Three patients relapsed and were treated with cytarabine by infusion and subcutaneously and with etoposide. All patients were alive, and the range of survival at the time of publication was 8 to 89 months, with a median survival of 27 months. The resistance of these cells to currently available therapy is gruesomely highlighted by the sense of success in prolonging the life of infants and young children by a few years. Intensive therapy can control disease, but curative chemotherapy has been elusive.1045 The inclusion of isotretinoin was based on a prior report of responsiveness to the drug used alone; however, this observation has not been confirmed.1046 The GM-CSF antagonist E21R, inhibitors of RAF-1 gene expression, blockers of RAS protein farnesylation, and angiogenesis inhibitors are among other drug approaches to the disease being studied.1026,1047

Allogeneic stem cell transplantation is an important approach to therapy and may provide the best chance of long-term survival in selected children.1048,1049 Hence, a rapid search for a matched-unrelated donor is important in patients without matched sibling donors. Transplantation from a histocompatible sibling or matched-unrelated donor resulted in an event-free survival at 4 years of 54 percent in one study of 27 patients that used a variety of conditioning regimens. A cytogenetic abnormality, such as monosomy 7, was a dismal prognostic finding in that study, and children transplanted before age 1 year had better results than did older children.1048 DLI has placed a posttransplantation patient in relapse into remission,1050 but in general this procedure is ineffective in patients who relapse after transplantation.

A minority of patients have a smoldering course for 2 to 4 years. Thereafter, the disease usually rapidly progresses, and patients die of infection or hemorrhage. Occasional patients have a very long survival (>10 years) despite persistence of abnormal blood counts and splenomegaly, independent of the type or intensity of therapy. Some children convert to a full-blown acute myelogenous leukemia with a rapidly fatal outcome. Cases of juvenile myelomonocytic leukemia may be associated with transformation to acute lymphoblastic leukemia.1051

Chronic Neutrophilic Leukemia

History, Pathogenesis, and Epidemiology

In 1920, Tuohey1052 described the first recorded case of an unusual sustained neutrophilia with splenomegaly without fever, inflammation, cancer, or other cause of a leukemoid reaction. Use of X-chromosome-linked polymorphic genes in blood cells and FISH of chromosome abnormalities have been indicative of a clonal myeloid disorder.1053–1055 Some cases may arise in the hematopoietic multipotential cell, others in a neutrophil progenitor cell (see Chap. 85).1053–1057 Evidence points to defective apoptotic signals accounting, in part, for the striking accumulation of segmented neutrophils in the blood.1058 The median age at onset is approximately age 65 years. Younger patients may be affected.1059 As in most clonal myeloid diseases, men are affected more frequently than are women.

Clinical Features

Symptoms and Signs

Patients may complain of weakness, anorexia, weight loss, abdominal pain, and easy bruising. Symptoms and signs of gouty arthritis occur in approximately one-third of cases. The spleen is enlarged in almost all cases, and the liver frequently is enlarged. Lymphadenopathy is very infrequent.1057 A hemorrhagic tendency is present in some patients.

Laboratory Findings

Although some patients have a normal hemoglobin concentration at the time of presentation, most have mild to moderate anemia on presentation. The reticulocyte count usually is between 0.5 and 3.0 percent. The platelet count rarely is less than 125,000/L (125 x 109/L) and usually is normal. Coagulation times are normal. The total leukocyte count usually is between 25,000 and 75,000/L (25 and 75 x 109/L) in most cases, and only rarely is less than 20,000/L (20 x 109/L) or exceeds 100,000/L (100 x 109/L). Neutrophils compose 85 to 95 percent of the white cells. Although segmented cells usually dominate, occasional cases have a high proportion of band forms. Very infrequently, metamyelocytes, myelocytes, and nucleated red cells may be present in patients. Basophil and eosinophil counts are not increased. Blasts nearly always are absent from the blood. Neutrophil alkaline phosphatase activity is increased in almost all cases.

The marrow invariably shows granulocytic hyperplasia with myeloid-to-erythroid (M:E) ratios as high as 10:1. Myeloblasts are not overtly increased in number (0.5–3.0%). Megakaryocytes are either normal or slightly increased in number and have normal distribution and morphology. Erythropoiesis usually is mildly decreased. Unlike CML, reticulin fibrosis is unusual. A few cases with dysmorphic features in the marrow (acquired Pelger-Hüet anomaly, erythroid, dysplasia, micromegakaryocytes) have been reported. By definition, the Ph chromosome, BCR gene rearrangements, and BCR-ABL transcripts are absent.1059–1062 Most patients have normal karyotypes, but approximately 25 percent of patients have nonrandom abnormalities of chromosomes.1062 Deletions of chromosome 20q and trisomy 21 or 9 are the most common abnormalities. Serum vitamin B12-binding protein and vitamin B12 levels both are markedly increased above normal. Serum uric acid concentration is increased, and serum lactic dehydrogenase activity may be increased.

Almost every case examined postmortem had liver and splenic enlargement. Portal hepatic and splenic red pulp infiltrates of neutrophils or islands of extramedullary hematopoiesis with immature myeloid cells and megakaryocytes are characteristic.

Differential Diagnosis

Most leukemoid reactions are associated with an obvious underlying cause, such as pancreatitis, carcinoma, connective tissue disease, smoker’s neutrophilia, and chronic bacterial or fungal infection. The leukocyte alkaline phosphatase level usually is markedly elevated in chronic neutrophilic leukemia and markedly decreased in CML. More to the point, molecular studies identifying BCR gene rearrangement or the presence of BCR-ABL transcripts should distinguish chronic neutrophilic leukemia (BCR-ABL–negative) from neutrophilic CML (BCR-ABL–positive; see “Special Clinical Features” above). In the latter case, more than half of the patients have thrombocytosis and megakaryocytic hyperplasia, which are uncharacteristic of chronic neutrophilic leukemia.

Treatment

No systematic studies of treatment have been reported. Although hydroxyurea, IFN-, or cytarabine may decrease the white count and spleen size, long-term benefit is unusual.1059–1062 Intensive therapy has led to early posttreatment deaths. Allogeneic stem cell transplantation in eligible patients may be curative.1063

Course and Prognosis

The disease is fatal, with a median survival of approximately 2.5 years and a range of 0.5 to 6 years.1059–1062 A case of spontaneous remission has been reported. The prognosis is considerably worse than the prognosis for CML despite the prevalence of mature neutrophils and the paucity of blasts. Causes of death have included (1) intracranial hemorrhage, sometimes in the presence of adequate platelet counts and coagulation times, suggesting a vascular infiltrative process; (2) severe infection; (3) transformation to acute myelogenous leukemia; and (4) the toxic effects of intensive therapy. The disease usually afflicts older subjects, and cardiac, pulmonary, and vascular diseases contribute to a fatal outcome.

A remarkable frequency of concordant essential monoclonal gammopathy or myeloma has been described.1055,1064–1072 In two cases, the extreme neutrophilia proved to be a polyclonal response to a plasma cell disorder.1055,1073 Chronic neutrophilic leukemia has evolved from polycythemia vera or oligoblastic leukemia,1074–1078 supporting its relationship to the clonal hemopathies.1055,1079,1080

BCR Rearrangement-Negative Chronic Myelogenous Leukemia

A very small proportion of patients (~4%) with clinical manifestations within the limits usually applied to the diagnosis of CML have neither a Ph chromosome (classic, variant, or masked) nor evidence of rearrangement of BCR on chromosome 22. This circumstance represents BCR-negative CML. The literature describing Ph-chromosome–negative CML prior to 1987 is difficult to evaluate because many cases were not studied carefully for masked or variant translocations and for the BCR gene rearrangement. Ph-chromosome–negative CML is a clonal disease974 that has the propensity for lymphoid and myeloid transformation.1081,1082 Although most cases of BCR rearrangement-negative CML are closer in manifestations to CMML,996,974,1083–1086 some cases are indistinguishable from classic CML but without the BCR-ABL after exhaustive molecular diagnostic evaluation.1087–1091 In a report of 76 such patients, the median age was 66 years (range: 24–88), splenomegaly was present in 50 percent of cases, the median white cell count was 38,000 cells/L (38 x 109 cells/L; range: 11–296), and the median hemoglobin was 11 g/dL (range: 7 to 16), with classical morphologic features in blood and marrow.1091 As the disease progressed, patients developed severe cytopenias.1088 Median survival was 24 months and only 7 percent survived for more than 5 years. Myeloid blast crisis occurred in one-third of those followed until their death. Occasional patients had extended complete remissions with INF- therapy.1090 Hydroxyurea can be useful as palliative therapy.

Some patients have transposition of ABL to chromosome 22 but not the classic translocation. In such cases, including TEL-ABL translocations, transient responses to imatinib mesylate have been observed.1092

Uncommon cases of coexisting BCR– and BCR+ clones have been described, and the basis for such cases is in dispute.1093 One proposed explanation is that this case is an example of “field carcinogenesis” in which multiple clones coexist.1093 An alternative explanation is that these cases represent the dual progeny of a single unstable clone.1094 The long-term survival of patients with CML may permit the emergence of a drug-induced or spontaneous second malignancy, and in the former it may be notably associated with imatinib (see above “Secondary Chromosomal Changes with Imatinib Mesylate” under “Tyrosine Kinase Inhibitors”).1095–1097

References

1. Bennett JH: Case of hypertrophy of the spleen and liver, in which death took place from suppuration of the blood. Edinburgh Med Surg J 64:313, 1845.
2. Virchow R: Weisses blut. Froieps Notizen 36:151, 1845.
3. Craige D: Case of disease of the spleen in which death took place in consequence of the presence of purulent matter in the blood. Edinburgh Med Surg J 64:400, 1845.
4. Virchow R: Die Leukaemie in Gesammelte Abhandlungen zur Wissen-Schaftlichen Medizin. Meidinger, Frankfort, 1865.
5. Neumann E: Ueber myelogene leukämie. Berl Klin Wochenschr 15:69, 1878.
6. Nowell PC, Hungerford DA: A minute chromosome in human chronic granulocytic leukemia. J Natl Cancer Inst 25:85, 1960. [PMID: 14427847]
7. Baike AG, Court Brown WM, Buckton KE, et al: A possible specific chromosome abnormality in human chronic myeloid leukemia. Nature 188:1165, 1960.
8. Nowell PC, Hungerford DA: Chromosome studies in human leukemia: II. Chronic granulocytic leukemia. J Natl Cancer Inst 27:1013, 1961. [PMID: 14480645]
9. Tough IM, Court Brown WM, Buckton KE, et al: Cytogenetic studies in chronic leukemia and acute leukemia associated with mongolism. Lancet 1:411, 1961. [PMID: 13777696]
10. Caspersson T, Zech L, Johansson C, Modest EJ: Identification of human chromosomes by DNA binding fluorescent agents. Chromosoma 30:215, 1970. [PMID: 4193398]
11. Caspersson T, Gahrton G, Lindsten J, Zech L: Identification of the Philadelphia chromosome as a number 22 by quinacrine mustard fluorescence analysis. Exp Cell Res 63:238, 1970. [PMID: 5276176]
12. Rowley JD: A new consistent abnormality in chronic myelogenous leukemia identified by quinacrine fluorescence and Giemsa staining. Nature 243:290, 1973. [PMID: 4126434]
13. de Klein A, Van Kessel AG, Grosveld G, et al: A cellular oncogene is translocated to the Philadelphia chromosome in chronic myelocytic leukemia. Nature 300:765, 1982.
14. Bartram CR, de Klein A, Hagemeijer A, et al: Translocation of c-abl oncogene correlates with the presence of a Philadelphia chromosome in chronic myelocytic leukemia. Nature 306:277, 1983. [PMID: 6580527]
15. Drucker BJ, Tamura S, Buchdunger E, et al: Effects of a selective inhibitor of the ABL tyrosine kinase in the growth of BCR-ABL positive cells. Nat Med 2:561, 1996.
16. Thomas ED, Clift RA, Fefer A, et al: Marrow transplantation for the treatment of chronic myelogenous leukemia. Ann Intern Med 104:155, 1986. [PMID: 3511810]
17. Redaelli A, Bell C, Casagrande J, et al: Clinical and epidemiologic burden of chronic myelogenous leukemia. Expert Rev Anticancer Ther 4:85, 2004. [PMID: 14748660]
18. Hemminki K, Jiang Y: Familial myeloid leukemias from the Swedish Family-Cancer database. Leuk Res 26:611, 2002. [PMID: 12007511]
19. Ichimaru M, Ichimaru T, Belsky JL: Incidence of leukemia in atomic bomb survivors belonging to a fixed cohort in Hiroshima and Nagasaki 1950–1971. J Radiat Res (Tokyo) 19:262, 1978. [PMID: 739446]
20. Court Brown WM, Doll R: Adult leukemia: Trends in mortality in relation to etiology. Br Med J 1:1063, 1959.
21. Court Brown WM, Doll R: Adult leukemia. Br Med J 1:1753, 1960.
22. Boice JD Jr, Day NE, Anderson A, et al: Second cancers following radiation treatment for cervical cancer. J Natl Cancer Inst 74:955, 1985. [PMID: 3858584]
23. Maloney WC: Radiation leukemia revisited. Blood 70:905, 1987.
24. Lichtman MA: Is there an entity of chemically induced BCR-ABL-positive chronic myelogenous leukemia? Oncologist 13:645, 2008. [PMID: 18586919]
25. Whang-Peng J, Knutsen T: Chromosomal abnormalities, in Chronic Granulocytic Leukaemia, edited by MT Shaw, p 49. Praeger, East Sussex, UK, 1982.
26. Spiers ASD, Bain BJ, Turner JE: The peripheral blood in chronic granulocytic leukemia: A study of 50 untreated Philadelphia positive cases. Scand J Haematol 18:25, 1977. [PMID: 265093]
27. Sandberg AA: The leukemias: The Philadelphia chromosome, in The Chromosomes in Human Cancer and Leukemia, 2nd ed, p 183. Elsevier, New York, 1990.
28. Fialkow PJ, Garther SM, Yoshida A: Clonal origin of chronic myelocytic leukemia in men. Proc Natl Acad Sci U S A 58:1468, 1967. [PMID: 5237880]
29. Fialkow PJ, Jacobsen RJ, Papayannopoulou T: Chronic myelocytic leukemia: Clonal origin in a stem cell common to granulocyte, erythrocyte, platelet, and monocyte/macrophage. Am J Med 63:125, 1977. [PMID: 267431]
30. Koeffler HP, Levine AM, Sparkes LM, Sparkes RS: Chronic myelocytic leukemia: Eosinophils involved in the malignant clone. Blood 55:1063, 1980. [PMID: 6929713]
31. Hayata I, Kakati S, Sandberg AA: On the monoclonal origin of chronic myelocytic leukemia. Proc Jpn Acad 30:351, 1974.
32. Lawler SD, O’Malley F, Lobb DS: Chromosome banding studies in Philadelphia chromosome positive myeloid leukemia. Scand J Haematol 17:17, 1976. [PMID: 1066809]
33. Harrison CJ, Chang J, Johnson D, et al: Chromosomal evidence of a common stem cell in acute lymphoblastic leukemia and chronic granulocytic leukemia. Cancer Genet Cytogenet 13:331, 1984. [PMID: 6595053]
34. Chaganti RSK, Bailey RB, Jhanwar SC, et al: Chronic myelogenous leukemia in the monosomic cell line of a fertile Turner syndrome mosaic (45, X/46, XX). Cancer Genet Cytogenet 5:215, 1982. [PMID: 6950810]
35. Fitzgerald PH, Pickering AF, Eiby JR: Clonal origin of the Philadelphia chromosome and chronic leukemia. Br J Haematol 21:473, 1971. [PMID: 5287677]
36. Groffen J, Stephenson JR, Heisterkamp N, et al: Philadelphia chromosomal breakpoints are clustered within a limited region, bcr, on chromosome 22. Cell 36:93, 1984. [PMID: 6319012]
37. Leibowitz D, Schaefer-Rego K, Popenoe DW, et al: Variable breakpoints on the Philadelphia chromosome in chronic myelogenous leukemia. Blood 66:243, 1985. [PMID: 2988670]
38. Yoffe G, Chinault AG, Talpaz M, et al: Clonal nature of Philadelphia chromosome positive and negative chronic myelogenous leukemia by DNA hybridization analysis. Exp Hematol 15:725, 1987. [PMID: 2440709]
39. Kavalerchik E, Goff D, Jamieson CH: Chronic myeloid leukemia stem cells. J Clin Oncol 26:2911, 2008. [PMID: 18539972]
40. Savona M, Talpaz M: Getting to the stem of chronic myeloid leukaemia. Nat Rev Cancer 8:341, 2008. [PMID: 18385684]
41. Radich JP, Dai H, Mao M, et al: Gene expression changes associated with progression and response in chronic myeloid leukemia. Proc Natl Acad Sci U S A 103:2794, 2006. [PMID: 16477019]
42. Fialkow PJ, Denman AM, Jacobsen RJ, Lowenthal MN: Chronic myelocytic leukemia. Origin of some lymphocytes from leukemic stem cells. J Clin Invest 62:815, 1978. [PMID: 308953]
43. Martin PJ, Najfeld V, Hansen JA, et al: Involvement of the B-lymphoid system in chronic myelogenous leukaemia. Nature 287:49, 1980. [PMID: 6968038]
44. Boggs DR: Hematopoietic stem cell theory in relation to possible lymphoblastic conversion in chronic myeloid leukemia. Blood 44:449, 1974. [PMID: 4546780]
45. Bernheim A, Berger R, Preud’homme JL, et al: Philadelphia chromosome positive blood B lymphocytes in chronic myelocytic leukemia. Leuk Res 5:331, 1981. [PMID: 7026904]
46. Collins S, Coleman H, Groudine M: Expression of bcr and bcr-abl fusion transcripts in normal and leukemic cells. Mol Cell Biol 7:2870, 1987. [PMID: 3670297]
47. Al-Amin A, Lennartz K, Runde V, et al: Frequency of clonal B lymphocytes in chronic myelogenous leukemia evaluated by fluorescence in situ hybridization. Cancer Genet Cytogenet 104:45, 1998. [PMID: 9648557]
48. Torlakovic E, Litz CE, McClure JS, Brunning RD: Direct detection of the Philadelphia chromosome in CD20-positive lymphocytes in chronic myelogenous leukemia by tri-color immunophenotyping/FISH. Leukemia 8:1940, 1994. [PMID: 7526092]
49. Kearney L, Orchard KH, Hibbin JA, Goldman JM: T-cell cytogenetics in chronic granulocytic leukaemia. Lancet 1:858, 1981.
50. Nogueira-Costa R, Spitzer G, Cock A, Trijillo JM: E rosette-positive agar colonies containing the Philadelphia chromosome in chronic myeloid leukemia. Scand J Haematol 34:184, 1985. [PMID: 3871961]
51. Bartram CR, Raghavachar A, Anger B, et al: T lymphocytes lack rearrangement of the bcr gene in Philadelphia chromosome-positive chronic myelogenous leukemia. Blood 69:1682, 1985.
52. Fauser AA, Kanz L, Bross KJ, et al: T cells and probably B cells arise from the malignant clone in chronic myelogenous leukemia. J Clin Invest 75:1080, 1985. [PMID: 3920244]
53. Nitta M, Kato Y, Strife A, et al: Incidence of the B and T lymphocyte lineages in chronic myelogenous leukemia. Blood 66:1053, 1985. [PMID: 2996659]
54. Ariad S, Dajee D, Willem P, Bezwoda WR: Lack of involvement of T-lymphocytes in the leukaemic population during prolonged chronic phase of Philadelphia chromosome positive chronic myeloid leukaemia. Leuk Lymphoma 10:217, 1993. [PMID: 8220120]
55. Tsukamoto N, Karasawa M, Maehara T, et al: The majority of T lymphocytes are polyclonal during the chronic phase of chronic myelogenous leukemia. Ann Hematol 72:61, 1996. [PMID: 8597608]
56. Garicochea B, Chase A, Lazaridou A, Goldman JM: T lymphocytes in chronic myelogenous leukaemia (CML). Leukemia 8:1197, 1994. [PMID: 8035612]
57. Jonas D, Lubbert M, Kawasaki ES, et al: Clonal analysis of bcr-abl rearrangement in T lymphocytes from patients in the chronic myelogenous leukemia. Blood 79:1017, 1992. [PMID: 1371078]
58. Haferlach T, Winkemann M, Nickening C, et al: Which components are involved in Philadelphia-chromosome-positive chronic leukemia? Br J Haematol 97:99, 1997. [PMID: 9136947]
59. Verfaillie C, Miller W, Kay N, McClave P: Adherent lymphokine-activated killer cells in chronic myelogenous leukemia: A benign cell population with potent cytotoxic activity. Blood 74:793, 1989. [PMID: 2473805]
60. Takahashi N, Miura I, Saitoh K, Miura AB: Lineage involvement of stem cells bearing the Philadelphia chromosome in chronic myeloid leukemia in the chronic phase as shown by a combination of fluorescence-activated cell sorting and fluorescence in situ hybridization. Blood 92:4758, 1998. [PMID: 9845542]
61. Muñoz L, Bellido M, Sierra J, Nomdedéu JF: Flow cytometric detection of B cell abnormal maturation in chronic myeloid leukemia. Leukemia 14:339, 2000. [PMID: 20119477]
62. Miura A: Progress in laboratory medicine in chronic myeloid leukemia. Rinsho Byori 46:1226, 1998. [PMID: 9916508]
63. Muñoz L, Bellido M, Sierra J, Nomdedéu JF: Flow cytometric detection of B cell abnormal maturation in chronic myeloid leukemia. Leukemia 14:339, 1999. [PMID: 20119477]
64. Gunsilius E, Duba H-C, Petzer AL, et al: Evidence from a leukaemia model for maintenance of vascular endothelium by bone-marrow-derived endothelial cells. Lancet 355:1688, 2000. [PMID: 10905245]
65. Fialkow PJ, Martin PJ, Najfeld V, et al: Evidence for a multistep pathogenesis of chronic myelogenous leukemia. Blood 58:158, 1981. [PMID: 6972238]
66. Lisker R, Casas L, Mutchinick O, et al: Late-appearing Philadelphia chromosome in two patients with chronic myelogenous leukemia. Blood 56:812, 1980. [PMID: 6932978]
67. Kamada N, Uchino H: Chronologic sequence in appearance of clinical and laboratory findings characteristic of chronic myelogenous leukemia. Blood 51:843, 1978. [PMID: 273449]
68. Smadja N, Krulik M, DeGramont A, et al: Acquisition of a Philadelphia chromosome concomitant with transformation of a refractory anemia into an acute leukemia. Cancer 55:1477, 1985. [PMID: 3856463]
69. Fegan C, Morgan G, Whittaker JA: Spontaneous remission in a patient with chronic myeloid leukaemia. Br J Haematol 72:594, 1989. [PMID: 2775664]
70. Brandt L, Mitelman F, Panani A, Lenner HC: Extremely long duration of chronic myeloid leukaemia with Ph1 negative and Ph1 positive bone marrow cells. Scand J Haematol 16:321, 1976. [PMID: 1065952]
71. Hagemeijer A, Smith EME, Lowenberg B, Abels J: Chronic myeloid leukemia with permanent disappearance of the Ph1 chromosome and development of new clonal subpopulations. Blood 53:1, 1979. [PMID: 281254]
72. Singer JN, Arlin ZA, Najfeld V, et al: Restoration of nonclonal hematopoiesis in chronic myelogenous leukemia (CML) following a chemotherapy induced loss of the Ph1 chromosome. Blood 56:356, 1980. [PMID: 6931619]
73. Sokal JE: Significance of Ph1-negative marrow cells in Ph1 positive chronic granulocytic leukemia. Blood 56:1072, 1980. [PMID: 6934013]
74. Smadja N, Krulik M, Audebert AA, et al: Spontaneous regression of cytogenetic and haematologic anomalies in Ph1-positive chronic myelogenous leukaemia. Br J Haematol 63:257, 1986. [PMID: 3459548]
75. Goldman JM, Kearney L, Pittman S, et al: Hemopoietic stem cell grafting for chronic granulocytic leukemia. Exp Hematol 10:76, 1982.
76. Reiffers J, Vezon G, David B, et al: Philadelphia negative cells in a patient treated with autografting for Ph1 positive chronic granulocytic leukaemia in transformation. Br J Haematol 55:382, 1983. [PMID: 6137235]
77. Reiffers J, Broustet A, Goldman JM: Philadelphia chromosome-negative progenitors in chronic granulocytic leukemia. N Engl J Med 309:1460, 1983. [PMID: 6579348]
78. Coulombel L, Kalousek DK, Eaves CJ, et al: Long-term marrow culture reveals chromosomally normal hemopoietic progenitor cells in patients with Philadelphia chromosome-positive chronic myelogenous leukemia. N Engl J Med 308:1493, 1983. [PMID: 6574315]
79. Degliantoni G, Mangori L, Rizzoli V: In vitro restoration of polyclonal hematopoiesis in a chronic myelogenous leukemia after in vitro treatment with 4-hydroperoxy-cyclophosphamide. Blood 65:753, 1985. [PMID: 3855667]
80. Barnett MJ, Eaves CJ, Phillips GL, et al: Successful autografting in chronic myeloid leukemia after maintenance of marrow in culture. Bone Marrow Transplant 4:345, 1989. [PMID: 2775921]
81. Verfaillie CM, Miller WJ, Boylan K, McGlave PB: Selection of benign primitive hematopoietic progenitors in chronic myelogenous leukemia on the basis of HLA-DR antigen expression. Blood 79:1003, 1992. [PMID: 1371077]
82. Leemhuis T, Leibowitz D, Cox G, et al: Identification of BCR/ABL-negative primitive hematopoietic progenitor cells within chronic myeloid leukemia marrow. Blood 81:801, 1993. [PMID: 7679000]
83. Wang JCY, Lapidot T, Cashman JD, et al: High level engraftment of NOD/SCID mice by primitive normal and leukemic hemopoietic cells from patients with chronic myeloid leukemia in chronic phase. Blood 91:2406, 1998. [PMID: 9516140]
84. Dunbar CE, Stewart FM: Separating the wheat from the chaff: Selection of benign hematopoietic cells in chronic myeloid leukemia. Blood 79:1107, 1992. [PMID: 1536939]
85. Strife A, Clarkson B: Biology of chronic myelogenous leukemia: Is discordant maturation the primary defect? Semin Hematol 25:1, 1988. [PMID: 3279512]
86. Heinzinger M, Waller CF, Rosentiel A, et al: Quality of IL-3 and GCSF-mobilized peripheral blood stem cells in patients with early chronic phase CML. Leukemia 12:333, 1998. [PMID: 9529127]
87. Verfaillie CM, Bhatia R, Miller W, et al: BCR/ABL-negative primitive progenitors suitable for transplantation can be selected from the marrow of most early-chronic phase but not accelerated-phase chronic myelogenous leukemia patients. Blood 87:4770, 1996. [PMID: 8639848]
88. Grand FH, Marley SB, Chase A, et al: BCR/ABL-negative progenitors are enriched in the adherent fraction of CD34+ cells circulating in the blood of chronic phase chronic myeloid leukemia patients. Leukemia 11:1486, 1997. [PMID: 9305602]
89. Carella AM, Podesta M, Frassoni R, et al: Collection of “normal” blood repopulating cells during early hemopoietic recovery after intensive conventional chemotherapy in chronic myelogenous leukemia. Bone Marrow Transplant 12:267, 1993. [PMID: 7694724]
90. Guyootat D, Wahabi K, Viallet A, et al: Selection of BCR/ABL-negative stem cells from marrow or blood of patients with chronic myeloid leukemia. Leukemia 13:991, 1999.
91. Hogge DE, Coulumbel L, Kalousek D, et al: Nonclonal hemopoietic progenitors in a G6PD heterozygote with chronic myelogenous leukemia revealed after long-term marrow culture. Am J Hematol 24:389, 1987. [PMID: 3471084]
92. Deforge M, Boogaerts MA, McGlave PB, Verfaillie CM: BCR/ABL-CD34+HLA-DR– progenitor cells in early phase, but not in more advanced phases, of chronic myelogenous leukemia are polyclonal. Blood 93:284, 1999.
93. Van den Berg D, Wessman M, Murray L, et al: Leukemic burden in subpopulations of CD34+ cells isolated from the mobilized peripheral blood of alpha-interferon-resistant or -intolerant patients with chronic myeloid leukemia. Blood 87:4348, 1996.
94. Podesta M, Piaggio G, Frassoni F, et al: Very primitive hemopoietic cells (LTC-IC) are present in Philadelphia negative cytaphereses collected during early recovery after chemotherapy for chronic myeloid leukemia (CML). Bone Marrow Transplant 16:549, 1995. [PMID: 8528171]
95. Kirk JA, Reems JA, Roecklein BA, et al: Benign marrow progenitors are enriched in the CD34+/HLA-DRlo population but not in the CD34+/CD38lo population in chronic myeloid leukemia: An analysis using interphase fluorescence in situ hybridization. Blood 86:737, 1995. [PMID: 7541674]
96. Lewis ID, Haylock DN, Moore S, et al: Peripheral blood is a source of BCR-ABL–negative pre-progenitors in early chronic phase chronic myeloid leukemia. Leukemia 11:581, 1997. [PMID: 9096699]
97. Maguer-Satta V, Petzer AL, Eaves AC, Eaves CJ: BCR-ABL expression in different subpopulations of functionally characterized Ph+ CD34+ cells from patients with chronic myeloid leukemia. Blood 88:1796, 1996. [PMID: 8781437]
98. Sirard C, Lapidot T, Vormoor J, et al: Normal and leukemia SCID-repopulating cells (SRC) coexist in the bone marrow and peripheral blood from CML patients in chronic phase, whereas leukemic SRC are detected in blast crisis. Blood 87:1539, 1996. [PMID: 8608245]
99. Dazzi F, Capelli D, Hasserjian R, et al: The kinetics and extent of engraftment of chronic myelogenous leukemia cells in nonobese diabetic/severe combined immunodeficiency mice reflect the phase of the donor’s disease: An in vivo model for chronic myelogenous leukemia biology. Blood 92:1390, 1998. [PMID: 9694728]
100. Holyoake TL, Jiang X, Drummond MW, et al: Elucidating critical mechanisms of deregulated stem cell turnover in the chronic phase of chronic myelogenous leukemia. Leukemia 16:549, 2002. [PMID: 11960331]
101. Eaves C, Cashman J, Eaves A: Defective regulation of leukemic hematopoiesis in chronic myeloid leukemia. Leuk Res 22:1085, 1998. [PMID: 9922073]
102. Clarkson BD, Strife A, Wisniewski D, et al: New understanding of the pathogenesis of CML: A prototype of early neoplasia. Leukemia 11:1404, 1997. [PMID: 9305592]
103. Bedi A, Zehnbauer BA, Collector MI, et al: BCR-ABL gene rearrangement and expression of primitive hematopoietic progenitors in chronic myeloid leukemia. Blood 81:2898, 1993. [PMID: 8499629]
104. Moore MA: In vitro culture studies in chronic granulocytic leukaemia. Clin Haematol 6:97, 1977. [PMID: 334418]
105. Siitonen T, Zheng A, Savolainen E-R, Koistinen P: Spontaneous granulocyte-macrophage colony growth by peripheral blood mononuclear cells in myeloproliferative disorders. Leuk Res 20:187, 1996. [PMID: 8628019]
106. Eaves CJ, Eaves AC: Cell culture studies in CML. Baillieres Clin Haematol 1:931, 1987. [PMID: 3332857]
107. Galbraith PR, Abu-Zahra HT: Granulopoiesis in chronic granulocytic leukemia. Br J Haematol 22:135, 1972. [PMID: 4500668]
108. Sjögren U, Brandt L: Composition and mitotic activity of the erythropoietic part of the bone marrow in chronic myeloid leukaemia. Scand J Haematol 12:18, 1974. [PMID: 20117103]
109. Verfaillie CM: Stem cells in chronic myelogenous leukemia. Hematol Oncol Clin North Am 11:1079, 1997. [PMID: 9443047]
110. Ghaffari S, Dougherty GJ, Lansdorp PM, et al: Differentiation-associated changes in CD44 isoform expression during normal hematopoiesis and their alteration in chronic myeloid leukemia. Blood 86:2976, 1995. [PMID: 7579390]
111. Kawaishi K, Kimura A, Katch O, et al: Decreased L-selectin expression in CD34-positive cells from patients with chronic myelocytic leukaemia. Br J Haematol 93:367, 1996. [PMID: 8639430]
112. Turkina AG, Baryshnikov AY, Sedyakhina NP, et al: Studies of P-glycoprotein in chronic myelogenous leukaemia patients: Expression, activity and correlations with CD34 antigen. Br J Haematol 92:88, 1996. [PMID: 8562416]
113. Agarwal R, Doren S, Hicks B, Dunbar CE: Long-term culture of chronic myelogenous leukemia marrow cells on stem cell factor-deficient stroma favors benign progenitors. Blood 85:1306, 1995. [PMID: 7532038]
114. Moore S, Haylock DN, Levesque J-P, et al: Stem cell factor as a single agent induces selective proliferation of the Philadelphia chromosome positive fraction of chronic myeloid leukemia CD34+ cells. Blood 92:2461, 1998. [PMID: 9746786]
115. Chasty RC, Lucas GS, Owen-Lynch PJ, et al: Macrophage inflammatory protein-1 alpha receptors are present on cells enriched for CD34 expression from patients with chronic myeloid leukemia. Blood 86:4270, 1995. [PMID: 7492787]
116. Cashman JD, Eaves CJ, Sarris AH, Eaves AC: MCP-1, not MIP-1, is the endogenous chemokine that cooperates with TGF- to inhibit the cycling of primitive normal but not leukemic (CML) progenitors in long-term human marrow cultures. Blood 92:2338, 1998. [PMID: 9746772]
117. Murohashi I, Endho K, Nishida S, et al: Differential effects of TGF-beta 1 on normal and leukemic human hematopoietic cell proliferation. Exp Hematol 23:970, 1995. [PMID: 7543418]
118. Gordon MY, Dowding C, Riley G, et al: Altered adhesive interactions with marrow stroma of haematopoietic progenitor cells in chronic myeloid leukaemia. Nature 328:342, 1987. [PMID: 3474529]
119. Dowding C, Guo A-P, Osterholz J, et al: Interferon- overrides the deficient adhesion of chronic myeloid leukemia primitive progenitor cells to bone marrow stromal cells. Blood 78:499, 1991. [PMID: 1906352]
120. Bhatia R, Wayner EA, McGlave PB, Verfaillie CM: Interferon- restores normal adhesion of chronic myelogenous leukemia hematopoietic progenitors to bone marrow stroma by correcting impaired 1 integrin receptor function. J Clin Invest 94:384, 1994. [PMID: 7518835]
121. Verfaillie CM: Stem cells in chronic myelogenous leukemia. Hematol Oncol Clin North Am 11:1079, 1997. [PMID: 9443047]
122. Bhatia R, Munthe HA, Verfaillie CM: Tyrphostin AG957, a tyrosine kinase inhibitor with anti-BCR/ABL tyrosine activity restores 1 integrin-mediated adhesion and inhibiting signaling in chronic myelogenous leukemia hematopoietic progenitors. Leukemia 12:1708, 1998. [PMID: 9823945]
123. Lundell BI, McCarthy JB, Kovach NL, Verfaillie CM: Activation of beta 1 integrins on CML progenitors reveals cooperation between beta1 integrins and CD44 in the regulation of adhesion and proliferation. Leukemia 11:822, 1997. [PMID: 9177435]
124. Ghaffari S, Dougherty GJ, Eaves AC, Eaves CJ: Altered patterns of CD44 epitope expression in human chronic and acute myeloid leukemia. Leukemia 10:1773, 1996. [PMID: 8892681]
125. Vijayan KV, Advani SH, Zingde SM: Chronic myeloid leukemic granulocytes exhibit reduced and altered binding to P-selectin; modification in the CD15 antigens and sialylation. Leuk Res 21:59, 1997. [PMID: 9029187]
126. Deininger MW, Vieira S, Mendiola R, et al: BCR-ABL tyrosine kinase activity regulates the expression of multiple genes implicated in the pathogenesis of chronic myeloid leukemia. Cancer Res 60:2049, 2000. [PMID: 10766197]
127. Verfaillie CM, Hurley R, Lundell BI, et al: Integrin-mediated regulation of hematopoiesis: Do BCR/ABL-induced defects in integrin function underlie the abnormal circulation and proliferation of CML progenitors? Acta Haematol 29:40, 1997.
128. Symington BE: Growth signalling through the alpha 5 beta 1 fibronectin receptor. Biochem Biophys Res Commun 208:126, 1995. [PMID: 7534071]
129. Bhatia R, McCarthy JB, Verfaillie CM: Interferon-alpha restores normal beta 1 integrin-mediated inhibition of hematopoietic progenitor proliferation by the marrow microenvironment in chronic myelogenous leukemia. Blood 87:3883, 1996. [PMID: 8611716]
130. Wertheim JA, Forsythe K, Druker BJ, et al: BCR-ABL-induced adhesion defects are tyrosine kinase-independent. Blood 99:4122, 2002. [PMID: 12010816]
131. Fruehauf S, Topaly J, Schad M, Paschka P, et al: Imatinib restores expression of CD62L in BCR-ABL-positive cells. J Leukoc Biol 73:600, 2003. [PMID: 12714574]
132. Salgia R, Li JL, Ewaniuk DS, et al: BCR/ABL induces multiple abnormalities of cytoskeletal function. J Clin Invest 100:46, 1997. [PMID: 9202056]
133. Lewis JM, Baskaran R, Taagepera S, et al: Integrin regulation of c-ABL tyrosine kinase activity and cytoplasmic-nuclear transport. Proc Natl Acad Sci U S A 93:15174, 1996. [PMID: 8986783]
134. Renshaw MW, McWhirter JR, Wang JY: The human leukemia onco-gene bcr-abl abrogates the anchorage requirement but not the growth factor requirement for proliferation. Mol Cell Biol 15:1286, 1995. [PMID: 7862122]
135. Salgia R, Brunkhorst B, Pisick E, et al: Increased tyrosine phosphorylation of focal adhesion proteins in myeloid cell lines expressing p210BCR/ABL. Oncogene 11:1149, 1995. [PMID: 7566975]
136. Rudkin GT, Hungerford DA, Nowell PC: DNA content of chromosome Ph1 and chromosome 21 in human chronic granulocytic leukemia. Science 144:1229, 1964. [PMID: 14150328]
137. O’Riordan ML, Robinson JA, Buckton KE, Evans HJ: Distinguishing between the chromosome involved in Down’s syndrome (trisomy 21) and chronic myeloid leukaemia (Ph1) by fluorescence. Nature 230:167, 1971. [PMID: 4100531]
138. Lawler SD: The cytogenetics of chronic granulocytic leukemia. Clin Haematol 6:55, 1977. [PMID: 334417]
139. Melo JV, Yan XH, Diamond J, Goldman JM: Balanced parental contribution to the ABL component of the BCR-ABL gene in chronic myeloid leukemia. Leukemia 9:734, 1995. [PMID: 7723412]
140. Chissoe SL, Bodenteich A, Wang YF, et al: Sequence and analysis of the human ABL gene, the BCR gene, and regions involved in the Philadelphia chromosomal translocation. Genomics 27:67, 1995. [PMID: 7665185]
141. Melo JV, Deininger MW: Biology of chronic myelogenous leukemia-signaling pathways of initiation and transformation. Hematol Oncol Clin North Am 18:545, 2004. [PMID: 15271392]
142. Daley GQ, Beu Neriah Y: Implicating the bcr/abl gene in the pathogenesis of Philadelphia chromosome-positive human leukemia. Adv Cancer Res 57:151, 1991. [PMID: 1950703]
143. Heisterkamp N, Groffen J, Stephenson JR, et al: Chromosomal localization of human cellular homologues of two viral oncogenes. Nature 299:747, 1982. [PMID: 7121606]
144. Heisterkamp N, Stephenson JR, Groffen J, et al: Localization of the c-abl oncogene adjacent to a translocation breakpoint in chronic myelocytic leukemia. Nature 306:239, 1983. [PMID: 6316147]
145. Konopka JB, Witte ON: Activation of the abl oncogene in murine and human leukemias. Biochim Biophys Acta 823:1, 1985. [PMID: 2996602]
146. Collins SJ, Groudine MT: Rearrangements and amplification of c-abl sequences in the human chronic myelogenous leukemia cell line K562. Proc Natl Acad Sci U S A 80:4813, 1983. [PMID: 6308652]
147. Canaani E, Gale RP, Steiner-Seltz D, et al: Altered transcription of an oncogene in chronic myelocytic leukemia. Lancet 1:593, 1984. [PMID: 6142307]
148. Gale RP, Canaani E: An 8 kilobase abl RNA transcript in chronic myelogenous leukemia. Proc Natl Acad Sci U S A 81:5648, 1984. [PMID: 6091099]
149. Collins SJ, Kubonishi I, Miyoshi I, Groudine MT: Altered transcription of the c-abl oncogene in K562 and other chronic myelogenous leukemia cells. Science 225:72, 1984. [PMID: 6587568]
150. Leibowitz D, Cubbon RM, Bank A: Increased expression of a novel c-abl related RNA in K562 cells. Blood 65:526, 1985. [PMID: 2578837]
151. Konopka JB, Watanabe SM, Witte ON: An alteration of the human c-abl protein in K562 leukemia cells unmasks associated tyrosine kinase activity. Cell 37:1035, 1984. [PMID: 6204766]
152. Konopka JB, Watanabe SM, Singer JW, et al: Cell lines and clinical isolates derived from Ph1-positive chronic myelogenous leukemia patients express c-abl proteins with a common structural alteration. Proc Natl Acad Sci U S A 82:1810, 1985. [PMID: 3856862]
153. Stam K, Heisterkamp N, Grosveld G, et al: Evidence of a new chimeric bcr/c-abl mRNA in patients with chronic myelocytic leukemia and the Philadelphia chromosome. N Engl J Med 313:1429, 1985. [PMID: 3864009]
154. Ben-Neriah Y, Daley GQ, Mes-Masson A-M, et al: The chronic myelogenous leukemia-specific P210 protein is the product of the bcr/abl hybrid gene. Science 233:212, 1985.
155. Maxwell SA, Kurzrock R, Parson SJ, et al: Analysis of P210bcr/abl tyrosine protein kinase activity in various subtypes of Philadelphia chromosome-positive cells from chronic myelogenous leukemia patients. Cancer Res 47:1731, 1987. [PMID: 2434223]
156. Kurzrock R, Kloetzer WS, Talpaz M, et al: Identification of molecular variants of P210BCR–ABL in chronic myelogenous leukemia. Blood 70:233, 1987. [PMID: 2885048]
157. Xu DQ, Galibert F: Restriction fragment length polymorphism caused by a deletion within the human c-abl gene (ABL). Proc Natl Acad Sci U S A 83:3447, 1986. [PMID: 3010299]
158. Popenoe DW, Schaefer-Rego K, Mears JC, et al: Frequent and extensive deletion during the 9,22 translocation in CML. Blood 68:1123, 1986. [PMID: 3021264]
159. Shtivelman E, Gale RP, Dreazen O, et al: Bcr-abl RNA in patients with chronic granulocytic leukemia. Blood 69:971, 1987. [PMID: 3101769]
160. McWhirter JR, Wang JJ: Activation of tyrosine kinase and microfilament-binding functions of c-abl by bcr sequences in bcr/abl fusion proteins. Mol Cell Biol 11:1553, 1991. [PMID: 1705008]
161. Bernards A, Rubin CM, Westbrook CA, et al: The first intron in the human c-abl gene is at least 200 kilobases long and is the target for translocations in chronic myelogenous leukemia. Mol Cell Biol 7:3231, 1987. [PMID: 3313010]
162. Eisenberg A, Silver R, Soper L, et al: The location of breakpoints within the breakpoint cluster region (bcr) of chromosome 22 in chronic myeloid leukemia. Leukemia 2:642, 1988. [PMID: 3172841]
163. Collins SJ: Breakpoints on chromosomes 9 and 22 in Philadelphia chromosome-positive chronic myelogenous leukemia. J Clin Invest 78:1392, 1986. [PMID: 3021820]
164. Heisterkamp N, Stam K, Groffen J, et al: Structural organization of the bcr gene and its role in the Ph1 translocation. Nature 315:758, 1985. [PMID: 2989703]
165. Gao L-M, Goldman J: Long-range mapping of the normal BCR gene. Leukemia 5:555, 1991. [PMID: 1649356]
166. Melo JV: BCR-ABL gene variants. Baillieres Clin Haematol 10:203, 1997. [PMID: 9376660]
167. Saglio G, Pane F, Gottardi E, et al: Consistent amounts of acute leukemia-associated P190BCR/ABL transcripts are expressed by chronic myelogenous leukemia patients at diagnosis. Blood 87:1075, 1996. [PMID: 8562932]
168. Honda H, Oda H, Suzuki T, et al: Development of acute lymphoblastic leukemia and myeloproliferative disorder in transgenic mice expressing p210bcr/abl: A novel transgenic model for human Ph1-positive leukemias. Blood 91:2067, 1998. [PMID: 9490692]
169. Maru Y, Witte ON: The BCR gene encodes a novel serine/threonine kinase activity within a single exon. Cell 67:459, 1991. [PMID: 1657398]
170. Muller AJ, Young JC, Pendergast A-M, et al: BCR first exon sequences specifically activate the BCR/ABL tyrosine kinase oncogene of Philadelphia chromosome-positive human leukemia. Mol Cell Biol 11:1785, 1991. [PMID: 2005881]
171. Diekmann D, Brill S, Garrett MD, et al: BCR encodes a GTPase-activating protein for p21rac. Nature 351:400, 1991. [PMID: 1903516]
172. Melo JV, Gordon DE, Goldman JM: The ABL-BCR fusion gene is expressed in chronic myeloid leukemia. Blood 81:158, 1993. [PMID: 8417787]
173. Bartram CR, de Klein A, Hagemeijer A, et al: Translocation of the human c-abl oncogene correlates with the presence of a Philadelphia chromosome in chronic myelocytic leukaemia. Nature 306:277, 1983. [PMID: 6580527]
174. Selleri L, Narni F, Emilia G, et al: Philadelphia-positive chronic myeloid leukemia with a chromosome 22 breakpoint outside the breakpoint cluster region. Blood 70:1659, 1987. [PMID: 3478105]
175. Mohamed AN, Koppitch F, Varterasian M, et al: BCR/ABL fusion located on chromosome 9 in chronic myeloid leukemia with a masked Ph chromosome. Genes Chromosomes Cancer 13:133, 1995. [PMID: 7542908]
176. Morris C, Jeffs A, Smith T, et al: BCR gene recombines with genomically distinct sites on band 11Q13 in complex BCR-ABL translocations of chronic myeloid leukemia. Oncogene 12:677, 1996. [PMID: 8637725]
177. Andreasson P, Johansson B, Carlsson M, et al: BCR/ABL-negative chronic myeloid leukemia with ETV6/ABL fusion. Genes Chromosomes Cancer 20:299, 1997. [PMID: 9365838]
178. Rozman C, Urbano-Ispizua A, Cervantes F, et al: Analysis of the clinical relevance of the breakpoint location within M-BCR and the type of chimeric mRNA in chronic myelogenous leukemia. Leukemia 9:1104, 1995. [PMID: 7596178]
179. Verschraegen CF, Kantarjian HM, Hirsch-Ginsberg C, et al: The breakpoint cluster region site in patients with Philadelphia chromosome-positive chronic myelogenous leukemia. Clinical, laboratory, and prognostic correlations. Cancer 76:992, 1995. [PMID: 8625225]
180. Zaccaria A, Martinelli G, Testoni N, et al: Does the type of BCR/ABL junction predict the survival of patients with Ph1-positive chronic myeloid leukemia? Leuk Lymphoma 16:231, 1995. [PMID: 7719231]
181. Ohno T, Hada S, Sugiyama T, et al: Chronic myeloid leukemia with minor bcr breakpoint developed hybrid type of blast crisis. Am J Hematol 57:320, 1998. [PMID: 9544977]
182. Melo JV: The diversity of BCR-ABL fusion proteins and their relationship to leukemia phenotype. Blood 88:2375, 1996. [PMID: 8839828]
183. Rubinstein R, Purves LR: A novel BCR-ABL rearrangement in a Philadelphia chromosome-positive chronic myelogenous leukaemia variant with thrombocythaemia. Leukemia 12:230, 1998. [PMID: 9519786]
184. Hochhaus A, Reither A, Skladny H, et al: A novel BCR-ABL fusion gene (e6a2) in a patient with Philadelphia chromosome-negative chronic myelogenous leukemia. Blood 88:2236, 1996. [PMID: 8822944]
185. Briz M, Vilches C, Cabrera R, et al: Typical chronic myelogenous leukemia with e19a2 junction BCR/ABL transcript. Blood 90:5024, 1997. [PMID: 9389724]
186. McLaughlin J, Chianese E, Witte ON: In vitro transformation of immature hemopoietic cells by P210 bcr/abl oncogene product of the Philadelphia chromosome. Proc Natl Acad Sci U S A 84:6558, 1987. [PMID: 3498165]
187. Daley GQ, McLaughlin J, Witte ON, Baltimore D: The CML-specific P210 bcr/abl protein, unlike v-abl, does not transform NIH/3T3 fibroblasts. Science 237:532, 1987. [PMID: 2440107]
188. Elefanty AG, Hariharan IK, Cory S: Bcr-abl, the hallmark of chronic myeloid leukaemia in man, induces multiple haemopoietic neoplasms in mice. EMBO J 9:1069, 1990. [PMID: 1691092]
189. Daley GQ, VanEtten RA, Baltimore D: Induction of chronic myelogenous leukemia in mice by the p210bcr/abl gene of the Philadelphia chromosome. Science 247:824, 1990. [PMID: 2406902]
190. Voncken JW, Morris C, Pattengale P, et al: Clonal development and karyotype evolution during leukemogenesis of BCR/ABL transgenic mice. Blood 79:1029, 1992. [PMID: 1737087]
191. Gishizky ML, Johnson-White J, Witte O: Efficient transplantation of BCR-ABL-induced chronic myelogenous leukemia-like syndrome in mice. Proc Natl Acad Sci U S A 90:3755, 1993. [PMID: 8475126]
192. Daley GQ: Animal models of BCR/ABL-induced leukemias. Leuk Lymphoma 11:57, 1993. [PMID: 8251918]
193. Voncken JW, Kaartinen V, Pattengale PK, et al: BCR/ABL P210 and P190 cause distinct leukemia in transgenic mice. Blood 86:4603, 1995. [PMID: 8541551]
194. Honda H, Oda H, Suzuki T, et al: Development of acute lymphoblastic leukemia and myeloproliferative disorder in transgenic mice expressing p210bcr/abl: A novel transgenic model for human Ph 1-positive leukemias. Blood 91:2067, 1998. [PMID: 9490692]
195. Pear WS, Miller JP, Xu L, et al: Efficient and rapid induction of a chronic myelogenous leukemia-like myeloproliferative disease in mice receiving P210 bcr/abl-transduced bone marrow. Blood 92:3780, 1998. [PMID: 9808572]
196. Honda M, Ohno S, Takahashi T, et al: Establishment, characterization, and chromosomal analysis of new leukemic cell lines derived from MT/p210/bcr/abl transgenic mice. Exp Hematol 26:188, 1998. [PMID: 9502614]
197. Zhang X, Ren R: Bcr-Abl efficiency induces in a myeloproliferative disease and production of excess interleukin-3 and granulocyte-macrophage colony-stimulating factor in mice: A novel model for chronic myelogenous leukemia. Blood 92:3829, 1998. [PMID: 9808576]
198. Elefanty AG, Corsy S: Bcr-abl-induced cell lines can switch from mast cell to erythroid or myeloid differentiation in vitro. Blood 79:1271, 1992. [PMID: 1536951]
199. Van Etten RA: Pathogenesis and treatment of Ph+ leukemia: Recent insights from mouse models. Curr Opin Hematol 8:224, 2001.
200. Bose S, Deininger M, Goora-Tybor J, et al: The presence of typical and atypical BCR-ABL fusion genes in leukocytes of normal individuals: Biological significance and implications for the assessment of minimal residual disease. Blood 92:3362, 1998. [PMID: 9787174]
201. Hirai HS, Tanaka M, Azuma Y, et al: Transforming genes in human leukemia cells. Blood 66:1371, 1985. [PMID: 2998510]
202. Clarkson BD, Strife A, Wisniewski D, et al: New understanding of the pathogenesis of CML: A prototype of early neoplasia. Leukemia 11:1404, 1997. [PMID: 9305592]
203. Verfaillie CM: Chronic myelogenous leukemia: From pathogenesis to therapy. J Hematother 8:3, 1999. [PMID: 10192297]
204. Pasternak G, Hochhaus A, Schultheis B, Hehlmann R: Chronic myelogenous leukemia: Molecular and cellular aspects. J Cancer Res Clin Oncol 124:643, 1998. [PMID: 9879825]
205. Gotoh A, Broxmeyer HE: The function of BCR/ABL and related protooncogenes. Curr Opin Hematol 4:3, 1997. [PMID: 9050373]
206. Sattler M, Salgia R: Activation of hematopoietic growth factor signal transduction pathways by the human oncogene BCR/ABL. Cytokine Growth Factor Rev 8:63, 1997. [PMID: 9174663]
207. Skorski T, Kanakaraj P, Nieborowska-Skorska M, et al: Phosphatidylinositol-3 kinase activity is regulated by BCR/ABL and is required for the growth of Philadelphia chromosome-positive cells. Blood 86:726, 1995. [PMID: 7606002]
208. Skorski T, Nieborowska-Skorska M, Szczylik C, et al: C-RAF-1 serine/threonine kinase is required in BCR/ABL-dependent and normal hematopoiesis. Cancer Res 55:2275, 1995. [PMID: 7757976]
209. Goga A, McLaughlin J, Afar DE, et al: Alternative signals to RAS for hematopoietic transformation by the BCR-ABL oncogene. Cell 82:981, 1995. [PMID: 7553858]
210. Salgia R, Uemura N, Okuda K, et al: CRKL links p210BCR/ABL with paxillin in chronic myelogenous leukemia cells. J Biol Chem 270:29145, 1995. [PMID: 7493940]
211. De Jong R, ten Hoeve J, Heisterkamp N, Groffen J: Crkl is complexed with tyrosine-phosphorylated Cbl in Ph-positive leukemia. J Biol Chem 270:21468, 1995.
212. Salgia R, Pisick E, Sattler M, et al: P130CAS forms a signalling complex with the adapter protein CRKL in hematopoietic cells transformed by the BCR/ABL oncogene. J Biol Chem 271:25198, 1996. [PMID: 8810278]
213. Sattler M, Griffin JD: Molecular mechanisms of transformation by the BCR-ABL oncogene. Semin Hematol 40:4, 2003. [PMID: 12783368]
214. Melo JV, Deininger MW: Biology of chronic myelogenous-signaling pathways of initiation and transformation. Hematol Oncol Clin North Am 18:545, 2004. [PMID: 15271392]
215. Wong S, Witte ON: The BCR-ABL story: Bench to bedside and back. Annu Rev Immunol 22:247, 2004. [PMID: 15032571]
216. Sattler M, Verma S, Shrinkhande G, et al: The BCR/ABL tyrosine kinase induces production of reactive species in hematopoietic cells. J Biol Chem 275:24273, 2000. [PMID: 10833515]
217. Sattler M, Salgia R, Okuda K, et al: The proto-oncogene product p120CBL and the adaptor proteins CRKL and c-CR link c-ABL, p190BCR/ABL and p210BCR/ABL to the phosphatidylinositol-3; kinase pathway. Oncogene 12:832, 1996.
218. Salgia R, Sattler M, Pisick E, et al: P210BCR/ABL induces formation of complexes containing focal adhesion proteins and the protooncogene product p120c-CBL. Exp Hematol 24:310, 1996. [PMID: 8641358]
219. De Jong R, van Wijk A, Haataja L, et al: BCR/ABL-induced leukemogenesis causes phosphorylation of Hef2 and its association with Crkl. J Biol Chem 272:32649, 1997.
220. Bollag G, Clapp DW, Shih S, et al: Loss of NF1 results in activation of the Ras signaling pathway and leads to aberrant growth in haematopoietic cells. Nat Genet 12:144, 1996. [PMID: 8563751]
221. Carpino N, Wisniewski D, Strife A, et al: P62dok: A constitutively tyrosine-phosphorylated, GAP-associated protein in chronic myelogenous leukemia progenitor cells. Cell 88:197, 1997. [PMID: 9008160]
222. Yamanashi Y, Baltimore D: Identification of the Abl- and ras GAP-associated 62 kDa protein as a docking protein, Dok. Cell 88:205, 1997. [PMID: 9008161]
223. Reuther JY, Reuther GW, Cortez D, et al: A requirement for NFkappaB activation in BCR/ABL-mediated transformation. Genes Dev 1:12:968, 1998.
224. LaMontagne KR, Flint AJ, Franza BR, et al: Protein tyrosine phosphatase 1B antagonizes signalling by oncoprotein tyrosine kinase p210 bcr/abl in vivo. Mol Cell Biol 18:2965, 1998. [PMID: 9566916]
225. Chai SK, Nichols GL, Rothman P: Constitutive activation of JAKs and STATs in BCR-abl-expressing cell lines and peripheral blood cells derived from leukemic patients. J Immunol 159:4720, 1997. [PMID: 9366395]
226. Shuai K, Halpern J, ten Hoeve J, et al: Constitutive activation of STAT5 by the BCR-ABL oncogene in chronic myelogenous leukemia. Oncogene 13:247, 1996. [PMID: 8710363]
227. Wilson-Rawls J, Xie S, Liu J, et al: P210 Bcr-Abl interacts with the interleukin 3 receptor beta (c) subunit and constitutively induces its tyrosine phosphorylation. Cancer Res 56:3426, 1996. [PMID: 8758906]
228. Chuang TH, Xu X, Kaartinen V, et al: Abl and Bcr are multifunctional regulators of the Rho GTP-binding protein family. Proc Natl Acad Sci U S A 92:10282, 1995. [PMID: 7479768]
229. Afar DE, Witte O: Characterization of breakpoint cluster region kinase and SH2-binding activities. Methods Enzymol 256:125, 1995. [PMID: 7476425]
230. Gishizky ML, Cortez D, Pendergast AM: Mutant forms of growth factor-binding protein-2 reverse BCR-ABL-induced transformation. Proc Natl Acad Sci U S A 92:10889, 1995. [PMID: 7479904]
231. Raitano AB, Halpern JR, Hambuch TM, Sawyers CL: The Bcr-Abl leukemia oncogene activates Jun kinase and requires Jun for transformation. Proc Natl Acad Sci U S A 92:11746, 1995. [PMID: 8524841]
232. Miyamura T, Nishimura J, Yufu Y, Nawata H: Interaction of BCRABL with the retinoblastoma protein in Philadelphia chromosome-positive cell lines. Int J Hematol 67:115, 1997.
233. Largaespada DA, Brannan CI, Jenkins NA, Copeland NG: NF1 deficiency causes Ras-mediated granulocyte/macrophage colony stimulating factor hypersensitivity and chronic myeloid leukaemia. Nat Genet 12:137, 1996. [PMID: 8563750]
234. Amos TA, Lewis JL, Grand FH, et al: Apoptosis in chronic myeloid leukaemia: Normal responses by progenitor cells to growth factor deprivation, X-irradiation and glucocorticoids. Br J Haematol 91:387, 1995. [PMID: 8547080]
235. Bedi A, Barber JP, Bedi GC, et al: BCR-ABL-mediated inhibition of apoptosis with delay of G2/M transition after DNA damage: A mechanism of resistance to multiple anticancer agents. Blood 86:1148, 1995. [PMID: 7620167]
236. Amarante-Mendes GP, Naekyung KC, Liu L, et al: Bcr-Abl exerts its antiapoptotic effect against diverse apoptotic stimuli through blockage of mitochondrial release of cytochrome C and activation of caspase-3. Blood 92:1700, 1998.
237. Maguer-Satta V, Burl S, Liu L, et al: BCR-ABL accelerates C2-ceramide-induced apoptosis. Oncogene 16:237, 1998. [PMID: 9464542]
238. Pierson BA, Miller JS: CD56+bright and CD56+dim natural killer cells in patients with chronic myelogenous leukemia progressively decrease in number, respond less to stimuli that recruit clonogenic natural killer cells, and exhibit decreased proliferation on a per cell basis. Blood 88:2279, 1996. [PMID: 8822949]
239. Gissinger H, Kurzrock R, Wetzler M, et al: Apoptosis in chronic myelogenous leukemia: Studies of stage-specific differences. Leuk Lymphoma 25:121, 1997.
240. Boultwood J, Peniket A, Watkins F, et al: Telomere length shortening in chronic myelogenous leukemia is associated with reduced time to accelerated phase. Blood 96:358, 2000. [PMID: 10891474]
241. Terasaki Y, Okamura H, Ohtake S, Nakao S: Accelerated telomere length shortening in granulocytes: A diagnostic marker for myeloproliferative diseases. Exp Hematol 30:1399, 2002. [PMID: 12482501]
242. Drummond MW, Lennard A, Brummendorf TH, Holyoake TL: Telomere shortening correlates with prognostic score at diagnosis and proceeds rapidly during progression of chronic myeloid leukemia. Leuk Lymphoma 45:1775, 2004. [PMID: 15223635]
243. Campbell LJ, Fidler C, Eagleton H, et al: HTERT, the catalytic component of telomerase, is downregulated in the haematopoietic stem cells of patients with chronic myeloid leukaemia. Leukemia 20:671, 2006. [PMID: 16498395]
244. Ohyashiki K, Ohyashiki JH, Iwama H, et al: Telomerase activity and cytogenetic changes in chronic myeloid leukemia with disease progression. Leukemia 11:190, 1997. [PMID: 9009079]
245. Brümmendorf TH, Ersöz I, Hartmann U, et al: Telomere length in peripheral blood granulocytes reflects response to treatment with imatinib in patients with chronic myeloid leukemia. Blood 101:375, 2003. [PMID: 20120246]
246. Thompson RB, Stainsby D: The clinical and haematological features of chronic granulocytic leukaemia in the chronic phase, in Chronic Granulocytic Leukaemia, edited by MT Shaw, p 137. Praeger, East Sussex, UK, 1982.
247. Cortes JE, Talpaz M, Kantarkian H: Chronic myelogenous leukemia: A review. Am J Med 100:555, 1996. [PMID: 8644769]
248. Goldman JM: Chronic myeloid leukemia. Curr Opin Hematol 4:277, 1997. [PMID: 9260056]
249. Lichtman MA, Rowe JM: Hyperleukocytic leukemias: Rheological, clinical and therapeutic considerations. Blood 60:279, 1982. [PMID: 7046844]
250. Rowe JM, Lichtman MA: Hyperleukocytosis and leukostasis: Common features of childhood chronic myelogenous leukemia. Blood 63:1230, 1984. [PMID: 6585236]
251. Lichtman MA, Heal J, Rowe JM: Hyperleukocytic leukaemia. Baillieres Clin Haematol 1:725, 1987. [PMID: 3327563]
252. Ungaro PC, Gonzalez JJ, Werk EE, MacKay JC: Chronic myelogenous leukemia presenting clinically as diabetes insipidus. N C Med J 45:640, 1984. [PMID: 6594583]
253. Juan D, Hsu S-D, Hunter J: Case report of vasopressin-responsive diabetes insipidus associated with chronic myelogenous leukemia. Cancer 56:1468, 1985. [PMID: 3861227]
254. Brydon J, Lucky PA, Duffy T: Acne urticaria associated with chronic myelogenous leukemia. Cancer 56:2083, 1985. [PMID: 3161608]
255. Cohen PR, Talpaz M, Kurzrock R: Malignancy-associated Sweet’s syndrome: A review of the world’s literature. J Clin Oncol 6:1887, 1988. [PMID: 3058878]
256. López JLB, Fonseca E, Mauso F: Sweet’s syndrome during the chronic phase of chronic myeloid leukemia. Acta Haematol 84:207, 1990. [PMID: 20120531]
257. Nestok BR, Goldstein JD, Lipkovic P: Splenic rupture as a cause of sudden death in undiagnosed chronic myelogenous leukemia. Am J Forensic Med Pathol 9:241, 1988. [PMID: 3177355]
258. Giagounidis AAN, Burk M, Meckenstock G, et al: Pathological rupture of the spleen in hematologic malignancies. Ann Hematol 73:297, 1996. [PMID: 9003161]
259. Hild DH, Myers TJ: Hyperviscosity in chronic granulocytic leukemia. Cancer 46:1418, 1980. [PMID: 6932252]
260. D’Hondt L, Guillaume TH, Hemblit Y, Symann M: Digital necrosis associated with chronic myeloid leukemia. Acta Clin Belg 52:49, 1997. [PMID: 9085619]
261. Arbaje YM, Betran G: Chronic myelogenous leukemia complicated by autoimmune hemolytic anemia. Am J Med 88:197, 1990. [PMID: 2301449]
262. Steegman JL, Pinilla I, Requena MJ, et al: The direct antiglobulin test is frequently positive in chronic myeloid leukemia patients treated with interferon-. Transfusion 37:446, 1997.
263. Hoppin EC, Lewis JP: Polycythemia rubra vera progressing to Ph1-positive chronic myelogenous leukemia. Ann Intern Med 83:820, 1975. [PMID: 1060394]
264. Shenkenberg TD, Waddell CC, Rice L: Erythrocytosis and marked leukocytosis in overlapping myeloproliferative diseases. South Med J 75:868, 1982. [PMID: 6953606]
265. Haas O, Hinterberger W, Morz R: Pure red cell aplasia as possible early manifestation of chronic myeloid leukemia. Am J Hematol 27:20, 1986.
266. Mijovic A, Rolovic Z, Novak A, et al: Chronic myeloid leukemia associated with pure red cell aplasia and terminating in promyelocytic transformation. Am J Hematol 31:128, 1989. [PMID: 2500017]
267. Inbal A, Aktein E, Barak I, Meytes D: Cyclic leukocytosis and long survival in chronic myeloid leukemia. Acta Haematol 69:353, 1983. [PMID: 6404119]
268. Umemura T, Hirata J, Kaneko S, et al: Periodic appearance of erythropoietin-independent erythropoiesis in chronic myelogenous leukemia with cyclic oscillation. Acta Haematol 76:230, 1986. [PMID: 3107311]
269. Mitus WJ, Kiossoglou KA: Leukocyte alkaline phosphatase in myeloproliferative syndrome. Ann N Y Acad Sci 155:976, 1968.
270. DePalma L, Delgado P, Werner M: Diagnostic discrimination and cost-effective assay strategy for leukocyte alkaline phosphate. Clin Chim Acta 6:83, 1996.
271. Pedersen F: Functional and biochemical phenotype in relation to cellular age of differentiated neutrophils in chronic myeloid leukemia. Br J Haematol 51:339, 1982. [PMID: 7049217]
272. Rambaldi A, Terao M, Bettoni S, et al: Differences in the expression of alkaline phosphatase in mRNA in chronic myelogenous leukemia and paroxysmal nocturnal hemoglobinuria polymorphonuclear leukocytes. Blood 73:1113, 1989. [PMID: 2930836]
273. Perillie PE: Studies of the changes in leukocyte alkaline phosphatase following pyrogen stimulation in chronic granulocytic leukemia. Blood 29:401, 1967. [PMID: 5227141]
274. Rustin GJS, Goldman JM, McCarthy D, et al: An extracellular factor controls neutrophil alkaline phosphatase in chronic granulocytic leukemia. Br J Haematol 45:381, 1980. [PMID: 6932953]
275. Matsuo T: In vitro modulation of alkaline phosphatase activity in neutrophils from patients with chronic myelogenous leukemia by monocyte-derived activity. Blood 67:492, 1986. [PMID: 3484643]
276. Tanaka KR, Valentine WN, Fredricks RE: Diseases or clinical conditions associated with low leukocyte alkaline phosphatase. N Engl J Med 262:912, 1960. [PMID: 13836802]
277. Stinson RA, McPhee J, Lewanczk R, Dinwoodie A: Neutrophil alkaline phosphatase in hypophosphatasia. N Engl J Med 312:1642, 1985. [PMID: 4000211]
278. Kamada N, Uchino H: Chronologic sequence in appearance of clinical and laboratory findings characteristic of chronic myelocytic leukemia. Blood 51:843, 1978. [PMID: 273449]
279. Denberg JA, Wilson WEC, Goodacre R, Brenenstock J: Chronic myeloid leukemia—Evidence for basophil differentiation and histamine synthesis from cultured peripheral blood cells. Br J Haematol 45:13, 1980.
280. Goh K-O, Anderson FW: Cytogenetic studies in basophilic chronic myelocytic leukemia. Arch Pathol Lab Med 103:288, 1979. [PMID: 287415]
281. Valent P, Agis H, Sperr W, et al: Diagnostic and prognostic value of new biochemical and immunohistochemical parameters in chronic myeloid leukemia. Leuk Lymphoma 49:635, 2008. [PMID: 18398724]
282. Samorapoompichit P, Kiener HP, Schernthaner G-H, et al: Detection of tryptase in cytoplasmic granules of basophils in patients with chronic myeloid leukemia and other myeloid neoplasms. Blood 98:2580, 2001. [PMID: 11588060]
283. Weil SC, Hrisinko MA: A hybrid eosinophilic-basophilic granulocyte in chronic granulocytic leukemia. Am J Clin Pathol 87:66, 1987. [PMID: 2432775]
284. Velardi A, Rambotti P, Cernetti C, et al: Monoclonal antibody defined T-cell phenotypes and phytohemagglutinin reactivity of E-rosette forming circulating lymphocytes from untreated chronic myelocyte leukemia patients. Cancer 53:913, 1984. [PMID: 6607098]
285. Dowding C, Th’ng KH, Goldman JM, Galton DAG: Increased T-lymphocyte numbers in chronic granulocytic leukemia before treatment. Exp Hematol 12:811, 1984. [PMID: 6239787]
286. Kaur J, Catovsky D, Spiers ASD, Galton DAG: Increase of T-lymphocytes in the spleen in chronic granulocytic leukaemia. Lancet 1:834, 1974. [PMID: 4132790]
287. Fujimiya Y, Bakke A, Chang WC, et al: Natural killer-cell immunodeficiency in patients with chronic myelogenous leukemia. Int J Cancer 37:639, 1986. [PMID: 3457769]
288. Fujimiya Y, Chang WC, Bakke A, et al: Natural killer cell immunodeficiency in patients with chronic myelogenous leukemia. Cancer Immunol Immunother 24:213, 1987. [PMID: 3496152]
289. Mellqvist U-H, Hansson M, Brune M, et al: Natural killer cell dysfunction and apoptosis induced by chronic myelogenous leukemia cells: Role of reactive oxygen species and regulation by histamine. Blood 96:1961, 2000. [PMID: 10961901]
290. Pierson BA, Miller JS: The role of autologous natural killer cells in chronic myelogenous leukemia. Leukemia 11:1404, 1997.
291. Mason JE, DeVita VT, Canellos GP: Thrombocytosis in chronic granulocytic leukemia: Incidence and clinical significance. Blood 44:483, 1974. [PMID: 4528824]
292. Pederson B: Kinetics and cell function, in Chronic Granulocytic Leukaemia, edited by MT Shaw, p 93. Praeger, East Sussex, UK, 1982.
293. Radhika V, Thennarasu S, Naik NR, et al: Granulocytes from chronic myeloid leukemia (CML) patients show differential response to different chemoattractants. Am J Hematol 52:155, 1996. [PMID: 8756080]
294. Kasimir-Bauer S, Ottinger H, Brittinger G, König W: Philadelphia chromosome-positive chronic myelogenous leukemia: Functional defects in circulating mature neutrophils of untreated and interferon–treated patients. Exp Hematol 22:426, 1994. [PMID: 8174673]
295. Adams T, Schultz L, Goldberg L: Platelet function abnormalities in the myeloproliferative disorders. Scand J Haematol 13:215, 1974. [PMID: 4533155]
296. Gerrard JM, Stoddard SF, Shapiro RS, et al: Platelet storage pool deficiency and prostaglandin synthesis in chronic granulocytic leukaemia. Br J Haematol 40:597, 1978. [PMID: 281970]
297. Knox WF, Bhavani M, Davson J, Geary CG: Histological classification of chronic granulocytic leukemia. Clin Lab Haematol 6:171, 1984. [PMID: 6593144]
298. Lorand-Metze I, Vassalo J, Souza CA: Histological and cytological heterogeneity of bone marrow in Philadelphia-positive chronic myelogenous leukaemia at diagnosis. Br J Haematol 67:45, 1987. [PMID: 3478078]
299. Inokuchi K, Yamaguchi H, Tarusawa M, et al: Abnormality of c-kit oncoprotein in certain patients with chronic myelogenous leukaemia—Potential clinical significance. Leukemia 16:170, 2002. [PMID: 11840282]
300. Cairoli R, Grillo G, Beghini A, et al: Chronic myelogenous leukemia with acquired c-kit activating mutation and transient bone marrow mastocytosis. Hematol J 5:273, 2004. [PMID: 15167915]
301. Agis H, Sotlar K, Valent P, Horny HP: Ph-Chromosome-positive chronic myeloid leukemia with associated bone marrow mastocytosis. Leuk Res 29:1227, 2005. [PMID: 16111540]
302. Kelsey PR, Geary CG: Sea-blue histiocytes and Gaucher’s cells in bone marrow of patients with chronic myeloid leukaemia. J Clin Pathol 41:960, 1988. [PMID: 3192753]
303. Dezmezian R, Kantarjian HM, Keating MJ, et al: The relevance of reticulin stain-measured fibrosis at diagnosis in chronic myelogenous leukemia. Cancer 59:1739, 1987.
304. Ghosh K, Varma N, Varma S, Dash S: Cellular composition and reticulin fibrosis in chronic myeloid leukaemia. Indian J Cancer 25:128, 1988. [PMID: 3224984]
305. Buhr T, Choritz H, Georgü A: The impact of megakaryocyte proliferation for the evolution of myelofibrosis. Virchows Arch 420:473, 1992. [PMID: 1609507]
306. Korkolopoulou P, Viniou N, Kavantzas N, et al: Clinicopathologic correlations of bone marrow angiogenesis in chronic myeloid leukemia: A morphometric study. Leukemia 17:89, 2003. [PMID: 12529665]
307. Aguayo A, Kantarjian H, Manshouri T, et al: Angiogenesis in acute and chronic leukemias and myelodysplastic syndromes. Blood 96:2240, 2000. [PMID: 10979972]
308. Zhelyazkova AG, Tonchev AB, Kolova P, et al: Prognostic significance of hepatocyte growth factor and microvessel bone marrow density in patients with chronic myeloid leukaemia. Scand J Clin Lab Invest 18:1, 2008.
309. Rumpel M, Friedrich T, Deininger MWN: Imatinib normalizes bone marrow vascularity in patients with chronic myeloid leukemia in first chronic phase. Blood 101:4641, 2003. [PMID: 12756164]
310. Adams SP, Sahota SS, Mijovic A, et al: Frequent expression of HAGE in presentation chronic myeloid leukemias. Leukemia 16:2238, 2002. [PMID: 12399967]
311. Udomsakdi C, Eaves CJ, Lansdorp PM, Eaves AC: Phenotypic heterogeneity of primitive leukemic hematopoietic cells in patients with chronic myeloid leukemia. Blood 80:2522, 1992. [PMID: 1384787]
312. Huret JL: Complex translocations, simple variant translocation and Ph-negative cases in chronic myelogenous leukaemia. Hum Genet 85:565, 1990. [PMID: 2227945]
313. Sakurai M, Sandberg AA: The chromosomes and causation of human cancer and leukemia: XVIII. The missing Y in acute myeloblastic leukemia (AML) and Ph1-positive chronic myelocytic leukemia. Cancer 38:762, 1976. [PMID: 1067892]
314. Berger R, Bernheim A: Y chromosome loss in leukemias. Cancer Genet Cytogenet 1:1, 1979.
315. Ishihara T, Sasaki M, Oshimura M, et al: A summary of cytogenetic studies on 534 cases of chronic myelogenous leukemia in Japan. Cancer Genet Cytogenet 9:81, 1983. [PMID: 6573228]
316. Mitelman F: Catalogue of chromosomal aberrations in cancer. Cytogenet Cell Genet 36:9, 1983.
317. Heim S, Billstrom R, Kristoffersson U, et al: Variant Ph translocations in chronic myeloid leukemia. Cancer Genet Cytogenet 18:215, 1985. [PMID: 3863698]
318. Bartram CR, Anger B, Carbonell F, Kleihauer E: Involvement of chromosome 9 in variant Ph1 translocation. Leuk Res 9:1133, 1985. [PMID: 3865031]
319. Morris CM, Rosman I, Archer SA, et al: A cytogenetic and molecular analysis of five variant Philadelphia translocations in chronic myeloid leukemia. Cancer Genet Cytogenet 35:179, 1988. [PMID: 3180020]
320. Teyssier JR, Bartram CR, DeVille J, et al: C-abl oncogene and chromosome 22 “bcr” juxtaposition in chronic myelogenous leukemia. N Engl J Med 312:1393, 1985. [PMID: 3857461]
321. Hagemeijer A, Bartram CR, Smith EME, et al: Is the chromosomal region 9q34 always involved in variants of the Ph1 translocation? Cancer Genet Cytogenet 13:1, 1984. [PMID: 6467178]
322. DeBraikeleer M, Chiu H-K, Fiser J, Gardner HA: A further case of Philadelphia chromosome-positive chronic myeloid leukemia with t(3;9;22). Cancer Genet Cytogenet 35:279, 1988.
323. Latoge-Pochitaloff-Huvalé M, Sainty D, Adriaansen HJ, et al: Translocation (3;21) in Philadelphia positive chronic myeloid leukemia. Leukemia 3:554, 1989.
324. Thompson PW, Whittaker JA: Translocation 3;21 in Philadelphia chromosome positive chronic myeloid leukemia at diagnosis. Cancer 39:143, 1989. [PMID: 2752367]
325. Engel E, McGee BJ, Flexner JM, et al: Philadelphia chromosome (Ph1) translocation in an apparently Ph1 negative, minus G22, case of chronic myeloid leukemia. N Engl J Med 291:154, 1974. [PMID: 4525414]
326. Verma RS, Dosik H: “Masked” Ph1 chromosome in chronic myelogenous leukaemia (CML). Blut 50:129, 1985. [PMID: 3856457]
327. Hagemeijer A, de Klein A, Godde-Salz E, et al: Translocation of c-abl to “masked” Ph in chronic myeloid leukemia. Cancer Genet Cytogenet 18:95, 1985. [PMID: 3863697]
328. Melo JV: The diversity of BCR-ABL fusion proteins and their relationship to leukemic phenotype. Blood 88:2375, 1996. [PMID: 8839828]
329. O’Brien S, Thall PR, Siciliano MJ: Cytogenetics of chronic myeloid leukemia. Baillieres Clin Haematol 10:259, 1997. [PMID: 9376663]
330. Bartram CR, Carbonell F: Bcr rearrangement in Ph-negative CML. Cancer Genet Cytogenet 21:183, 1986. [PMID: 3456260]
331. Bartram CR: Rearrangement of bcr and c-abl sequences in Ph-positive acute leukemias and Ph-negative CML—An update. Curr Stud Hematol Blood Transfus 31:160, 1987. [PMID: 3481751]
332. Ganesan TS, Rassool F, Guo A-P, et al: Rearrangement of the bcr gene in Philadelphia-chromosome negative chronic myeloid leukemia. Curr Stud Hematol Blood Transfus 31:153, 1987. [PMID: 3481750]
333. Wiedemann LM, Karhi K, Chan LC: Similar molecular alterations occur in related leukemias with and without the Philadelphia chromosome. Curr Stud Hematol Blood Transfus 31:149, 1987. [PMID: 3127279]
334. Benn P, Loper L, Eisenberg A, et al: Utility of molecular genetic analysis of bcr rearrangement in the diagnosis of chronic myeloid leukemia. Cancer Genet Cytogenet 29:1, 1987. [PMID: 2822224]
335. Epner DE, Koeffler AP: Molecular genetic advances in chronic myelogenous leukemia. Ann Intern Med 113:3, 1990. [PMID: 2073245]
336. Dubé I, Dixon J, Beckett T, et al: Location of breakpoints within the major breakpoint cluster region (bcr) in 33 patients with bcr rearrangement-positive chronic myeloid leukemia with complex or absent Philadelphia chromosomes. Genes Chromosomes Cancer 1:106, 1989. [PMID: 20074029]
337. Morris C, Heisterkamp N, Kennedy MA, et al: Ph-negative chronic myeloid leukemia: Molecular analysis of ABL insertion into M-BCR on chromosome 22. Blood 76:1812, 1990. [PMID: 2171702]
338. Blennerhassett GT, Furth ME, Anderson A, et al: Clinical evaluation of DNA probe assay for the Philadelphia (Ph1) translocation in chronic myelogenous leukemia. Leukemia 2:648, 1988. [PMID: 3050293]
339. Lange W, Snyder DS, Castro R, et al: Detection by enzymatic amplification of bcr-abl mRNA in peripheral blood and bone marrow cells of patients with chronic myelogenous leukemia. Blood 73:1735, 1989. [PMID: 2496773]
340. Dhingra K, Talpaz M, Riggs MC, et al: Hybridization protection assay: A rapid, sensitive, and specific method for detection of Philadelphia chromosome-positive leukemias. Blood 77:238, 1991. [PMID: 1985690]
341. Stock W, Westbrook CA, Peterson B, et al: Value of molcular monitoring during the treatment of chronic myeloid leukemia: A Cancer and Leukemia Group B study. J Clin Oncol 15:26, 1997. [PMID: 8996121]
342. Frenoy N, Chabli A, Sol D, et al: Application of a new protocol for nested PCR to the detection of minimal residual bcr/abl transcripts. Leukemia 8:1411, 1994. [PMID: 7520102]
343. Melo JV, Yan XH, Diamond J, et al: Reverse transcription/polymerase chain reaction (RT/PCR) amplification of very small numbers of transcripts: The risk in misinterpreting negative results. Leukemia 10:1217, 1996. [PMID: 8684005]
344. Lin F, Chase A, Bunget J, et al: Correlation between the proportion of Philadelphia chromosome-positive metaphase cells and levels of BCRABL mRNA in chronic myeloid leukaemia. Genes Chromosomes Cancer 13:110, 1995. [PMID: 7542905]
345. VanDenderen J, Hermans A, Meeuwsen T, et al: Antibody recognition of the tumor-specific bcr-abl joining region in chronic myeloid leukemia. J Exp Med 169:87, 1989.
346. Hagemeyer A, vanderPlas DC, Solkarman D, et al: The Philadelphia translocation in CML and ALL: Recent investigations, new detection methods. Nouv Rev Fr Hematol 32:83, 1990.
347. Maxwell SA, Kurzrock R, Parsons SJ, et al: Analysis of p210 bcr-abl tyrosine protein kinase activity in various subtypes of Philadelphia chromosome-positive cells from chronic myelogenous leukemia patients. Cancer Res 47:1731, 1987. [PMID: 2434223]
348. Guo JQ, Lian JY, Xian YM, et al: BCR-ABL protein expression in peripheral blood cells of chronic myelogenous leukemia patients undergoing therapy. Blood 83:3629, 1994. [PMID: 8204887]
349. Dewald GW, Schad CR, Christensen ER, et al: The application of in situ fluorescent hybridization to detect M bcr/abl fusion in variant Ph chromosomes in CML and ALL. Cancer Genet Cytogenet 71:7, 1993. [PMID: 8275455]
350. Cox MC, Maffei L, Buffolino S, et al: A comparative analysis of FISH, RT-PCR, and cytogenetics for the diagnosis of bcr-abl-positive leukemias. Am J Clin Pathol 109:24, 1998. [PMID: 9426514]
351. Sinclair PB, Green AR, Grace C, Nacheva EP: Improved sensitivity of BCR-ABL detection: A triple-probe three-color fluorescence in situ hybridization system. Blood 90:1395, 1997. [PMID: 9269756]
352. Acar H, Stewart J, Boyd E, Connor MJ: Identification of variant translocations in chronic myeloid leukemia by fluorescence in situ hybridization. Cancer Genet Cytogenet 93:115, 1997. [PMID: 9078295]
353. Schoch C, Schnittger S, Bursch S, et al: Comparison of chromosome banding analysis, interphase- and hypermetaphase-FISH, qualitative and quantitative PCR for diagnosis and for follow-up in chronic myeloid leukemia: A study of 350 cases. Leukemia 16:53, 2002. [PMID: 11840263]
354. Yanagi M, Shinjo K, Takeshita A, et al: Simple and reliably sensitive diagnosis and monitoring of Philadelphia chromosome-positive cells in chronic myeloid leukemia by interphase fluorescence in situ hybridization of peripheral blood cells. Leukemia 13:542, 1999. [PMID: 10214860]
355. Werner M, Ewig M, Nasarek A, et al: Value of fluorescence in situ hybridization for detecting the bcr/abl gene fusion in interphase cells of routine bone marrow specimens. Diagn Mol Pathol 6:282, 1997. [PMID: 9458387]
356. Chase A, Grand F, Zhang JG, et al: Factors influencing the false positive and negative rates of BCR-ABL fluorescence in situ hybridization. Genes Chromosomes Cancer 18:246, 1997. [PMID: 9087564]
357. Pelz AF, Kroning H, Franke A, Wieacker P: High reliability and sensitivity of the BCR/ABL1 D-FISH test for the detection of BCR/ABL rearrangements. Ann Hematol 81:147, 2002. [PMID: 11904740]
358. Hochhaus A, Reiter A, Skladny H, et al: Molecular monitoring of residual disease in chronic myelogenous leukemia patients after therapy. Recent Results Cancer Res 144:36, 1998. [PMID: 9304705]
359. Wells SJ, Phillips CN, Winton EF, Farhi DC: Reverse transcriptase polymerase chain reaction for bcr-abl fusion in chronic myelogenous leukemia. Am J Clin Pathol 105:756, 1996. [PMID: 8659451]
360. Cox MC, Maffei L, Buffolino S, et al: A comparative analysis of FISH, RT-PCR, and cytogenetics for the diagnosis of bcr-abl-positive leukemias. Am J Clin Pathol 109:24, 1998. [PMID: 9426514]
361. Krackoff IH: Studies of uric acid biosynthesis in the chronic leukemias. Arthritis Rheum 8:772, 1965.
362. Vogler WR, Bain JA, Huguley CM Jr, et al: Metabolic and therapeutic effects of allopurinol in patients with leukemia and gout. Am J Med 40:548, 1966.
363. Zittoun J, Marquet J, Zittoun R: The intracellular content of the three cobalamins at various stages of normal and leukaemic myeloid cell development. Br J Haematol 31:299, 1975. [PMID: 1201243]
364. Zittoun J, Zittoun R, Marquet J, Sultan C: The three transcobalamins in myeloproliferative disorders and acute leukemia. Br J Haematol 31:287, 1975. [PMID: 1059479]
365. Rosner F, Schreiber ZA: Serum vitamin B12 and vitamin B12 binding capacity in chronic myelogenous leukemia and other disorders. Am J Med Sci 263:473, 1972. [PMID: 4505888]
366. Sternman U-H: Intrinsic factor and the B12 binding proteins. Clin Haematol 5:473, 1976.
367. Corcino JJ, Zalusky R, Greenberg M, Herbert V: Coexistence of pernicious anaemia and chronic myeloid leukaemia: An experiment of nature involving vitamin B12 metabolism. Br J Haematol 20:511, 1971. [PMID: 4103047]
368. Agis H, Sperr WR, Herndlhofer S, et al: Clinical and prognostic significance of histamine monitoring in patients with CML during treatment with imatinib (STI571). Ann Oncol 18:1834, 2007. [PMID: 17804468]
369. Youman JD, Taddeini L, Cooper T: Histamine excess symptoms in basophilic chronic granulocytic leukemia. Arch Intern Med 131:560, 1973. [PMID: 4121666]
370. Rosenthal S, Schwartz JH, Canellos GP: Basophilic chronic granulocytic leukemia with hyperhistaminemia. Br J Haematol 36:367, 1977. [PMID: 268209]
371. Gomez GA, Sokal JE, Walsh D: Prognostic features at diagnosis of chronic myelocytic leukemia. Cancer 47:2470, 1981. [PMID: 6944141]
372. Bellevue R, Dosik H, Spergel G, Gussoff BD: Pseudohyperkalemia and extreme leukocytosis. J Lab Clin Med 85:660, 1975. [PMID: 1120932]
373. Ballard HS, Marcus AJ: Hypercalcemia in chronic myelogenous leukemia. N Engl J Med 282:663, 1970. [PMID: 5264635]
374. Evans JJ, Bozdech MJ: Hypokalemia in nonblastic chronic myelogenous leukemia. Arch Intern Med 141:786, 1981. [PMID: 6940517]
375. Perillie PE, Finch SC: Muramidase studies in Philadelphia-chromosome-positive and chromosome-negative chronic granulocytic leukemia. N Engl J Med 283:456, 1970. [PMID: 5270611]
376. Gilbert HS, Ginsberg H: Hypocholesterolemia as a manifestation of disease activity in chronic myeloid leukemia. Cancer 51:1428, 1983. [PMID: 6572087]
377. Muller CP, Wagner AN, Maucher C, Steinke B: Hypocholesterolemia, an unfavorable feature of prognostic value in chronic myeloid leukemia. Eur J Haematol 43:235, 1989. [PMID: 2806478]
378. Musolino C, Alonci A, Bellomo G, et al: Levels of soluble angiogenin in chronic myeloid malignancies. Eur J Haematol 72:416, 2004. [PMID: 15128420]
379. Calabro L, Fonsatti E, Bellomo G, et al: Differential levels of soluble endoglin (CD105) in myeloid malignancies. J Cell Physiol 194:171, 2003. [PMID: 12494455]
380. Morris CM, Fitzgerald PH, Hollings PE, et al: Essential thrombocythemia and the Philadelphia chromosome. Br J Haematol 70:13, 1988. [PMID: 3179224]
381. Stoll DB, Peterson P, Exten R, et al: Clinical presentation and natural history of patients with essential thrombocythemia and the Philadelphia chromosome. Am J Hematol 27:77, 1988. [PMID: 3422539]
382. Sessarego M, Defferrari R, Dejana AM, et al: Cytogenetic analysis in essential thrombocythemia at diagnosis and at transformation. Cancer Genet Cytogenet 43:57, 1989. [PMID: 2790773]
383. Pajor L, Kereskai L, Zsdral K, et al: Philadelphia chromosome and/or bcr-abl mRNA-positive primary thrombocytosis: Morphometric evidence for the transition from essential thrombocythemia to chronic myeloid leukaemia type myeloproliferation. Histopathology 42:53, 2003. [PMID: 12493025]
384. Blickstein D, Aviram A, Luboshitz J, et al: BCR-ABL transcripts in bone marrow aspirates of Philadelphia-negative essential thrombocythemia patients: Clinical presentation. Blood 90:2768, 1997. [PMID: 9326244]
385. Cervantes F, Colomer D, Vives-Corrons JL, et al: Chronic myeloid leukemia of thrombocythemic onset: A CML subtype with distinct hematological and molecular features. Leukemia 10:1241, 1996. [PMID: 8684009]
386. Martiat P, Ifrah N, Rassool F, et al: Molecular analysis of Philadelphia positive essential thrombocythemia. Leukemia 3:563, 1989. [PMID: 2747291]
387. Michiels JJ, Berneman Z, Schroyens W, et al: Philadelphia (Ph) chromosome–positive thrombocythemia without features of chronic myeloid leukemia in peripheral blood; natural history and diagnostic differentiation from Ph-negative essential thrombocythemia. Ann Hematol 83:504, 2004. [PMID: 15164229]
388. Blickstein D, Aviram A, Luboshitz J, et al: BCR-ABC transcripts in bone marrow aspirates of Philadelphia-negative essential thrombocythemia patients: Clinical presentation. Blood 90:2768, 1997. [PMID: 9326244]
389. Pajor L, Kereskai L, Zsdral K, et al: Philadelphia chromosome and/or bcr-abl mRNA positive primary thrombocytosis: Morphometric evidence for the transition from essential thrombocythaemia to chronic myeloid leukaemia type of myeloproliferation. Histopathology 42:53, 2003. [PMID: 12493025]
390. Damaj G, delabesse E, Le Bihan C, et al: Typical essential thrombocythaemia does not express bcr-abelson fusion transcript. Br J Haematol 116:812, 2002. [PMID: 11886385]
391. Hsu H-C, Tan L-Y, Au L-C, et al: Detection of bcr-abl gene expression at a low level in blood cells of some patients with essential thrombocythemia. J Lab Clin Med 143:125, 2004. [PMID: 14966468]
392. Paietta E, Rosen N, Roberts M, et al: Philadelphia chromosome positive essential thrombocythemia evolving into lymphoid blast crisis. Cancer Genet Cytogenet 25:227, 1987. [PMID: 3470113]
393. Michiels JJ, Prins ME, Hagermeijer A, et al: Philadelphia chromosome-positive thrombocythemia and megakaryoblast leukemia. Am J Clin Pathol 88:645, 1987. [PMID: 3479003]
394. Kwong YL, Chiu EK, Liang RH, et al: Essential thrombocythemia with BCR/ABL rearrangement. Cancer Genet Cytogenet 89:74, 1996. [PMID: 8689617]
395. Marasca R, Luppi M, Zucchini P, et al: Might essential thrombocythemia carry Ph anomaly? Blood 91:3084, 1998. [PMID: 9531623]
396. Sanadi I, Yamamoto S, Ogata M, et al: Detection of the Philadelphia chromosome in chronic neutrophilic leukemia. Jpn J Clin Oncol 15:553, 1985.
397. Christopoulus C, Kottoris K, Mikraki V, Anevlavis E: Presence of bcr/abl rearrangement in a patient with chronic neutrophilic leukaemia. J Clin Pathol 49:1013, 1996.
398. Pane F, Frigeri F, Sindina M, et al: Neutrophilic-chronic myeloid leukemia: A distinct disease with a specific molecular marker (BCR/ABL with C3/A2 junction). Blood 88:2410, 1996. [PMID: 8839830]
399. Verstovsek S, Lin H, Kantarjian H, et al: Neutrophilic-chronic myeloid leukemia: Low levels of p 230 BCR/ABL mRNA and undetectable BCR/ABL protein may predict an indolent course. Cancer 94:2416, 2002. [PMID: 12015767]
400. Ohsaka A, Shiina S, Kobayashi M, et al: Philadelphia chromosome-positive chronic myeloid leukemia expressing p190(BCR-ABL). Intern Med 41:1183, 2002. [PMID: 12521212]
401. Barnes DJ, Melo JV: Cytogenetic and molecular genetic aspects of chronic myeloid leukaemia. Acta Haematol 108:180, 2002. [PMID: 12432215]
402. Knowles DM: Thymoma and chronic myelogenous leukemia. Cancer 38:414, 1976.
403. Vannier JP, Bizet M, Bastard C, et al: Simultaneous occurrence of a T-cell lymphoma and a chronic myelogenous leukemia with an unusual karyotype. Leuk Res 8:647, 1984. [PMID: 6590932]
404. Djulbegovi B, Hadley T, Yen F: Occurrence of high-grade T-cell lymphoma in a patient with Philadelphia chromosome-negative chronic myelogenous leukemia with breakpoint cluster region rearrangement. Am J Hematol 36:63, 1991.
405. Tittley P, Trempe JM, van der Jagt R, et al: Occurrence of T-cell lymphoma in a patient with Philadelphia chromosome-positive chronic myelogenous leukemia with rearrangements of BCR and TCR- genes in the lymph nodes. Am J Hematol 42:229, 1993. [PMID: 8438886]
406. Hornstein P, Nordenson I, Wahlin A: Philadelphia chromosome negative acute lymphoblastic leukemia preceding Philadelphia positive chronic myelogenous leukemia. Cancer Genet Cytogenet 39:147, 1989.
407. Ichinohasama R, Miura I, Takahashi N, et al: Ph-negative non-Hodgkin’s lymphoma occurring in chronic phase of Ph-positive chronic myelogenous leukemia is defined as a genetically different neoplasm from extramedullary localized blast crisis: Report of two cases and review of the literature. Leukemia 14:169, 2000. [PMID: 10637493]
408. Rodler E, Welborn J, Hatcher S, et al: Blastic mantle cell lymphoma developing concurrently in a patient with chronic myelogenous leukemia and a review of the literature. Am J Hematol 75:231, 2004. [PMID: 15054816]
409. Naparstek Y, Zlotnick A, Polliack A: Coexistent chronic myeloid leukemia and IgA monoclonal gammopathy: Report of a case and review of the literature. Am J Med Sci 292:111, 1980.
410. Shoenfeld Y, Berliner S, Ayalone A, et al: Monoclonal gammopathy in patients with chronic and acute myeloid leukemia. Cancer 54:280, 1984. [PMID: 6426774]
411. Tanaka M, Kimura R, Matsutani A, et al: Coexistence of chronic myelogenous leukemia and multiple myeloma. Acta Haematol 99:221, 1998. [PMID: 9644301]
412. Schwartzmeier JD, Shehata M, Ackermann J, et al: Simultaneous occurrence of chronic myeloid leukemia and multiple myeloma: Evaluation by FISH analysis and in vitro expansion of bone marrow cells. Leukemia 17:1426, 2003.
413. Nitta M, Tsuboi K, Yamashita S, et al: Multiple myeloma preceding development of chronic myelogenous leukemia. Int J Hematol 69:170, 1999. [PMID: 10222655]
414. Vitali C, Bombardieri S, Spremolla G: Chronic myeloid leukemia in Waldenström’s macroglobulinemia. Arch Intern Med 141:1349, 1981. [PMID: 6791603]
415. Whang-Peng J, Gralnick HR, Johnson RE, et al: Chronic granulocytic leukemia (CGL) during the course of chronic lymphocytic leukemia (CLL): Correlation of blood, marrow, and spleen morphology and cytogenetics. Blood 43:333, 1974. [PMID: 4521368]
416. Schrieber ZA, Axelrod MR, Abebe LS: Coexistence of chronic myelogenous leukemia and chronic lymphocytic leukemia. Cancer 54:697, 1984.
417. Specchia G, Buquicchio C, Albano F, et al: Non-treatment-related chronic myeloid leukemia as a second malignancy. Leuk Res 28:115, 2004. [PMID: 14654074]
418. Esteve J, Cervantes F, Rives S, et al: Simultaneous occurrence of B-cell chronic lymphocytic leukemia and chronic myeloid leukemia with further evolution to lymphoid blast status. Haematologica 82:596, 1997. [PMID: 9407730]
419. Leoni F, Ferrini PR, Castoldi GL, et al: Simultaneous occurrence of chronic granulocytic leukemia and chronic lymphoid leukemia. Haematologica 72:253, 1987. [PMID: 3114094]
420. Faguet GB, Little T, Agee JF, Garver FA: Chronic lymphatic leukemia evolving into chronic myelocytic leukemia. Cancer 52:1647, 1983. [PMID: 6577937]
421. Crescenzi B, Sacchi S, Marasca R, et al: Distinct genomic events in the myeloid and lymphoid lineages in simultaneous presentation of chronic myeloid leukemia and B-chronic lymphocytic leukemia. Leukemia 16:955, 2002. [PMID: 11986962]
422. Mansat-De Mas V, Regal-Huguet F, Cassar G, et al: Chronic myeloid leukemia associated with B-cell chronic lymphocytic leukemia: Evidence of two separate clones as shown by combined cell-sorting and fluorescence in situ hybridization. Leuk Lymphoma 44:867, 2003.
423. Lucia E, Martino B, Mammi C, et al: The incidence of JAK2 V617F mutation in bcr/abl-negative chronic myeloproliferative disorders: Assessment by two different detection methods. Leuk Lymphoma 18:1, 2008.
424. Jantunen E, Nousiainen T: Ph-positive chronic myelogenous leukemia evolving after polycythemia vera. Am J Hematol 37:212, 1991. [PMID: 1858777]
425. Hoppen EC, Lewis JP: Polycythemia rubra vera progressing to Ph-positive chronic myelogenous leukemia. Ann Intern Med 83:820, 1975.
426. Haq AU: Transformation of polycythemia vera to Ph-positive chronic myelogenous leukemia. Am J Hematol 356:110, 1990.
427. Roth AD, Oral A, Przepiorka D, et al: Chronic myelogenous leukemia and acute lymphoblastic leukemia occurring in the course of polycythemia vera. Am J Hematol 43:123, 1993. [PMID: 8342539]
428. Foviester RH, Louro JM: Philadelphia chromosome abnormality in angiogenic myeloid metaplasia. Ann Intern Med 64:622, 1966.
429. Nowell PC, Kant JA, Finan JB, et al: Marrow fibrosis associated with a Philadelphia chromosome. Cancer Genet Cytogenet 59:89, 1992. [PMID: 1372840]
430. Roth DG, Richman CM, Rowley JD: Chronic myelodysplastic syndrome (preleukemia) with the Philadelphia chromosome. Blood 56:262, 1980. [PMID: 6930980]
431. Berrebi A, Bruck R, Shtalrid M, Chemke J: Philadelphia chromosome in idiopathic acquired sideroblastic anemia. Acta Haematol 72:343, 1984. [PMID: 6441415]
432. Suzan F, Terré C, Garcia I, et al: Three cases of typical aplastic anaemia associated with a Philadelphia chromosome. Br J Haematol 112:385, 2001. [PMID: 11167835]
433. Sica S, Chiusolo P, Zollino M, et al: The association of severe aplastic anaemia with the Philadelphia chromosome and the bcr/abl transcript. Br J Haematol 114:961, 2001. [PMID: 11564100]
434. Hande K: Hyperuricemia, uric acid nephropathy and the tumor lysis syndrome, in Renal Complications of Neoplasia, edited by TD McKinney, p 134. Praeger, New York, 1986.
435. Navolanic PM, Pui CH, Larson RA, et al: ElitekTM-rasburicase: An effective means to prevent and treat hyperuricemia associated with tumor lysis syndrome, a Meeting Report, Dallas, TX, January, 2002. Leukemia 17:499, 2003. [PMID: 12646938]
436. Jeha S, Pui CH: Recombinant urate oxidase (rasburicase) in the prophylaxis and treatment of tumor lysis syndrome. Contrib Nephrol 147:69, 2005. [PMID: 15604607]
437. Liu CY, Sims-McCallum RP, Schiffer CA: A single dose of rasburicase is sufficient for the treatment of hyperuricemia in patients receiving chemotherapy. Leuk Res 29:463, 2005. [PMID: 15725482]
438. Arnold TM, Reuter JP, Delman BS, Shanholtz CB: Use of single-dose rasburicase in an obese female. Ann Pharmacother 38:1428, 2004. [PMID: 15292497]
439. Bazatbashi MS, Smith MR, Karanes C, et al: Successful management of Ph chromosome chronic myelogenous leukemia with leukapheresis during pregnancy. Am J Hematol 38:235, 1991.
440. Strobl FJ, Voelkerding KY, Smith EP: Management of chronic myeloid leukemia during pregnancy with leukapheresis. J Clin Apher 14:42, 1999. [PMID: 10355663]
441. Kennedy BJ: The evolution of hydroxyurea therapy in chronic myelogenous leukemia. Semin Oncol 19(Suppl 9):21, 1992.
442. Tsimberidou AM, Colburn DE, Welch MA, et al: Anagrelide and imatinib mesylate combination therapy in patients with chronic myeloproliferative disorders. Cancer Chemother Pharmacol 52:229, 2003. [PMID: 12783207]
443. Baccarani M, Saglio G, Goldman J, et al: Evolving concepts in the management of chronic myeloid leukemia. Recommendations from an expert panel of behalf of the European LeukemiaNet. Blood 108:1809, 2006. [PMID: 16709930]
444. NCCN Practice Guidelines in Oncology. v.3.2008. Available at: www.nccn.org/professionsals/physician_gls/.
445. Kantarjian HM, Talpaz M, O’Brien S, et al: Dose escalation of imatinib mesylate can overcome resistance to standard-dose therapy in patients with chronic myelogenous leukemia. Blood 101:473, 2003. [PMID: 12393385]
446. Marin D, Goldman JM, Olavarria E, Apperley JF: Transient benefit only from increasing the imatinib dose in CML patients who do not achieve complete cytogenetic remissions on conventional doses. Blood 102:2702, 2003. [PMID: 14504074]
447. Zonder JA, Pemberton P, Brandt H, et al: The effect of dose increase of imatinib mesylate in patients with chronic or accelerated phase chronic myelogenous leukemia with inadequate hematologic or cytogenetic response to initial treatment. Clin Cancer Res 9:2092, 2003. [PMID: 12796373]
448. Kantarjian H, Talpaz M, O’Brien S, et al: High-dose imatinib mesylate therapy in newly diagnosed Philadelphia chromosome-positive chronic phase chronic myeloid leukemia. Blood 103:2873, 2004. [PMID: 15070658]
449. Kanda Y, Okamoto S, Tauchi T, et al: Multicenter prospective trial evaluating the tolerability of imatinib for Japanese patients with chronic myelogenous leukemia in the chronic phase: Does body weight matter? Am J Hematol 83:835, 2008. [PMID: 18785644]
450. Kobayashi S, Kimura F, Kobayashi A, et al: Efficacy of low-dose imatinib in chronic-phase chronic myelogenous leukemia patients. Ann Hematol 88:311, 2009. [PMID: 18704417]
451. Atallah E, Cortes J: Optimal initial therapy for patients with newly diagnosed chronic myeloid leukemia in chronic phase. Curr Opin Hematol 14:138, 2007. [PMID: 17255791]
452. Branford S, Seymour JF, Grigg A, et al: BCR-ABL messenger RNA levels continue to decline in patients with chronic phase chronic myeloid leukemia treated with imatinib for more than 5 years and approximately half of all first-line treated patients have stable undetectable BCR-ABL using strict sensitivity criteria. Clin Cancer Res 13:7080, 2007. [PMID: 18056186]
453. de Lavallade H, Apperley JF, Khorashad JS, et al: Imatinib for newly diagnosed patients with chronic myeloid leukemia: Incidence of sustained responses in an intention-to-treat analysis. J Clin Oncol 26:3358, 2008.
454. El-Zimaity MM, Kantarjian H, Talpaz M, et al: Results of imatinib mesylate therapy in chronic myelogenous leukaemia with variant Philadelphia chromosome. Br J Haematol 125:187, 2004. [PMID: 15059141]
455. Synder DS, McMahon R, Cohen SR, Slovak ML: Chronic myeloid leukemia with an e13a3 BCR-ABL fusion: Benign course responsive to imatinib with an RT-PCR advisory. Am J Hematol 75:92, 2004.
456. de Lemos JA, de Oliveira CM, Scerni AC, et al: Differential molecular response of the transcripts B2A2 and B3A2 to imatinib mesylate in chronic myeloid leukemia. Genet Mol Res 4:803, 2005.
457. Agirre X, Román-Gómez J, Vázquez I, et al: Coexistence of different clonal populations harboring the b3a2 (p210) and e1a2 (p190) BCR-ABL1 fusion transcripts in chronic myelogenous leukemia resistant to imatinib. Cancer Genet Cytogenet 160:22, 2005. [PMID: 15949566]
458. Champagne MA, Capdeville R, Krailo M, et al: Imatinib mesylate (STI571) for treatment of children with Philadelphia chromosome-positive leukemia: Results from a Children’s Oncology Group phase I study. Blood 104:2655, 2004. [PMID: 15231574]
459. Cortes J, Talpaz M, O’Brien S, et al: Effects of age on prognosis with imatinib mesylate therapy for patients with Philadelphia chromosome-positive chronic myelogenous leukemia. Cancer 98:1105, 2003. [PMID: 12973833]
460. Latagliata R, Breccia M, Carmosino I, et al: Elderly patients with Ph+ chronic myelogenous leukemia (CML): Results of imatinib mesylate treatment. Leuk Res 29:287, 2005. [PMID: 15661264]
461. Guilhot F: Indications for imatinib mesylate therapy and clinical management. Oncologist 9:271, 2004. [PMID: 15169982]
462. Marin D, Marktel S, Foot N, et al: Granulocyte colony-stimulating factor reverses cytopenia and may permit cytogenetic responses in patients with chronic myeloid leukemia treated with imatinib mesylate. Haematologica 88:227, 2003. [PMID: 12604417]
463. Quintas-Cardama A, Kantarjian H, O’Brien S, et al: Granulocyte-colony-stimulating factor (filgrastim) may overcome imatinib-induced neutropenia in patients with chronic-phase chronic myelogenous leukemia. Cancer 100:2592, 2004. [PMID: 15197801]
464. van Deventer HW, Hall MD, Orlowski RZ, et al: Clinical course of thrombocytopenia in patients treated with imatinib mesylate for accelerated phase chronic myelogenous leukemia. Am J Hematol 71:184, 2002.
465. Sneed TB, Kantarjian HM, Talpaz M, et al: The significance of myelosuppression during therapy with imatinib mesylate in patients with chronic myelogenous leukemia in chronic phase. Cancer 100:116, 2004. [PMID: 14692031]
466. Lokeshwar N, Kumar L, Kumari M: Severe bone marrow aplasia following imatinib mesylate in a patient with chronic myelogenous leukemia. Leuk Lymphoma 46:781, 2005. [PMID: 16019519]
467. Hensley ML, Ford JM: Imatinib treatment: Specific issues related to safety, fertility, and pregnancy. Semin Hematol 40:21, 2003. [PMID: 12783371]
468. Ferrero D, Pogliani EM, Rege-Cambrin G, et al: Corticosteroids can reverse severe imatinib-induced hepatotoxicity. Haematologica 91(6 Suppl):ECR27, 2006.
469. Cross TJ, Bagot C, Portmann B, et al: Imatinib mesylate as a cause of acute liver failure. Am J Hematol 83:189, 2006.
470. Esmaeli B, Prieto VG, Butler CE, et al: Severe periorbital edema secondary to STI571 (Gleevec). Cancer 95:881, 2002. [PMID: 12209733]
471. Elliott MA, Mesa RA, Tefferi A: Adverse events after imatinib mesylate therapy. N Engl J Med 346:712, 2002. [PMID: 11870257]
472. Kusumi E, Arakawa A, Kami M, et al: Visual disturbance due to retinal edema as a complication of imatinib. Leukemia 18:1138, 2004. [PMID: 15085156]
473. Mattiuzzi GN, Cortes JE, Talpaz M, et al: Development of Varicella-Zoster virus infection in patients with chronic myelogenous leukemia treated with imatinib mesylate. Clin Cancer Res 9:976, 2003. [PMID: 12631595]
474. Gambacorti-Passerini C, Tornaghi L, Cavagnini F, et al: Gynaecomastia in men with chronic myeloid leukaemia after imatinib. Lancet 361:1954, 2003. [PMID: 12801741]
475. Novaretti MCZ, Fonseca GHH, Conchon M, et al: First case of immune-mediated haemolytic anaemia associated with imatinib mesylate. Eur J Haematol 71:455, 2003. [PMID: 14703696]
476. Kyathari S, Chao K, Liu D, Seiter K: Severe imatinib-associated muscle edema in patients with chronic Myelogenous leukemia and marked leukocytosis. Leuk Lymphoma 49:1002, 2008. [PMID: 18452084]
477. Ostro D, Lipton J: Unusual fluid retention with imatinib therapy for chronic myeloid leukemia. Leuk Lymphoma 48:195, 2007. [PMID: 17325868]
478. Ohnishi K, Sakai F, Kudoh S, Ohno R: Twenty-seven cases of drug-induced interstitial lung disease associated with imatinib mesylate. Leukemia 20:1162, 2006. [PMID: 16598305]
479. Rajda J, Phatak PD: Reversible drug-induced interstitial pneumonitis following imatinib mesylate therapy. Am J Hematol 79:80, 2005. [PMID: 15849772]
480. Assouline S, Laneuville P, Gambacorti-Passerini C: Panniculitis during dasatinib therapy for imatinib-resistant chronic myelogenous leukemia. N Engl J Med 354:2623, 2006. [PMID: 16775249]
481. Osorio S, Noblejas AG, Durán A, Steegmann JL: Imatinib mesylate induces hypophosphatemia in patients with chronic myeloid leukemia in late chronic phase, and this effect is associated with response. Am J Hematol 82:394, 2007. [PMID: 17117416]
482. Fitter S, Dewar AL, Kostakis P, et al: Long-term imatinib therapy promotes bone formation in CML patients. Blood 111:2538, 2008. [PMID: 18042796]
483. Berman E, Nicolaides M, Maki RG, et al: Altered bone and mineral metabolism in patients receiving imatinib mesylate. N Engl J Med 354:2006, 2006. [PMID: 16687713]
484. Sanchez-Gonzalez B, Pascual-Ramirez JC, Fernandez-Abellian P, et al: Severe skin reaction to imatinib in a case of Philadelphia-positive acute lymphoblastic leukemia. Blood 101:2446, 2003. [PMID: 12609964]
485. Drummond A, Micallef-Eynaud P, Douglas WS, et al: A spectrum of skin reactions caused by the tyrosine kinase inhibitor imatinib mesylate (STI 571, Glivec). Br J Haematol 120:911, 2003. [PMID: 12614232]
486. Rule SAJ, O’Brien SG, Crossman LC: Managing cutaneous reactions to imatinib therapy. Blood 100:3434, 2002. [PMID: 12412577]
487. Nelson RP Jr, Cornetta K, Ward KE:, et al Desensitization to imatinib in patients with leukemia. Ann Allergy Asthma Immunol 97:216, 2006. [PMID: 16937754]
488. Liu D, Seiter K, Mathews T, et al: Sweet’s syndrome with CML cell infiltration of the skin in a patient with chronic-phase CML while taking imatinib mesylate. Leuk Res 28SI:S61, 2004.
489. Brazzelli V, Prestinari F, Roveda E, et al: Pityriasis rosea-like eruption during treatment with imatinib mesylate: Description of 3 cases. J Am Acad Dermatol 53(Suppl 1):S240, 2005.
490. Deguchi N, Kawamura T, Shimizu A, et al: Imatinib mesylate causes palmoplantar hyperkeratosis and nail dystrophy in three patients with chronic myeloid leukaemia. Br J Dermatol 154:1216, 2006. [PMID: 16704666]
491. Pascual JC, Matarredona J, Miralles J, et al: Oral and cutaneous lichenoid reaction secondary to imatinib: Report of two cases. Int J Dermatol 45:1471, 2006. [PMID: 17184272]
492. Etienne G, Cony-Makhoul P, Mahon FX: Imatinib mesylate and gray hair. N Engl J Med 346:645, 2002.
493. Tjao AS, Kantarjian H, Cortes J, et al: Imatinib mesylate causes hypopigmentation in the skin. Cancer 98:2483, 2003.
494. Legros L, Cassuto JP, Ortonne JP: Imatinib mesilate (Glivec): A systemic depigmenting agent for extensive vitiligo? Br J Dermatol 153:691, 2005. [PMID: 16120179]
495. Beham-Schmid C, Apfelbeck U, Sill H, et al: Treatment of chronic myelogenous leukemia with the tyrosine kinase inhibitor STI571 results in marked regression of bone marrow fibrosis. Blood 99:381, 2002. [PMID: 11756197]
496. Kantarjian HM, Bueso-Ramos CE, Talpaz M, et al: The degree of bone marrow fibrosis in chronic myelogenous leukemia is not a prognostic factor with imatinib mesylate therapy. Leuk Lymphoma 46:993, 2005. [PMID: 16019549]
497. Buesche G, Ganser A, Schlegelberger B, et al: Marrow fibrosis and its relevance during imatinib treatment of chronic myeloid leukemia. Leukemia 21:2420, 2007. [PMID: 17805334]
498. Ebos JM, Tran J, Master Z, et al: Imatinib mesylate (STI-571) reduces the Bcr-Abl-mediated vascular endothelial growth factor secretion in chronic myelogenous leukemia. Mol Cancer Res 1:89, 2002. [PMID: 12496355]
499. Kvasnicka HM, Thiele J, Staib P, et al: Reversal of bone marrow angiogenesis in chronic myeloid leukemia following imatinib mesylate (STI571) therapy. Blood 103:3549, 2004. [PMID: 14726401]
500. Larson RA, Druker BJ, Guilhot F, et al: Imatinib pharmacokinetics and its correlation with response and safety in chronic-phase chronic myeloid leukemia: A subanalysis of the IRIS study. Blood 111:4022, 2008. [PMID: 18256322]
501. Picard S, Titier K, Etienne G, et al: Trough imatinib plasma levels are associated with both cytogenetic and molecular responses to standard-dose imatinib in chronic myeloid leukemia. Blood 109:3496, 2007. [PMID: 17192396]
502. Schmidli H, Peng B, Riviere GJ, et al: Population pharmacokinetics of imatinib mesylate in patients with chronic-phase chronic myeloid leukaemia: Results of a phase III study. Br J Clin Pharmacol 60:35, 2005. [PMID: 15963092]
503. Ozdemir E, Koc Y, Kansu E: Successful treatment of chronic myeloid leukemia with imatinib mesylate in a patient with chronic renal failure on hemodialysis. Am J Hematol 81:474, 2006. [PMID: 16680756]
504. Darkow T, Henk HJ, Thomas SK, et al: Treatment interruptions and non-adherence with imatinib and associated healthcare costs: A retrospective analysis among managed care patients with chronic myelogenous leukaemia. Pharmacoeconomics 25:481, 2007. [PMID: 17523753]
505. Cortes J, Giles F, O’Brien S, et al: Results of imatinib mesylate therapy in patients with refractory or recurrent acute myeloid leukemia, high-risk myelodysplastic syndrome, and myeloproliferative disorders. Cancer 97:2760, 2003. [PMID: 12767088]
506. Kantarjian H, Talpaz M, O’Brien S, et al: Prediction of initial cytogenetic response for subsequent major and complete cytogenetic response to imatinib mesylate therapy in patients with Philadelphia chromosome-positive chronic myelogenous leukemia. Cancer 98:1776, 2003.
507. Rousselot P, Huguet F, Rea D, et al: Imatinib mesylate discontinuation in patients with chronic myelogenous leukemia in complete molecular remission for more than 2 years. Blood 109:58, 2007. [PMID: 16973963]
508. Carella AM, Lerma E: Durable responses in chronic myeloid leukemia patients maintained with lower doses of imatinib mesylate after achieving molecular remission. Ann Hematol 86:749, 2007. [PMID: 17576558]
509. Ghanima W, Kahrs J, Dahl TG 3rd, Tjonnfjord GE: Sustained cytogenetic response after discontinuation of imatinib mesylate in a patient with chronic myeloid leukaemia. Eur J Haematol 72:441, 2004. [PMID: 15128424]
510. Cortes J, O’Brien S, Kantarjian H: Discontinuation of imatinib therapy after achieving a molecular response. Blood 104:2204, 2004. [PMID: 15377577]
511. Okabe S, Tauchi T, Ishii Y, et al: Sustained complete cytogenetic remission in a patient with chronic myeloid leukemia after discontinuation of imatinib mesylate therapy. Int J Hematol 85:173, 2007. [PMID: 17321998]
512. Merante S, Orlandi E, Bernasconi P, et al: Outcome of four patients with chronic myeloid leukemia after imatinib mesylate discontinuation. Haematologica 90:979, 2005. [PMID: 15996937]
513. Graham SM, Jorgensen HG, Allan E, et al: Primitive, quiescent, Philadelphia-positive stem cells from patients with chronic myeloid leukemia are insensitive to STI571 in vitro. Blood 99:319, 2002. [PMID: 11756187]
514. Meera V, Jijina F, Shrikande M, et al: Twin pregnancy in a patient of chronic myeloid leukemia on imatinib therapy. Leuk Res 32:1620, 2008. [PMID: 18420270]
515. Skoumalova I, Vondrakova J, Rohon P, et al: Successful childbirth in a patient with chronic myelogenous leukemia treated with imatinib mesylate during early pregnancy. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 152:121, 2008. [PMID: 18795086]
516. Ali R, Ozkalemka F, Ozçelik T, et al: Pregnancy under treatment of imatinib and successful labor in a patient with chronic myelogenous leukemia (CML). Outcome of discontinuation of imatinib therapy after achieving a molecular remission. Leuk Res 29:971, 2005. [PMID: 15978950]
517. Shapira T, Pereg D, Lishner M: How I treat acute and chronic leukemia in pregnancy. Blood Rev 22:247, 2008. [PMID: 18472198]
518. Pye SM, Cortes J, Ault P, Hatfield A, et al: The effects of imatinib on pregnancy outcome. Blood 111:5505, 2008. [PMID: 18322153]
519. Ault P, Kantarjian H, O’Brien S, et al: Pregnancy among patients with chronic myeloid leukemia treated with imatinib. J Clin Oncol 24:1204, 2006. [PMID: 16446320]
520. Ramasamy K, Hayden J, Lim Z, et al: Successful pregnancies involving men with chronic myeloid leukaemia on imatinib therapy. Br J Haematol 137:374, 2007. [PMID: 17408403]
521. Breccia M, Cannella L, Montefusco E, et al: Male patients with chronic myeloid leukemia treated with imatinib involved in healthy pregnancies: Report of five cases. Leuk Res 32:519, 2008. [PMID: 17804066]
522. Gambacorti-Passerini CB, Tornaghi L, Marangon E, et al: Imatinib concentrations in human milk. Blood 109:1790, 2007. [PMID: 17284536]
523. O’Dwyer ME, Gatter KM, Loriaux M, et al: Demonstration of Philadelphia chromosome negative abnormal clones in patients with chronic myelogenous leukemia during major cytogenetic responses induced by imatinib mesylate. Leukemia 17:481, 2003. [PMID: 12646934]
524. Guilbert-Douet N, Morel F, LeBris M-J, et al: Clonal chromosomal abnormalities in the Philadelphia chromosome negative cells of chronic myeloid leukemia patients treated with imatinib. Leukemia 18:1140, 2004. [PMID: 15085159]
525. Deininger MWN: Cytogenetic studies in patients on imatinib. Semin Hematol 40:50, 2003. [PMID: 12783376]
526. Bumm T, Muller C, Al-Ali K, et al: Emergence of clonal cytogenetic abnormalities in Ph-cells in some CML patients in cytogenetic remission to imatinib but restoration of polyclonal hematopoiesis in the majority. Blood 101:1941, 2003. [PMID: 12411298]
527. Goldberg SL, Medan RA, Rowley SD, et al: Myelodysplastic subclones in chronic myeloid leukemia: Implications for imatinib mesylate therapy. Blood 101:781, 2003. [PMID: 12509385]
528. Farag SS, Ruppert AS, Mrozek K, et al: Prognostic significance of additional cytogenetic abnormalities in newly diagnosed patients with Philadelphia chromosome-positive chronic myelogenous leukemia treated with interferon-: A Cancer and Leukemia Group B study. Int J Oncol 25:143, 2004. [PMID: 15201999]
529. Andersen MK, Pedersen-Bjergaard J, Kjeldsen I, et al: Clonal Ph-negative hematopoiesis in CML after therapy with imatinib mesylate is frequently characterized by trisomy 8. Leukemia 16:1390, 2002. [PMID: 12094265]
530. Terre C, Eclache V, Rousselot P, et al: Report of 34 patients with clonal chromosomal abnormalities in Philadelphia-negative cells during imatinib treatment of Philadelphia-positive chronic myeloid leukemia. Leukemia 18:1340, 2004. [PMID: 15190256]
531. Jabbour E, Kantarjian HM, Abruzzo LV, et al: Chromosomal abnormalities in Philadelphia chromosome negative metaphases appearing during imatinib mesylate therapy in patients with newly diagnosed chronic myeloid leukemia in chronic phase. Blood 110:2991, 2007. [PMID: 17625066]
532. Cortes JE, Talpaz M, Giles F, et al: Prognostic significance of cytogenetic clonal evolution in patients with chronic myelogenous leukemia on imatinib mesylate therapy. Blood 101:3794, 2003. [PMID: 12560227]
533. Chee YL, Vickers MA, Stevenson D, et al: Fatal myelodysplastic syndrome developing during therapy with imatinib mesylate and characterised by the emergence of complex Philadelphia negative clones. Leukemia 17:634, 2003. [PMID: 12646955]
534. Nimmanapalli R, O’Bryan E, Huang M, et al: Molecular characterization and sensitivity of STI-571 (imatinib mesylate, Gleevec)-resistant, Bcr-Abl-positive, human acute leukemia cells to SRC kinase inhibitor PD180970 and 17-allylamino-17-demethoxygeldanamycin. Cancer Res 62:5761, 2002. [PMID: 12384536]
535. Paterson SC, Smith KD, Holyoake TL, Jorgensen HG: Is there a cloud in the silver lining for imatinib? Br J Cancer 88:983, 2003. [PMID: 12671692]
536. Hochhaus A, La Rosse P: Imatinib therapy in chronic myelogenous leukemia: Strategies to avoid and overcome resistance. Leukemia 18:1320, 2004.
537. Cowan-Jacob SW, Guez V, Fendrich G, et al: Imatinib (STI571) resistance in chronic myelogenous leukemia: Molecular basis of the underlying mechanism and potential strategies for treatment. Mini Rev Med Chem 4:285, 2004. [PMID: 15032675]
538. Weisberg E, Griffin JD: Resistance to imatinib (Glivec): Update on clinical mechanisms. Drug Resist Updat 6:231, 2003. [PMID: 14643293]
539. Melo JV: Resistance to imatinib mesylate in CML: All BCR-ABL mutations “are created equal but some are more equal than others.” Blood 101:4231, 2003.
540. Villuendas R, Steegmann JL, Pollán M, et al: Identification of genes involved in imatinib resistance in CML: A gene-expression profiling approach. Leukemia 20:1047, 2006. [PMID: 16598311]
541. Jiang X, Zhao Y, Forrest D, et al: Stem cell biomarkers in chronic myeloid leukemia. Dis Markers 24:201, 2008. [PMID: 18525114]
542. Barnes DJ, Melo JV: Primitive, quiescent and difficult to kill: The role of non-proliferating stem cells in chronic myeloid leukemia. Cell Cycle 5:2862, 2006. [PMID: 17172863]
543. Michor F, Hughes TP, Iwasa Y, et al: Dynamics of chronic myeloid leukaemia. Nature 435:1267, 2005. [PMID: 15988530]
544. Jørgensen HG, Copland M, Allan EK, et al: Intermittent exposure of primitive quiescent chronic myeloid leukemia cells to granulocyte-colony stimulating factor in vitro promotes their elimination by imatinib mesylate. Clin Cancer Res 12:626, 2006. [PMID: 20120524]
545. Holtz M, Forman SJ, Bhatia R: Growth factor stimulation reduces residual quiescent chronic myelogenous leukemia progenitors remaining after imatinib treatment. Cancer Res 67:1113, 2007. [PMID: 17283145]
546. Jin L, Tabe Y, Konoplev S, et al: CXCR4 up-regulation by imatinib induces chronic myelogenous leukemia (CML) cell migration to bone marrow stroma and promotes survival of quiescent CML cells. Mol Cancer Ther 7:48, 2008. [PMID: 18202009]
547. le Coutre P, Tassi E, Varella-Garcia M, et al: Induction of resistance to the Abelson inhibitor STI571 in human leukemic cells through gene amplification. Blood 95:1758, 2000.
548. Campbell LJ, Patsouris C, Rayeroux KC, et al: BCR/ABL amplification in chronic myelocytic leukemia blast crisis following imatinib mesylate administration. Cancer Genet Cytogenet 139:30, 2002. [PMID: 12547154]
549. Donato NJ, Wu JY, Stapley J, et al: BCR-ABL independence and LYN kinase overexpression in chronic myelogenous leukemia cells selected for resistance to STI571. Blood 101:690, 2003. [PMID: 12509383]
550. Illmer T, Schaich M, Platzbecker U, et al: P-glycoprotein-mediated drug efflux is a resistance mechanism of chronic myelogenous leukemia cells to treatment with imatinib mesylate. Leukemia 18:401, 2004. [PMID: 14724652]
551. Mahon FX, Belloc F, Lagarde V, et al: MDR1 gene overexpression confers resistance to imatinib mesylate in leukemia cell line models. Blood 101:2368, 2003. [PMID: 12609962]
552. Gambacorti-Passerini C, Barni R, le Coutre P, et al: Role of alpha1 acid glycoprotein in the in vivo resistance of human BCR-ABL(+) leukemic cells to the abl inhibitor STI571. J Natl Cancer Inst 92:1641, 2000. [PMID: 11036109]
553. White DL, Saunders VA, Dang P, et al: OCT-1-mediated influx is a key determinant of the intracellular uptake of imatinib but not nilotinib (AMN107): Reduced OCT-1 activity is the cause of low in vitro sensitivity to imatinib. Blood 108:697, 2006. [PMID: 16597591]
554. Hiwase DK, Saunders V, Hewett D, et al: Dasatinib cellular uptake and efflux in chronic myeloid leukemia cells: Therapeutic implications. Clin Cancer Res 14:3881, 2008. [PMID: 18559609]
555. Jordanides NE, Jorgensen HG, Holyoake TL, et al: Functional ABCG2 is overexpressed on primary CML CD34+ cells and is inhibited by imatinib mesylate. Blood 108:1370, 2006. [PMID: 16627755]
556. Ossard-Receveur A, Bernheim A, Clausse B, et al: Duplication of the Ph-chromosome as a possible mechanism of resistance to imatinib mesylate in patients with chronic myelogenous leukemia. Cancer Genet Cytogenet 163:189, 2005. [PMID: 16337868]
557. Szych CM, Liesveld JL, Iqbal MA, et al: Isodicentric Philadelphia chromosomes in imatinib mesylate (Gleevec)-resistant patients. Cancer Genet Cytogenet 174:132, 2007. [PMID: 17452255]
558. Roche-Lestienne C, Preudhomme C: Mutations in the ABL kinase domain pre-exist the onset of imatinib treatment. Semin Hematol 21:80, 2003.
559. Corbin AS, LaRosee P, Stoffregen EP, et al: Several Bcr-Abl kinase domain mutants associated with imatinib mesylate resistance remain sensitive to imatinib. Blood 101:4611, 2003. [PMID: 12576318]
560. Khorashad JS, Anand M, Marin D, et al: The presence of a BCR-ABL mutant allele in CML does not always explain clinical resistance to imatinib. Leukemia 20:658, 2006. [PMID: 16467863]
561. Wei Y, Hardling M, Olsson B, et al: Not all imatinib resistance in CML are BCR-ABL kinase domain mutations. Ann Hematol 85:841, 2006. [PMID: 17006667]
562. Soverini S, Colarossi S, Gnani A, et al: Contribution of ABL kinase domain mutations to imatinib resistance in different subsets of Philadelphia-positive patients: By the GIMEMA Working Party on Chronic Myeloid Leukemia. Clin Cancer Res 12:7374, 2006. [PMID: 17189410]
563. Sherbenou DW, Wong MJ, Humayun A, et al: Mutations of the BCR-ABL-kinase domain occur in a minority of patients with stable complete cytogenetic response to imatinib. Leukemia 21:489, 2007. [PMID: 17252009]
564. Miething C, Mugler C, Grundler R, et al: Phosphorylation of tyrosine 393 in the kinase domain of Bcr-Abl influences the sensitivity towards imatinib in vivo. Leukemia 17:1695, 2003. [PMID: 12970766]
565. Branford S, Rudzki Z, Walsh S, et al: Detection of BRC-ABL mutations in patients with CML treated with imatinib is virtually always accompanied by clinical resistance, and mutations in the ATP phosphate-binding loop (P-loop) are associated with a poor prognosis. Blood 102:276, 2003. [PMID: 12623848]
566. Nicolini FE, Corm S, Lê QH, et al: Mutation status and clinical outcome of 89 imatinib mesylate-resistant chronic myelogenous leukemia patients: A retrospective analysis from the French intergroup of CML (Fi(phi)-LMC GROUP). Leukemia 20:1061, 2006. [PMID: 16642048]
567. Quintás-Cardama A, Cortes J: Therapeutic options against BCR-ABL1 T315I-positive chronic myelogenous leukemia. Clin Cancer Res 14:4392, 2008. [PMID: 20059135]
568. Modugno M, Casale E, Soncini C, et al: Crystal structure of the T315I Abl mutant in complex with the aurora kinases inhibitor PHA-739358. Cancer Res 67:7987, 2007. [PMID: 17804707]
569. Jabbour E, Kantarjian H, Jones D, et al: Characteristics and outcomes of patients with chronic myeloid leukemia and T315I mutation following failure of imatinib mesylate therapy. Blood 112:53, 2008. [PMID: 18403620]
570. de Lavallade H, Khorashad JS, Davis HP, et al: Interferon-alpha or homoharringtonine as salvage treatment for chronic myeloid leukemia patients who acquire the T315I BCR-ABL mutation. Blood 110:2779, 2007.
571. Jilani I, Kantarjian H, Gorre M, et al: Phosphorylation levels of BCR-ABL, CrkL, AKT and STAT5 in imatinib-resistant chronic myeloid leukemia cells implicate alternative pathway usage as a survival strategy. Leuk Res 32:643, 2008. [PMID: 17900686]
572. Pocaly M, Lagarde V, Etienne G, et al: Overexpression of the heat-shock protein 70 is associated to imatinib resistance in chronic myeloid leukemia. Leukemia 21:93, 2007. [PMID: 17109025]
573. Carter BZ, Mak DH, Schober WD, et al: Regulation of survivin expression through Bcr-Abl/MAPK cascade: Targeting survivin overcomes imatinib resistance and increases imatinib sensitivity in imatinib-responsive CML cells. Blood 107:1555, 2006. [PMID: 16254145]
574. Wu J, Meng F, Kong LY, et al: Association between imatinib-resistant BCR-ABL mutation-negative leukemia and persistent activation of LYN kinase. J Natl Cancer Inst 100:926, 2008. [PMID: 18577747]
575. Hochhaus A, Erben P, Ernst T, Mueller MC: Resistance to targeted therapy in chronic myelogenous leukemia. Semin Hematol 44:S15, 2007.
576. Feller SM, Tuchscherer G, Voss J: Hihg affinity molecular disruption of GRB2 protein complexes as a therapeutic strategy for chronic myelogenous leukemia. Leuk Lymphoma 44:411, 2003. [PMID: 12688310]
577. Hochhaus A: Cytogenetic and molecular mechanisms of resistance to imatinib. Semin Hematol 40:69, 2003. [PMID: 12783379]
578. Ohno R, Nakamura Y: Prediction of response to imatinib by cDNA microarray analysis. Semin Hematol 40:42, 2003. [PMID: 12783375]
579. Cortes J, Kantarjian H: Beyond dose escalation: Clinical options for relapse or resistance in chronic myelogenous leukemia. J Natl Compr Canc Netw 6 Suppl 2:S22, 2008.
580. Talpaz M, Shah NP, Kantarjian H, et al: Dasatinib in imatinib-resistant Philadelphia chromosome-positive leukemias. N Engl J Med 354:2531, 2006. [PMID: 16775234]
581. Martinelli G, Soverini S, Rosti G, Baccarani M: Dual tyrosine kinase inhibitors in chronic myeloid leukemia. Leukemia 19:1872, 2005. [PMID: 16179913]
582. Wong SF: Dasatinib dosing strategies in Philadelphia chromosome-positive leukemia. J Oncol Pharm Pract 15:17, 2009. [PMID: 18753186]
583. Shah NP, Kantarjian HM, Kim DW, et al: Intermittent target inhibition with dasatinib 100 mg once daily preserves efficacy and improves tolerability in imatinib-resistant and -intolerant chronic-phase chronic myeloid leukemia. J Clin Oncol 26:3204, 2008. [PMID: 18541900]
584. Hochhaus A, Kantarjian HM, Baccarani M, et al: Dasatinib induces notable hematologic and cytogenetic responses in chronic-phase chronic myeloid leukemia after failure of imatinib therapy. Blood 109:2303, 2007. [PMID: 17138817]
585. Kantarjian H, Pasquini R, Hamerschlak N, et al: Dasatinib or high-dose imatinib for chronic-phase chronic myeloid leukemia after failure of first-line imatinib: A randomized phase 2 trial. Blood 109:5143, 2007. [PMID: 17317857]
586. Porkka K, Koskenvesa P, Lundán T, et al: Dasatinib crosses the blood-brain barrier and is an efficient therapy for central nervous system Philadelphia chromosome-positive leukemia. Blood 112:1005, 2008. [PMID: 18477770]
587. Baraska M, Lewandowski K, Gniot M, et al: Dasatinib treatment can overcome imatinib and nilotinib resistance in CML patient carrying F359I mutation of BCR-ABL oncogene. J Appl Genet 49:201, 2008.
588. Hiwase DK, Saunders V, Hewett D, et al: Dasatinib cellular uptake and efflux in chronic myeloid leukemia cells: Therapeutic implications. Clin Cancer Res 14:3881, 2008. [PMID: 18559609]
589. Soverini S, Colarossi S, Gnani A, et al: Resistance to dasatinib in Philadelphia-positive leukemia patients and the presence or the selection of mutations at residues 315 and 317 in the BCR-ABL kinase domain. Haematologica 92:401, 2007. [PMID: 17339191]
590. Golemovic M, Verstovsek S, Giles F, et al: AMN107, a novel amino pyrimidine inhibitor of Bcr-Abl, has in vitro activity against imatinib-resistant chronic myeloid leukemia. Clin Cancer Res 11:4941, 2005. [PMID: 16000593]
591. Jørgensen HG, Allan EK, Jordanides NE, et al: Nilotinib exerts equipotent antiproliferative effects to imatinib and does not induce apoptosis in CD34+ CML cells. Blood 109:4016, 2007. [PMID: 20120524]
592. Kantarjian HM, Giles F, Gattermann N, et al: Nilotinib (formerly AMN107), a highly selective BCR-ABL tyrosine kinase inhibitor, is effective in patients with Philadelphia chromosome-positive chronic myelogenous leukemia in chronic phase following imatinib resistance and intolerance. Blood 110:3540, 2007. [PMID: 17715389]
593. Hazarika M, Jiang X, Liu Q, et al: Tasigna for chronic and accelerated phase Philadelphia chromosome-positive chronic myelogenous leukemia resistant to or intolerant of imatinib. Clin Cancer Res 14:5325, 2008. [PMID: 18765523]
594. Weisberg E, Catley L, Wright RD, et al: Beneficial effects of combining nilotinib and imatinib in preclinical models of BCR-ABL+ leukemias. Blood 109:2112, 2007. [PMID: 17068153]
595. Tipping AJ, Mahon FX, Zafirides G, et al: Drug responses of imatinib mesylate-resistant cells: Synergism of imatinib with other chemotherapeutic drugs. Leukemia 16:2349, 2002. [PMID: 12454739]
596. Tipping AJ, Melo JV: Imatinib mesylate in combination with other hemotherapeutic drugs: In vitro studies. Semin Hematol 40:83, 2003. [PMID: 12783381]
597. Kantarjian HM, Talpaz M, Smith TL, et al: Homoharringtonine and low-dose cytarabine in the management of late chronic-phase chronic myelogenous leukemia. J Clin Oncol 18:3513, 2000. [PMID: 11032593]
598. O’Dwyer ME, La Rosee P, Nimmanapalli R, et al: Recent advances in Philadelphia chromosome-positive malignancies: The potential role of arsenic trioxide. Semin Hematol 39:18, 2002. [PMID: 12012318]
599. Kantarjian HM, O’Brien S, Corteo J, et al: Results of decitabine (5-aza-2′-deoxycytidine) therapy in 130 patients with chronic myelogenous leukemia. Cancer 98:522, 2003. [PMID: 12879469]
600. Issa JP, Garcia-Manero G, Giles FJ, et al: Phase 1 study of low-dose prolonged exposure schedules of the hypomethylating agent 5-aza-2′-deoxycytidine (decitabine) in hematopoietic malignancies. Blood 103:1635, 2004. [PMID: 14604977]
601. Chand M, Thakuri M, Keung YK: Imatinib mesylate associated with delayed hematopoietic recovery after concomitant chemotherapy. Leukemia 18:886, 2004. [PMID: 14961027]
602. O’Hare T, Walters DK, Stoffregen EP, et al: Combined Abl inhibitor therapy for minimizing drug resistance in chronic myelogenous leukemia. Src/Abl inhibitors are compatible with imatinib. Clin Cancer Res 11:6987, 2005. [PMID: 16203792]
603. Sun X, Layton JE, Elefanty A, Lieschke GJ: Comparison of effects of the tyrosine kinase inhibitors AG957, AG490, and STI571 on BCRABL-expressing cells, demonstrating synergy between AG490 and STI571. Blood 97:2008, 2001. [PMID: 11264165]
604. Mohi MG, Boulton C, Gu TL, et al: Combination of rapamycin and protein tyrosine kinase (PTK) inhibitors for the treatment of leukemias caused by oncogenic PTKs. Proc Natl Acad Sci U S A 101:3130, 2004. [PMID: 14976243]
605. Gatto S, Scappini B, Pham L, et al: The proteasome inhibitor PS-341 inhibits growth and induces apoptosis in Bcr/Abl-positive cell lines sensitive and resistant to imatinib mesylate. Haematologica 88:853, 2003. [PMID: 12935973]
606. Dai Y, Rahmani M, Pei XY, et al: Bortezomib and flavopiridol interact synergistically to induce apoptosis in chronic myeloid leukemia cells resistant to imatinib mesylate through both Bcr/Abl-dependent and -independent mechanisms. Blood 104:509, 2004. [PMID: 15039284]
607. Yu C, Rahmani M, Conrad D, et al: The proteasome inhibitor bortezomib interacts synergistically with histone deacetylase inhibitors to induce apoptosis in Bcr/Abl+ cells sensitive and resistant to STI571. Blood 102:3765, 2003. [PMID: 12893773]
608. Fiskus W, Pranpat M, Bali P, et al: Combined effects of novel tyrosine kinase inhibitor AMN107 and histone deacetylase inhibitor LBH589 against Bcr-Abl-expressing human leukemia cells. Blood 108:645, 2006. [PMID: 16537804]
609. Chu S, Holtz M, Gupta M, Bhatia R: BCR/ABL kinase inhibition by imatinib mesylate enhances MAP kinase activity in chronic myelogenous leukemia CD34+ cells. Blood 103:3167, 2004. [PMID: 15070699]
610. Kuroda J, Kimura S, Segawa H, et al: The third-generation bisphosphonate zoledronate synergistically augments the anti-Ph+ leukemia activity of imatinib mesylate. Blood 102:2229, 2003. [PMID: 12763930]
611. Keating A: Chronic myeloid leukemia: Current therapies and the potential role of farnesyltransferase inhibitors. Semin Hematol 39:11, 2002. [PMID: 12214288]
612. Daley GQ: Towards combination target-directed chemotherapy for chronic myeloid leukemia: Role of farnesyl transferase inhibitors. Semin Hematol 40:11, 2003. [PMID: 12783369]
613. Nakajima A, Tauchi T, Sumi M, et al: Efficacy of SCH66336, a farnesyl transferase inhibitor, in conjunction with imatinib against BCRABL-positive cells. Mol Cancer Ther 2:219, 2003. [PMID: 12657715]
614. Hoover RR, Mahon FX, Melo JV, Daley GQ: Overcoming STI571 resistance with the farnesyl transferase inhibitor SCH66336. Blood 100:1068, 2002. [PMID: 12130526]
615. Druker BJ: Overcoming resistance to imatinib by combining targeted agents. Mol Cancer Ther 2:225, 2003. [PMID: 12657716]
616. 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, 2003. [PMID: 12411300]
617. Sawyers CL, Hochhaus A, Feldman E, et al: Imatinib induces hematologic and cytogenetic responses in patients with chronic myelogenous leukemia in myeloid blast crisis: Results of a phase II study. Blood 99:3530, 2002. [PMID: 11986204]
618. Gorre ME, Ellwood-Yen K, Chiosis G, et al: BCR-ABL point mutants isolated from patients with imatinib mesylate-resistant chronic myeloid leukemia remain sensitive to inhibitors of the BCR-ABL chaperone heat shock protein 90. Blood 100:3041, 2002. [PMID: 12351420]
619. Deininger M: Resistance and relapse with imatinib in CML: Causes and consequences. J Natl Compr Canc Netw 6 Suppl 2:S11, 2008.
620. Wu CJ, Neuberg D, Chillemi A, et al: Quantitative monitoring of BCR/ABL transcript during STI-571 therapy. Leuk Lymphoma 43:2281, 2002. [PMID: 12613514]
621. Druker BJ: Imatinib as a paradigm of targeted therapies. J Clin Oncol 21:239, 2003.
622. Druker BJ: STI571 (Gleevec) as a paradigm for cancer therapy. Trends Mol Med 8:S14, 2002.
623. Merx K, Muller MC, Kreil S, et al: Early reduction of BCR-ABL mRNA transcript levels predicts cytogenetic response in chronic phase CML patients treated with imatinib after failure of interferon alpha. Leukemia 16:1579, 2002. [PMID: 12200666]
624. Tam CS, Kantarjian H, Garcia-Manero G, et al: Failure to achieve a major cytogenetic response by 12 months defines inadequate response in patients receiving nilotinib or dasatinib as second or subsequent line therapy for chronic myeloid leukemia. Blood 112:516, 2008. [PMID: 18492956]
625. Carella AM: Questioning the aim of CML therapy in the era of Imatinib? Leukemia 17:1199, 2003. [PMID: 12764394]
626. DeAngelo DJ, Ritz J: Imatinib therapy for patients with chronic myelogenous leukemia: Are patients living longer? Clin Cancer Res 10:1, 2004. [PMID: 14734443]
627. Goldman JM, Marin D, Olavarria E, Apperley JF: Clinical decisions for chronic myeloid leukemia in the imatinib era. Semin Hematol 40:98, 2003. [PMID: 12783383]
628. Goldman JM: Chronic myeloid leukemia—Still a few questions. Exp Hematol 32:2, 2004. [PMID: 14725895]
629. Goldman JM: How I treat chronic myeloid leukemia in the imatinib era. Blood 110:2828, 2007. [PMID: 17626839]
630. Bonifazi F, Bandini G, Rondelli D, et al: Reduced incidence of GVHD without increase in relapse with low-dose rabbit ATG in the preparative regimen for unrelated bone marrow transplants in CML. Bone Marrow Transplant 32:237, 2003. [PMID: 12858193]
631. Baccarani M, Russo D, Rosti G, Martinelli G: Interferon-alpha for chronic myeloid leukemia. Semin Hematol 40:22, 2003. [PMID: 12563609]
632. Kluin-Nelemans HC, Buck G, Le Cessie S, et al: Randomized comparison of low-dose versus high-dose interferon-alfa in chronic myeloid leukemia: Prospective collaboration of 3 joint trials by the MRC and HOVON groups. Blood 103:4408, 2004. [PMID: 15010373]
633. Michallet M, Maloisel F, Delain M, et al: Pegylated recombinant interferon alpha-2b vs recombinant interferon alpha-2b for the initial treatment of chronic-phase chronic myelogenous leukemia: A phase III study. Leukemia 18:309, 2004. [PMID: 14671645]
634. Lipton JH, Khoroshko N, Golenkov A, et al: Phase II, randomized, multicenter, comparative study of peginterferon-alpha-2a (40 kD) (Pegasys) versus interferon alpha-2a (Roferon-A) in patients with treatment-naïve, chronic-phase chronic myelogenous leukemia. Leuk Lymphoma 48:497, 2007. [PMID: 17454589]
635. Garcia-Manero G, Talpaz M, Giles FJ, et al: Treatment of Philadelphia chromosome-positive chronic myelogenous leukemia with weekly polyethylene glycol formulation of interferon-alpha-2b and low-dose cytosine arabinoside. Cancer 97:3010, 2003. [PMID: 12784336]
636. Roy L, Guilhot J, Krahnke T, et al: Survival advantage from imatinib compared with the combination interferon-alpha plus cytarabine in chronic-phase chronic myelogenous leukemia: Historical comparison between two phase 3 trials. Blood 108:1478, 2006. [PMID: 16627756]
637. Talpaz M: Interferon-alfa-based treatment of chronic myeloid leukemia and implications of signal transduction inhibition. Semin Hematol 38:22, 2001. [PMID: 11526598]
638. Kujawski LA, Talpaz M: The role of interferon-alpha in the treatment of chronic myeloid leukemia. Cytokine Growth Factor Rev 18:459, 2007. [PMID: 17703986]
639. Alimena G, Breccia M, Luciano L, et al: Imatinib mesylate therapy in chronic myeloid leukemia patients in stable complete cytogenic response after interferon-alpha results in a very high complete molecular response rate. Leuk Res 32:255, 2008. [PMID: 17692911]
640. Branford S, Hughes T, Milner A, et al: Efficacy and safety of imatinib in patients with chronic myeloid leukemia and complete or near-complete cytogenetic response to interferon-alpha. Cancer 110:801, 2007. [PMID: 17607681]
641. Kolitz JE, Kempin SF, Schluger A, et al: A phase II trial of high-dose hydroxyurea in chronic myelogenous leukemia. Semin Oncol 19:27, 1992. [PMID: 1641654]
642. Abhyankar D, Shende C, Saikia T, Advani SH: Hydroxyurea induced leg ulcers. J Assoc Physicians India 48:926, 2000. [PMID: 11198798]
643. Guilhot F, Chastang C, Michallet M, et al: Interferon alfa-2b combined with cytarabine versus interferon alone in chronic myelogenous leukemia. N Engl J Med 337:223, 1997. [PMID: 9227927]
644. Hehlmann R, Heimpel H, Hasford J, et al: Randomized comparison of busulfan and hydroxyurea in chronic myelogenous leukemia: Prolongation of survival by hydroxyurea. Blood 82:398, 1993. [PMID: 8329700]
645. O’Brien S, Kantarjian H, Keating M, et al: Homoharringtonine therapy induces responses in patients with chronic myelogenous leukemia in late chronic phase. Blood 86:3322, 1995. [PMID: 7579434]
646. Clarkson B: Chronic myelogenous leukemia: Is aggressive treatment indicated? J Clin Oncol 3:135, 1985. [PMID: 2578556]
647. Cortes J, Jabbour E, Daley GQ, et al: Phase 1 study of lonafarnib (SCH 66336) and imatinib mesylate in patients with chronic myeloid leukemia who have failed prior single-agent therapy with imatinib. Cancer 110:1295, 2007. [PMID: 17623836]
648. Cortes J, Quintás-Cardama A, Garcia-Manero G, et al: Phase 1 study of tipifarnib in combination with imatinib for patients with chronic myelogenous leukemia in chronic phase after imatinib failure. Cancer 110:2000, 2007. [PMID: 17849425]
649. Issa JP, Gharibyan V, Cortes J, et al: Phase II study of low-dose decitabine in patients with chronic Myelogenous leukemia resistant to imatinib mesylate. J Clin Oncol 23:3948, 2005. [PMID: 15883410]
650. Xu R, Dong Q, Yu Y, et al: Berbamine: A novel inhibitor of bcr/abl fusion gene with potent anti-leukemia activity. Leuk Res 30:17, 2006. [PMID: 16023722]
651. Chandra J, Tracy J, Loegering D, et al: Adaphostin-induced oxidative stress overcomes BCR/ABL mutation-dependent and -independent imatinib resistance. Blood 107:2501, 2006. [PMID: 16291594]
652. Yokota A, Kimura S, Masuda S, et al: INNO-406, a novel BCR-ABL/Lyn dual tyrosine kinase inhibitor, suppresses the growth of Ph+ leukemia cells in the central nervous system, and cyclosporine A augments its in vivo activity. Blood 109:306, 2007. [PMID: 16954504]
653. Kamitsuji Y, Kuroda J, Kimura S, et al: The Bcr-Abl kinase inhibitor INNO-406 induces autophagy and different modes of cell death execution in Bcr-Abl-positive leukemias. Cell Death Differ 15:1712, 2008. [PMID: 18617896]
654. Carew JS, Nawrocki ST, Kahue CN, et al: Targeting autophagy augments the anticancer activity of the histone deacetylase inhibitor SAHA to overcome Bcr-Abl-mediated drug resistance. Blood 110:313, 2007. [PMID: 17363733]
655. Puttini M, Coluccia AM, Boschelli F, et al: In vitro and in vivo activity of SKI-606, a novel Src-Abl inhibitor, against imatinib-resistant Bcr-Abl+ neoplastic cells. Cancer Res 66:11314, 2006. [PMID: 17114238]
656. Noronha G, Cao J, Chow CP, et al: Inhibitors of ABL and the ABL-T315I mutation. Curr Top Med Chem 8:905, 2008. [PMID: 18673174]
657. Barbarotto E, Calin GA: Potential therapeutic applications of miRNA-based technology in hematological malignancies. Curr Pharm Des 14:2040, 2008. [PMID: 18691114]
658. Koldehoff M, Steckel NK, Beelen DW, Elmaagacli AH: Therapeutic application of small interfering RNA directed against bcr-abl transcripts to a patient with imatinib-resistant chronic myeloid leukaemia. Clin Exp Med 7:47, 2007. [PMID: 17609876]
659. James HA: The potential application of ribozymes for the treatment of hematological disorders. J Leukoc Biol 66:361, 1999. [PMID: 10496304]
660. Mendoza-Maldonado R, Zentilin L, Fanin R, Giacca M: Purging of chronic myelogenous leukemia cells by retrovirally expressed anti-bcrabl ribozymes with specific cellular compartmentalization. Cancer Gene Ther 9:71, 2002. [PMID: 11916246]
661. Cotter FE: Antisense oligonucleotides for haematological malignancies. Haematologica 84:19, 1999. [PMID: 10907458]
662. Verfaillie CM, McIvor S, Zhao RCH: Gene therapy for chronic myelogenous leukemia. Mol Med Today 5:359, 1999. [PMID: 10431169]
663. Clark RE, Dodi A, Hill SC, et al: Direct evidence that leukemic cells present HLA-associated immunogenic peptides derived from the BCRABL b3a2 fusion protein. Blood 98:2887, 2001. [PMID: 11698267]
664. Schwartz J, Pinilla-Ibarz J, Yuan RR, Scheinberg DA: Novel targeted and immunotherapeutic strategies in chronic myeloid leukemia. Semin Hematol 40:87, 2003. [PMID: 12563615]
665. Nossner E, Gastpar R, Milani V, et al: Tumor-derived heat shock protein 90 peptide complexes are cross-presented by human dendritic cells. J Immunol 169:5424, 2002.
666. Pinilla-Ibarz J, Cathcart K, Korontsvit T, et al: Vaccination of patients with chronic myelogenous leukemia with bcr-abl oncogene breakpoint fusion peptides generates specific immune responses. Blood 95:1781, 2000. [PMID: 10688838]
667. Maslak PG, Dao T, Gomez M, et al: A pilot vaccination trial of synthetic analog peptides derived from the BCR-ABL breakpoints in CML patients with minimal disease. Leukemia 22:1613, 2008. [PMID: 18256684]
668. Yasukawa M, Ohminami H, Kojima K, et al: HLA class II-restricted antigen presentation of endogenous bcr-abl fusion protein by chronic myelogenous leukemia-derived dendritic cells to CD4+ T lymphocytes. Blood 98:1498, 2001. [PMID: 11520800]
669. Tschiedel S, Gentilini C, Lange T, et al: Identification of NM23-H2 as a tumour-associated antigen in chronic myeloid leukaemia. Leukemia 22:1542, 2008. [PMID: 18496563]
670. Borrello I, Sotomayor EM, Rattis F-M, et al: Sustaining the graft-versus-tumor effect through posttransplant immunization with granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing tumor vaccines. Blood 95:3011, 2000. [PMID: 10807763]
671. Kessler JH, Bres-Vloemans SA, van Veelen PA, et al: BCR-ABL fusion regions as a source of multiple leukemia-specific CD8+ T-cell epitopes. Leukemia 20:1738, 2006. [PMID: 16932347]
672. Rojas JM, Knight K, Wang L, Clark RE: Clinical evaluation of BCR-ABL peptide immunisation in chronic myeloid leukaemia: Results of the EPIC study. Leukemia 21:2287, 2007. [PMID: 17637811]
673. Wagner H, McKeough PG, Desforges J, Madoc-Jones H: Splenic irradiation in the treatment of patients with chronic myelogenous leukemia or myelofibrosis and myeloid metaplasia. Cancer 58:1204, 1986. [PMID: 2427184]
674. McFarland JT, Kuzma C, Millard FE, Johnstone PA: Palliative irradiation of the spleen. Am J Clin Oncol 26:178, 2003. [PMID: 12714892]
675. The Italian Cooperative Study Group on Chronic Myeloid Leukemia: Results of a prospective randomized trial of early splenectomy in chronic myeloid leukemia. Cancer 54:333, 1984.
676. Mesa RA, Elliott MA, Tefferi A: Splenectomy in chronic myeloid leukemia and myelofibrosis with myeloid metaplasia. Blood Rev 14:121, 2000. [PMID: 10986148]
677. Kalhs P, Schwarzinger I, Anderson G, et al: A retrospective analysis of the long-term effect of splenectomy on late infections, graft-versus-host disease, relapse, and survival after allogeneic marrow transplantation for chronic myelogenous leukemia. Blood 86:2028, 1995. [PMID: 7655031]
678. Rodrigues CA, Fermino FA, Vasconcelos Y, De Oliveira JS: Refractory chronic GVHD emerging after splenectomy in a marrow transplant recipient with accelerated phase CML. Bone Marrow Transplant 32:333, 2003. [PMID: 12858207]
679. Fadilah SA, Ahmad-Zailani R, Soon-Keng C, Norlaila M: Successful treatment of chronic myeloid leukemia during pregnancy with hydroxyurea. Leukemia 16:1202, 2002. [PMID: 12040456]
680. Mubarek AA, Kakil IR, Al-Homsi U, et al: Normal outcome of pregnancy in chronic myeloid leukemia treated with interferon-alpha in 1st trimester: Report of 3 cases and review of the literature. Am J Hematol 69:115, 2002.
681. CML Autograft Trials Collaboration: Autologous stem cell transplantation in chronic myeloid leukaemia: A meta-analysis of six randomized trials. Cancer Treat Rev 33:39, 2007.
682. Goldman J: Autologous stem-cell transplantation for chronic myelogenous leukemia. Semin Hematol 30:53, 1993. [PMID: 7901914]
683. Talpaz M, Kantarjian H, Liang J, et al: Percentage of Philadelphia chromosome (Ph)-negative and Ph-positive cells found after autologous transplantation for chronic myelogenous leukemia depends on percentage of diploid cells induced by conventional dose chemotherapy before collection of autologous cells. Blood 85:3257, 1995. [PMID: 7756658]
684. Drummond MW, Marin D, Clark RE, et al: Mobilization of Ph chromosome-negative peripheral blood stem cells in chronic myeloid leukemia patients with imatinib mesylate-induced complete cytogenetic remission. Br J Haematol 123:479, 2003. [PMID: 14617010]
685. Hui CH, Goh KY, White D, et al: Successful peripheral blood stem cell mobilisation with filgrastim in patients with chronic myeloid leukaemia achieving complete cytogenetic response with imatinib, without increasing disease burden as measured by quantitative real-time PCR. Leukemia 17:821, 2003. [PMID: 12750692]
686. Kreuzer KA, Kluhs C, Baskaynak G, et al: Filgastrim-induced stem cell mobilization in chronic myeloid leukaemia patients during imatinib therapy: Safety, feasibility and evidence for an efficient in vivo purging. Br J Haematol 124:195, 2004. [PMID: 14687030]
687. Gordon MK, Sher D, Karrison T, et al: Successful autologous stem cell collection in patients with chronic myeloid leukemia in complete cytogenetic response, with quantitative measurement of BCR-ABL expression in blood, marrow, and apheresis products. Leuk Lymphoma 49:531, 2008. [PMID: 18297531]
688. Olavarria E: Autologous stem cell transplantation in chronic myeloid leukemia. Semin Hematol 44:252, 2007. [PMID: 17961724]
689. Perseghin P, Gambacorti-Passerini C, Tornaghi L, et al: Peripheral blood progenitor cell collection in chronic myeloid leukemia patients with complete cytogenetic response after treatment with imatinib mesylate. Transfusion 45:1214, 2005. [PMID: 15987369]
690. Cervantes F, Hernandez-Boluda JC, Odriozola J, et al: Imatinib mesylate (STI571) treatment in patients with chronic-phase chronic myelogenous leukaemia previously submitted to autologous stem cell transplantation. Br J Haematol 120:500, 2003. [PMID: 12580969]
691. Simon W, Segel GB, Lichtman MA: Early allogeneic stem cell transplantation for chronic myelogenous leukemia in the imatinib era: A preliminary assessment. Blood Cells Mol Dis 37:116, 2006. [PMID: 16904348]
692. Goldman J: Allogeneic stem cell transplantation for chronic myeloid leukemia—Status in 2007. Bone Marrow Transplant 42:S11, 2008.
693. Giralt SA, Arora M, Goldman JM, et al: Chronic Leukemia Working Committee, Center for International Blood and Marrow Transplant Research. Impact of imatinib therapy on the use of allogeneic haematopoietic progenitor cell transplantation for the treatment of chronic myeloid leukaemia. Br J Haematol 137:461, 2007. [PMID: 17459051]
694. Maziarz RT: Who with chronic myelogenous leukemia to transplant in the era of tyrosine kinase inhibitors? Curr Opin Hematol 15:127, 2008. [PMID: 18300759]
695. Hehlmann R, Berger U, Pfirrmann M, et al: Drug treatment is superior to allografting as first-line therapy in chronic myeloid leukemia. Blood 109:4686, 2007. [PMID: 17317858]
696. Thomas ED, Clift RA, Fefer A, et al: Marrow transplantation for the treatment of chronic myelogenous leukemia. Ann Intern Med 104:155, 1986. [PMID: 3511810]
697. Apperley JF: Hematopoietic stem cell transplantation in chronic myeloid leukemia. Curr Opin Hematol 5:445, 1998. [PMID: 9814654]
698. Cooperative Study Group on Chromosomes in Transplanted Patients: Cytogenetic follow-up of 100 patients submitted to bone marrow transplantation for Philadelphia chromosome-positive chronic myeloid leukemia. Eur J Haematol 40:50, 1988.
699. McGlave P, Bartoch G, Anasetti C, et al: Unrelated donor marrow transplantation therapy for chronic myelogenous leukemia. Blood 81:543, 1993. [PMID: 8422471]
700. Fernandez HF, Tran HT, Albrecht F, et al: Evaluation of safety and pharmacokinetics of administering intravenous busulfan in a twice-daily or daily schedule to patients with advanced hematologic malignant disease undergoing stem cell transplantation. Biol Blood Marrow Transplant 8:486, 2002. [PMID: 12374453]
701. Radich JP, Gooley T, Bensinger W, et al: HLA-matched related hematopoietic cell transplantation for chronic-phase CML using a targeted busulfan and cyclophosphamide preparative regimen. Blood 102:31, 2003. [PMID: 12595317]
702. Goldman J: Implications of imatinib mesylate for hematopoietic stem cell transplantation. Semin Hematol 38:28, 2001. [PMID: 11526599]
703. Barrett J: Allogeneic stem cell transplantation for chronic myeloid leukemia. Semin Hematol 40:59, 2003. [PMID: 12563612]
704. Messner HA, Curtis JE, Lipton JL, et al: Three decades of allogeneic bone marrow transplants at the Princess Margaret Hospital. Clin Transplant 289, 1999.
705. Byrne JL, Stainer C, Hyde H, et al: Low incidence of acute graft-versus-host disease and recurrent leukaemia in patients undergoing allogeneic haemopoietic stem cell transplantation from sibling donors with methotrexate and dose-monitored cyclosporin A prophylaxis. Bone Marrow Transplant 22:541, 1988.
706. Goldman J, Apperley J, Kanfer E, et al: Imatinib or transplant for chronic myeloid leukemia? Lancet 362:172, 2003. [PMID: 12867128]
707. Van Rhee F, Szydlo RM, Hermans J, et al: Long-term results after allogeneic bone marrow transplantation for chronic myelogenous leukemia in chronic phase: A report from the Chronic Leukemia Working Party of the European Groups for Blood and Marrow Transplantation. Bone Marrow Transplant 20:553, 1997.
708. Szydlo R, Goldman JM, Klein JP, et al: Results of allogeneic bone marrow transplants using donors other than HLA-identical siblings. J Clin Oncol 15:1767, 1997. [PMID: 9164184]
709. Petersdorf EW, Longton GM, Anasetti C, et al: The significance of HLA-DRBI matching on clinical outcome after HLA-A band DR identical unrelated donor transplantation. Blood 86:1606, 1995. [PMID: 7632970]
710. Weisdorf DJ, Anasetti C, Antin JH, et al: Allogeneic bone marrow transplantation for chronic myelogenous leukemia: Comparative analysis of unrelated versus matched sibling donor transplantation. Blood 99:1971, 2002. [PMID: 11877268]
711. Laporte JP, Gorin NC, Rubinstein P, et al: Cord-blood transplantation from an unrelated donor in an adult with chronic myelogenous leukemia. N Engl J Med 335:167, 1997.
712. Oehler VG, Gooley T, Snyder DS, et al: The effects of imatinib mesylate treatment before allogeneic transplantation for chronic myeloid leukemia. Blood 109:1782, 2007. [PMID: 17062727]
713. Weisser M, Schmid C, Schoch C, et al: Resistance to pretransplant imatinib therapy may adversely affect the outcome of allogeneic stem cell transplantation in CML. Bone Marrow Transplant 36:1017, 2005. [PMID: 16184176]
714. Jabbour E, Cortes J, Kantarjian H, et al: Novel tyrosine kinase inhibitor therapy before allogeneic stem cell transplantation in patients with chronic myeloid leukemia: No evidence for increased transplant-related toxicity. Cancer 110:340, 2007. [PMID: 17559140]
715. Radich JP, Gehly G, Gooley T, et al: Polymerase chain reaction detection of the BCR-ABL fusion transcript after allogeneic marrow transplantation for chronic myeloid leukemia: Results and implications in 346 patients. Blood 85:2632, 1995. [PMID: 7727789]
716. Goldman JM: Therapeutic strategies for chronic myeloid leukemia in chronic (stable) phase. Semin Hematol 40:10, 2003. [PMID: 12682876]
717. Miller JS, Cooley S, Parham P, et al: Missing KIR ligands are associated with less relapse and increased graft-versus-host disease (GVHD) following unrelated donor allogeneic HCT. Blood 109:5058, 2007. [PMID: 17317850]
718. Elmaagacli AH, Ottinger H, Koldehoff M, et al: Reduced risk for molecular disease in patients with chronic myeloid leukemia after transplantation from a KIR-mismatched donor. Transplantation 79:1741, 2005. [PMID: 15973179]
719. Nadal E, Garin M, Kaeda J, et al: Increased frequencies of CD4(+)CD25(high) T(regs) correlate with disease relapse after allogeneic stem cell transplantation for chronic myeloid leukemia. Leukemia 21:472, 2007. [PMID: 17215853]
720. Crawley C, Szydlo R, Lalancette M, et al: Outcomes of reduced-intensity transplantation for chronic myeloid leukemia: An analysis of prognostic factors from the Chronic Leukemia Working Party of the EBMT. Blood 106:2969, 2005. [PMID: 15998838]
721. Kebriaei P, Detry MA, Giralt S: Long-term follow-up of allogeneic hematopoietic stem-cell transplantation with reduced-intensity conditioning for patients with chronic myeloid leukemia. Blood 110:3456, 2007. [PMID: 17652620]
722. Uzunel M, Mattsson J, Brune M, et al: Kinetics of minimal residual disease and chimerism in patients with chronic myeloid leukemia after nonmyeloablative conditioning and allogeneic stem cell transplantation. Blood 101:469, 2003. [PMID: 12393598]
723. Or R, Shapira MY, Resnick I, et al: Nonmyeloablative allogeneic stem cell transplantation for the treatment of chronic myeloid leukemia in first chronic phase. Blood 101:441, 2003.
724. Bornhauser M, Kiehl M, Siegert W, et al: Dose-reduced conditioning for allografting in 44 patients with chronic myeloid leukaemia: A retrospective analysis. Br J Haematol 115:119, 2001. [PMID: 11722421]
725. Das M, Saikia TK, Advani SH, et al: Use of a reduced-intensity conditioning regimen for allogeneic transplantation in patients with chronic myeloid leukemia. Bone Marrow Transplant 32:125, 2003. [PMID: 12838275]
726. Feinstein L, Storb R: Reducing transplant toxicity. Curr Opin Hematol 8:342, 2001. [PMID: 11604573]
727. Koh LP, Hwang WY, Chuah CT, et al: Imatinib mesylate (STI-571) given concurrently with nonmyeloablative stem cell transplantation did not compromise engraftment and resulted in cytogenetic remission in a patient with chronic myeloid leukemia in blast crisis. Bone Marrow Transplant 31:305, 2003. [PMID: 12621468]
728. McCann SR: Molecular response to imatinib mesylate following relapse after allogeneic SCT for CML. Blood 101:1200, 2003. [PMID: 12529295]
729. Vandenberghe P, Boeckx N, Ronsyn E, et al: Imatinib mesylate induces durable complete remission of advanced CML persisting after allogeneic bone marrow transplantation. Leukemia 17:458, 2003. [PMID: 12592347]
730. Ullmann AJ, Hess G, Kolbe K, et al: Current results on the use of imatinib mesylate in patients with relapsed Philadelphia chromosome positive leukemia after allogeneic or syngeneic hematopoietic stem cell transplantation. Keio J Med 52:182, 2003. [PMID: 14529151]
731. Olavarria E, Craddock C, Dazzi F, et al: Imatinib mesylate (STI571) in the treatment of relapse of chronic myeloid leukemia after allogeneic stem cell transplantation. Blood 99:3861, 2002. [PMID: 11986250]
732. Carpenter PA, Snyder DS, Flowers ME, et al: Prophylactic administration of imatinib after hematopoietic cell transplantation for high-risk Philadelphia chromosome-positive leukemia. Blood 109:2791, 2007. [PMID: 17119111]
733. Olavarria E, Siddique S, Griffiths MJ, et al: Posttransplantation imatinib as a strategy to postpone the requirement for immunotherapy in patients undergoing reduced-intensity allografts for chronic myeloid leukemia. Blood 110:4614, 2007. [PMID: 17881635]
734. Weisser M, Tischer J, Schnittger S, et al: A comparison of donor lymphocyte infusions or imatinib mesylate for patients with chronic myelogenous leukemia who have relapsed after allogeneic stem cell transplantation. Haematologica 91:663, 2006. [PMID: 16627251]
735. Sullivan KM: Marrow transplantation for disorders of hematopoiesis. Leukemia 7:1098, 1993. [PMID: 8321033]
736. Kolb HJ, Mittermuller J, Clemm CH, et al: Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood 76:2462, 1990. [PMID: 2265242]
737. Dazzi F, Szydlo RM, Goldman JM: Donor lymphocyte infusion for relapse of chronic myeloid leukemia after allogeneic stem cell transplant: Where we now stand. Exp Hematol 27:1477, 1999. [PMID: 10517488]
738. Van Rhee F, Lin F, Cullis JO, et al: Relapse of chronic myeloid leukemia after allogeneic bone marrow transplant: The case of giving donor leukocyte transfusions before the onset of hematologic relapse. Blood 83:3377, 1994.
739. Leis J, Porter DL: Unrelated donor leukocyte infusions to treat relapse after unrelated donor bone marrow transplantation. Leuk Lymphoma 43:9, 2002. [PMID: 11908741]
740. Dazzi F, Goldman J: Donor lymphocyte infusions. Curr Opin Hematol 6:394, 1999. [PMID: 10546793]
741. Dazzi F, Szydlo RM, Cross NCP, et al: Durability of responses following donor lymphocyte infusions for patients who relapse after allogeneic stem cell transplantation for chronic myeloid leukemia. Blood 96:2712, 2000. [PMID: 11023502]
742. Dazzi F: Monitoring of minimal residual disease after allografting: A requirement to guide DLI treatment. Ann Hematol 81:S29, 2002.
743. Porter D, Levine JE: Graft-versus-host disease and graft-versus-leukemia after donor leukocyte infusion. Semin Hematol 43:53, 2006. [PMID: 16412789]
744. Makinnon S: Donor leukocyte infusions. Baillieres Clin Haematol 10:357, 1997.
745. Giralt S, Hester J, Huh T, et al: CD8-depleted donor lymphocyte infusion as treatment for relapsed chronic myelogenous leukemia after allogeneic bone marrow transplantation. Blood 86:4337, 1995. [PMID: 7492795]
746. Guglielma C, Arcese W, Dazzi F, et al: Donor lymphocyte infusion for relapsed chronic myelogenous leukemia: Prognostic relevance of the initial cell dose. Blood 100:397, 2002.
747. Verzeletti S, Bonini C, Marktel S, et al: Herpes simplex virus thymidine kinase gene transfer for controlled graft-versus-host disease and graft-versus-leukemia: Clinical follow-up and improved new vectors. Hum Gene Ther 9:2243, 1998. [PMID: 9794208]
748. Maravcova J, Nadvornikova S, Zmekova V, et al: Molecular monitoring of responses to DLI and DLI + IFN treatment of post-SCT relapse in patients with CML. Leuk Res 27:719, 2003.
749. Vela-Ojeda J, Garcia-Ruiz Esparza MA, Reyes-Maldonado E, et al: Donor lymphocyte infusions for relapse of chronic myeloid leukemia after allogeneic stem cell transplantation: Prognostic significance of the dose of CD3+ and CD4+ lymphocytes. Ann Hematol 83:295, 2004. [PMID: 15060749]
750. Savani BN, Montero A, Kurlander R, et al: Imatinib synergizes with donor lymphocyte infusions to achieve rapid molecular remission of CML relapsing after allogeneic stem cell transplantation. Bone Marrow Transplant 36:1009, 2005. [PMID: 16205732]
751. Porter DL, Antin JH: Donor leukocyte infusions in myeloid malignancies: New strategies. Best Pract Res Clin Haematol 19:737, 2006. [PMID: 16997180]
752. Kardinal CG, Bateman JR, Weiner J: Chronic myeloid leukemia. Review of 356 cases. Arch Intern Med 136:305, 1976. [PMID: 1062970]
753. Tura S, Baccarini M, Corbelli G: Staging of chronic myeloid leukemia. Br J Haematol 47:105, 1981. [PMID: 6934007]
754. Gomez GA, Sokal JE, Walsh D: Prognostic features at diagnosis of chronic myelogenous leukemia. Cancer 47:2470, 1981. [PMID: 6944141]
755. Cervantes F, Rozman C: A multivariate analysis of prognostic factors in chronic myeloid leukemia. Blood 60:1298, 1982. [PMID: 6958336]
756. Sokal JE, Cox EB, Baccarani M, et al: Prognostic discrimination in “good-risk” chronic granulocytic leukemia. Blood 63:789, 1984. [PMID: 6584184]
757. Sokal JE, Baccarini M, Tura S, et al: Prognostic discrimination among younger patients with chronic granulocytic leukemia: Relevance to bone marrow transplantation. Blood 66:1352, 1985. [PMID: 3904870]
758. Kantarjian HM, Keating MJ, Walters RS, et al: Clinical and prognostic features of Philadelphia chromosome-negative chronic myelogenous leukemia. Cancer 58:2023, 1986. [PMID: 3463397]
759. Sokal JE, Baccarini M, Russo D, Tura S: Staging and prognosis in chronic myelogenous leukemia. Semin Hematol 25:49, 1988. [PMID: 3279515]
760. Kantarjian HM, Keating MK, Smith TL, et al: Proposal for a single synthesis prognostic staging system in chronic myelogenous leukemia. Am J Med 88:1, 1990. [PMID: 2294759]
761. Dreazen I, Berman M, Gaoe RP: Molecular abnormalities of bcr and c-abl in chronic myelogenous leukemia associated with a long chronic phase. Blood 71:797, 1988. [PMID: 2449925]
762. Nowell PC, Jackson L, Weiss A, Kurzrock P: Historical communication: Philadelphia positive chronic myelogenous leukemia followed for 27 years. Cancer Genet Cytogenet 34:57, 1988. [PMID: 3165050]
763. Selleir L, Emilia G, Temperani P, et al: Philadelphia-positive chronic myelogenous leukemia with typical bcr/abl molecular features and atypical, prolonged survival. Leukemia 3:538, 1989.
764. Birnie GD, MacKenzie ED, Goyns MH, Pollock A: Sequestration of Philadelphia chromosome-positive cells in the bone marrow of a chronic myeloid leukemia patient in very prolonged remission. Leukemia 4:452, 1990. [PMID: 2359346]
765. Lamy TH, Dauriac C, Le Prise PY: Long-term survival in chronic granulocytic leukemia. Br J Haematol 73:279, 1989. [PMID: 2818949]
766. Johansson B, Martens F, Fioretos T, et al: Remarkably long survival of a patient with Ph1-positive chronic myeloid leukemia and 5/bcr rearrangement. Leukemia 4:448, 1990. [PMID: 2359344]
767. Singer CRJ, McDonald GA, Douglas AS: Twenty-five year survival of chronic granulocytic leukemia with spontaneous karyotype conversion. Br J Haematol 57:309, 1984. [PMID: 6587904]
768. Wodzinski MA, Potter AM, Lawence ACK: Prolonged survival in chronic granulocytic leukemia associated with loss of the Philadelphia chromosome. Br J Haematol 71:296, 1989. [PMID: 2923815]
769. Kantarjian HM, Smith TL, McCredie KB, et al: Chronic myelogenous leukemia: A multivariate analysis of the associations of patient characteristics and therapy with survival. Blood 66:1326, 1985. [PMID: 3864497]
770. Kantarjian HM, Talpaz M: Treatment of chronic myelogenous leukemia. Hematology 14:105, 1991.
771. Baccarini M, Russo D, Zuffa E, et al: The prognosis of chronic myeloid leukemia. Bone Marrow Transplant 1:126, 1989.
772. Hasford J, Pfirrmann M, Hockhaus A: How long will chronic myeloid leukemia patients treated with imatinib live? Leukemia 19:497, 2005. [PMID: 15861176]
773. Hasford J, Pfirrmann M, Hochhaus A: How long will chronic myeloid leukemia patients treated with imatinib mesylate live? Leukemia. 19:497, 2005. [PMID: 15861176]
774. O’Brien SG, Guilhot F, Goldman JM, et al: International randomized study of interferon versus STI571 (IRIS) 7-year follow-up: Sustained survival, low rate of transformation, and increased rate of major molecular response (MMR) in patients (pts) with newly diagnosed chronic myeloid leukemia in chronic phase (CML-CP) treated with imatinib (IM). Blood 112:76, 2008.
775. Druker BJ, Guilhot F, O’Brien SG, et al: Five-year follow-up of patients receiving imatinib for chronic myeloid leukemia. N Engl J Med 355:2408, 2006. [PMID: 17151364]
776. Rosti G, Iacobucci I, Bassi S, et al: Impact of age on the outcome of patients with chronic myeloid leukemia in late chronic phase: Results of a phase II study of the GIMEMA CML Working Party. Haematologica 92:101, 2007. [PMID: 17229641]
777. Kantarjian H, O’Brien S, Talpaz M, et al: Outcome of patients with Philadelphia chromosome-positive chronic Myelogenous leukemia post-imatinib mesylate failure. Cancer 109:1556, 2007. [PMID: 17342766]
778. Grossman A, Silver RT, Arlin Z, et al: Fine mapping of chromosome 22 breakpoints within the breakpoint cluster region (bcr) implies a role for bcr exon 3 in determining disease duration in chronic myeloid leukemia. Am J Hum Genet 45:729, 1989. [PMID: 2683759]
779. Morris SW, Daniel L, Ahmed CMI, et al: Relationship of bcr breakpoint to chronic phase duration, survival, and blast crisis lineage in chronic myelogenous leukemia patients presenting in early chronic phase. Blood 75:2035, 1990. [PMID: 2337672]
780. Giralt S, Kantarjian H, Talpaz M: The natural history of chronic myelogenous leukemia in the interferon era. Semin Hematol 32:152, 1995. [PMID: 7652582]
781. Smadja N, Krulik M, Audebert AA, et al: Spontaneous regression of cytogenetic and hematologic anomalies in Ph1-positive chronic myelogenous leukemia. Br J Haematol 63:257, 1986. [PMID: 3459548]
782. Musashi M, Abe S, Yamada T, et al: Spontaneous remission in a patient with chronic myelogenous leukemia. N Engl J Med 336:337, 1997. [PMID: 9011786]
783. Provan AB, Smith AG: Re-emergence of Philadelphia chromosome positive clone on a patient with previous spontaneous remission of chronic myeloid leukemia. Leukemia 9:1600, 1995. [PMID: 7658732]
784. Korkolopoulou P, Viniou N, Kavantzas N, et al: Clinicopathologic correlations of bone marrow angiogenesis in chronic myeloid leukemia: A morphometric study. Leukemia 17L:89, 2003.
785. Boultwood J, Peniket A, Watkins F, et al: Telomere length shortening in chronic myelogenous leukemia is associated with reduced time to accelerated phase. Blood 96:358, 2000. [PMID: 10891474]
786. Verstovsek S, Kantarjian H, Manshouri T, et al: Prognostic significance of cellular vascular endothelial growth factor expression in chronic phase chronic myeloid leukemia. Blood 99:2265, 2002. [PMID: 11877311]
787. Sinclair PB, Nacheva EP, Leversha M, et al: Large deletions at the t(9;22) breakpoint are common and may identify a poor-prognosis subgroup of patients with chronic myeloid leukemia. Blood 95:738, 2000. [PMID: 10648381]
788. Huntly BJP, Reid AG, Bench AJ, et al: Deletions of the derivative chromosome 9 occur at the time of the Philadelphia translocation and provide a powerful and independent prognostic indicator in chronic myeloid leukemia. Blood 98:1732, 2001. [PMID: 11535505]
789. Cohen N, Rozenfeld-Granot G, Hardan I, et al: Subgroup of patients with Philadelphia-positive chronic myelogenous leukemia characterized by a deletion of 9q proximal to ABL gene: Expression profiling, resistance to interferon therapy, and poor prognosis. Cancer Genet Cytogenet 128:114, 2001. [PMID: 11463449]
790. Kvasnicka HM, Thiele J, Schmitt-Graeff A, et al: Prognostic impact of bone marrow erythropoietic precursor cells and myelofibrosis at diagnosis of Ph1+ chronic myelogenous leukaemia—A multicentre study on 495 patients. Br J Haematol 112:727, 2001. [PMID: 11260078]
791. Normann AP, Egeland T, Madshus IH, et al: CD7 expression by CD34+ cells in CML patients, of prognostic significance? Eur J Haematol 71:266, 2003. [PMID: 12950236]
792. Roman-Gomez J, Castillejo JA, Jimenez A, et al: Cadherin-13, a mediator of calcium-dependent cell-cell adhesion, is silenced by methylation in chronic myeloid leukemia and correlates with pretreatment risk profile and cytogenetic response to interferon alfa. J Clin Oncol 21:1472, 2003. [PMID: 12697869]
793. Bhatia R, Holtz M, Niu N, et al: Persistence of malignant hematopoietic progenitors in chronic myelogenous leukemia patients in complete cytogenetic remission following imatinib mesylate treatment. Blood 101:4701, 2003. [PMID: 12576334]
794. Morel F, Ka C, Le Bris MJ, et al: Deletion of the 5′ ABL region in Philadelphia chromosome positive chronic myeloid leukemia: Frequency, origin and prognosis. Leuk Lymphoma 44:1333, 2003. [PMID: 12952226]
795. Prejzner W: Relationship of the BCR gene breakpoint and the type of BCR/ABL transcript to clinical course, prognostic indexes and survival in patients with chronic myeloid leukemia. Med Sci Monit 8:193, 2002.
796. Bauduer F, Ducout L, Dastugue N, Marolleau JP: Chronic myeloid leukemia as a secondary neoplasm after anti-cancer radiotherapy: A report of three cases and a brief review of the literature. Leuk Lymphoma 43:1057, 2002. [PMID: 12148886]
797. Agis H, Sperr WR, Herndlhofer S, et al: Clinical and prognostic significance of histamine monitoring in patients with CML during treatment with imatinib (STI571). Ann Oncol 18:1834, 2007. [PMID: 17804468]
798. Passweg JR, Walker I, Sobocinski KA, et al: Validation and extension of the EBMT Risk Score for patients with chronic myeloid leukemia (CML) receiving allogeneic haematopoietic stem cell transplants. Br J Haematol 125:613, 2004. [PMID: 15147377]
799. Hasford J, Pfirrmann M, Hehlmann R, et al: Prognosis and prognostic factors for patients with chronic myeloid leukemia: Nontransplant therapy. Semin Hematol 40:4, 2003. [PMID: 12563607]
800. Qazilbash MH, Devetten MP, Abraham J, et al: Utility of a prognostic scoring system for allogeneic stem cell transplantation in patients with chronic myeloid leukemia. Acta Haematol 109:119, 2003. [PMID: 12714820]
801. Usman M, Syed NN, Kakepoto GN, et al: Chronic phase chronic myeloid leukemia: Response of imatinib mesylate and significance of Sokal score, age and disease duration in predicting the hematological and cytogenetic response. J Assoc Physicians India 55:103, 2007. [PMID: 17571738]
802. Marin D, Marktel S, Bua M, et al: Prognostic factors for patients with chronic myeloid leukemia in chronic phase treated with imatinib mesylate after failure of interferon alfa. Leukemia 17:1448, 2003. [PMID: 12886230]
803. Kantarjian HM, O’Brien S, Cortes JE, et al: Complete cytogenetic and molecular responses to interferon-alpha-based therapy for chronic myelogenous leukemia are associated with excellent long-term prognosis. Cancer 97:1033, 2003. [PMID: 12569603]
804. Rosti G, Martinelli G, Bassi S, et al: Molecular response to imatinib in late chronic-phase chronic myeloid leukemia. Blood 103:2284, 2004. [PMID: 14645009]
805. Yee K, Anglin P, Keating A: Molecular approaches to the detection and monitoring of chronic myeloid leukemia: Theory and practice. Blood Rev 13:105, 1999. [PMID: 10414946]
806. Kaeda J, Chase A, Goldman JM: Cytogenetic and molecular monitoring of residual disease in chronic myeloid leukaemia. Acta Haematol 107:64, 2002. [PMID: 11919387]
807. Hughes T, Branford S: Molecular monitoring of chronic myeloid leukemia. Semin Hematol 40:62, 2003. [PMID: 12783378]
808. Kantarjian H, Schiffer C, Jones D, Cortes J: Monitoring the response and course of chronic myeloid leukemia in the modern era of BCR-ABL tyrosine kinase inhibitors: Practical advice on the use and interpretation of monitoring methods. Blood 111(4):1774, 2008. [PMID: 18055868]
809. Lowenberg B: Minimal residual disease in chronic myeloid leukemia. N Engl J Med 349:1399, 2003. [PMID: 14534331]
810. Gabert J: Detection of recurrent translocations using real time PCR; assessment of the technique for diagnosis and detection of minimal residual disease. Haematologica 84:107, 1999. [PMID: 10907485]
811. Negrin RS, Blume KG: The use of polymerase chain reaction for the detection of minimal residual malignant disease. Blood 78:255, 1991. [PMID: 2070062]
812. Lee M-S, Kantarjian H, Talpaz M, et al: Detection of minimal residual disease by polymerase chain reaction in Philadelphia chromosome-positive chronic myelogenous leukemia following interferon therapy. Blood 79:1920, 1992. [PMID: 1562719]
813. Schulze E, Krahl R, Thalmeier K, Helbig W: Detection of bcr-abl mRNA in single progenitor colonies from patients with chronic myeloid leukemia by PCR: Comparison with cytogenetics and PCR from uncultured cells. Exp Hematol 23:1649, 1995. [PMID: 8542960]
814. Lin F, Chase A, Bungey J, et al: Correlation between the proportion of Philadelphia chromosome-positive metaphase cells and levels of BCR-ABL mRNA in chronic myeloid leukaemia. Genes Chromosomes Cancer 13:110, 1995. [PMID: 7542905]
815. Thompson JD, Brodsky I, Yunis JJ: Molecular quantification of residual disease in chronic myelogenous leukemia after bone marrow transplantation. Blood 79:1629, 1992. [PMID: 1547352]
816. Lin F, Chase A, Bungey J, et al: Correlation between the proportion of Philadelphia chromosome-positive metaphase cells and levels of BCR-ABL mRNA in chronic myeloid leukaemia. Genes Chromosomes Cancer 18:110, 1995.
817. Branford S, Fletcher L, Cross NC, et al: Desirable performance characteristics for BCR-ABL measurement on an international reporting scale to allow consistent interpretation of individual patient response and comparison of response rates between clinical trials. Blood 112:3330, 2008. [PMID: 18684859]
818. Sahay T, Schiffer CA: Monitoring minimal residual disease in patients with chronic myeloid leukemia after treatment with tyrosine kinase inhibitors. Curr Opin Hematol 15:134, 2008. [PMID: 18300760]
819. Stock W, Yu D, Karrison T, et al: Quantitative real-time RT-PCR monitoring of BCR-ABL in chronic Myelogenous leukemia shows lack of agreement in blood and bone marrow samples. Int J Oncol 28:1099, 2006. [PMID: 16596225]
820. Gaiger A, Henn T, Horth E, et al: Increase of bcr-abl chimeric mRNA expression in tumor cells of patients with chronic myeloid leukemia precedes disease progression. Blood 86:2371, 1995. [PMID: 7662984]
821. Guo JQ, Lin H, Kantarjian H, et al: Comparison of competitive-nested PCR and real-time PCR in detecting BCR-ABL fusion transcripts in chronic myeloid leukemia patients. Leukemia 16:2447, 2002. [PMID: 12454751]
822. Iacobucci I, Saglio G, Rosti G, et al: Achieving a major molecular response at the time of a complete cytogenetic response (CCgR) predicts a better duration of CCgR in imatinib-treated chronic myeloid leukemia patients. Clin Cancer Res 12:3037, 2006. [PMID: 16707599]
823. Press RD, Love Z, Tronnes AA, et al: BCR-ABL mRNA levels at and after the time of a complete cytogenetic response (CCR) predict the duration of CCR in imatinib mesylate-treated patients with CML. Blood 107:4250, 2006. [PMID: 16467199]
824. Jabbour E, Cortes JE, Kantarjian HM: Molecular monitoring in chronic myeloid leukemia: Response to tyrosine kinase inhibitors and prognostic implications. Cancer 112:2112, 2008. [PMID: 18348294]
825. O’Dwyer ME: How to monitor patients with chronic myelogenous leukemia. JNCCN 1:513, 2003. [PMID: 19774742]
826. Landstrom AP, Tefferi A: Fluorescent in situ hybridization in the diagnosis, prognosis, and treatment monitoring of chronic myeloid leukemia. Leuk Lymphoma 47:397, 2006. [PMID: 16396761]
827. Cohen N, Novikov I, Hardan I, et al: Standardization criteria for the detection of BCR/ABL fusion in interphase nuclei of chronic myelogenous leukemia patients by fluorescence in situ hybridization. Cancer Genet Cytogenet 123:102, 2000. [PMID: 11150599]
828. Rheinhold U, Hennig E, Leiblein S, et al: FISH for BCR-ABL on interphases of peripheral blood neutrophils but not of unselected white cells correlates with bone marrow cytogenetics in CML patients treated with imatinib. Leukemia 17:1925, 2003.
829. Kim YJ, Kim DW, Lee S, et al: Comprehensive comparison of FISH, RT-PCR, and RQ-PCR for monitoring the BCR-ABL gene after hematopoietic stem cell transplantation in CML. Eur J Haematol 68:272, 2002. [PMID: 12144533]
830. Bao F, Munker R, Lowery C, et al: Comparison of FISH and quantitative RT-PCR for the diagnosis and follow-up of BCR-ABL-positive leukemias. Mol Diagn Ther 11:239, 2007. [PMID: 17705578]
831. Uzunel M, Mattsson J, Brune M, et al: Kinetics of minimal residual disease and chimerism in patients with chronic myeloid leukemia after nonmyeloablative conditioning and allogeneic stem cell transplantation. Blood 101:469, 2003. [PMID: 12393598]
832. Hochhaus A, Weisser A, LaRosee P, et al: Detection and quantification of residual disease in chronic myelogenous leukemia. Leukemia 14:998, 2000. [PMID: 10865964]
833. Olavarria E, Kanfer E, Szydlo R, et al: Early detection of BCR-ABL transcripts by quantitative reverse transcriptase-polymerase chain reaction predicts outcome after allogeneic stem cell transplantation for chronic myeloid leukemia. Blood 97:1560, 2001. [PMID: 11238091]
834. Radich JP, Gooley T, Bryant E, et al: The significance of bcr-abl molecular detection in chronic myeloid leukemia patients “late” 18 months or more after transplantation. Blood 98:1701, 2001. [PMID: 11535500]
835. Lion T: Minimal residual disease. Curr Opin Hematol 6:406, 1999. [PMID: 10546795]
836. Thiele J, Wickenhauser C, Kvasnicka HM, et al: Mixed chimerism of bone marow CD34+ progenitor cells (genotyping, bcr/abl analysis) after allogeneic transplantation for chronic myelogenous leukemia. Transplantation 74:982, 2002. [PMID: 12394842]
837. Serrano J, Roman J, Sanchez J, et al: Molecular analysis of lineage specific chimerism and minimal residual disease by RT-PCR of p210 (BCR-ABL) and p190 (BCR-ABL) after allogeneic bone marrow transplantation for chronic myeloid leukemia: Increasing mixed myeloid chimerism and p190 (BCR-ABL) detection precede cytogenetic relapse. Blood 15:2659, 2000.
838. Muller MC, Gattermann N, Lahaye T, et al: Dynamics of BCR-ABL mRNA expression in first-line therapy of chronic myelogenous leukemia patients with imatinib or interferon alpha/ara-C. Leukemia 17:2392, 2003. [PMID: 14523462]
839. Hochhaus A: Minimal residual disease in chronic myeloid leukaemia patients. Best Pract Res Clin Haematol 15:159, 2002. [PMID: 11987922]
840. Lin F, Drummond M, O’Brien S, et al: Molecular monitoring in chronic myeloid leukemia patients who achieve complete cytogenetic remission on imatinib. Blood 102:1143, 2003. [PMID: 12869491]
841. Ross DM, Watkins DB, Hughes TP, Branford S: Reverse transcription with random pentadecamer primers improves the detection limit of a quantitative PCR assay for BCR-ABL transcripts in chronic myeloid leukemia: Implications for defining sensitivity in minimal residual disease. Clin Chem 54:1568, 2008. [PMID: 18641042]
842. Sun X, Li J, Chen J, et al: Flow cytometric assay of phosphotyrosine levels in Bcr-Abl-positive chronic myelogenous leukemias: A potential prognostic marker. Ann Hematol 88:29, 2009. [PMID: 18696070]
843. Giles FJ, Cortes JE, Kantarjian HM, O’Brien S: Accelerated and blastic phase of chronic myelogenous leukemia. Hematol Oncol Clin North Am 18:753, 2004. [PMID: 15271404]
844. Jamieson CH, Ailles LE, Dylla SJ, et al: Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. N Engl J Med 351:657, 2004. [PMID: 15306667]
845. Michor F: Chronic myeloid leukemia blast crisis arises from progenitors. Stem Cells 25:1114, 2007. [PMID: 17218393]
846. Wodarz D: Stem cell regulation and the development of blast crisis in chronic myeloid leukemia: Implications for the outcome of Imatinib treatment and discontinuation. Med Hypotheses 70:128, 2008. [PMID: 17566666]
847. Melo JV, Barnes DJ: Chronic myeloid leukaemia as a model of disease evolution in human cancer. Nat Rev Cancer 7:441, 2007. [PMID: 17522713]
848. Brazma D, Grace C, Howard J, et al: Genomic profile of chronic myelogenous leukemia: Imbalances associated with disease progression. Genes Chromosomes Cancer 46:1039, 2007. [PMID: 17696194]
849. Gaiger A, Henn T, Horth E, et al: Increase of bcr/abl chimeric mRNA expression in tumor cells of patients with chronic myeloid leukemia precedes disease progression. Blood 86:2371, 1995. [PMID: 7662984]
850. Elmaaglacli AH, Beelen DW, Opalka B, et al: The amount of BCR/ABL fusion transcripts detected by real-time quantitative polymerase chain reaction method in patients with Philadelphia chromosome positive chronic myeloid leukemia disease stages correlates with the disease stages. Ann Hematol 79:424, 2000.
851. Lowenberg B, Hagemeijer A, Swart K, Abels J: Serial follow-up of patients with chronic myeloid leukemia (CML) with combined cytogenetic and colony culture methods. Exp Hematol 10:123, 1982.
852. Haas OA, Schwarzmeier JD, Nachera E, et al: Investigations on karyotype evolution in patients with chronic myeloid leukemia (CML). Blut 48:33, 1984. [PMID: 6580930]
853. Swolin B, Weinfeld A, Westin J, et al: Karyotypic evolution in Ph-positive chronic myeloid leukemia in relation to management and disease progression. Cancer Genet Cytogenet 18:65, 1985. [PMID: 3861236]
854. Cortes J, O’Dwyer ME: Clonal evolution in chronic myelogenous leukemia. Hematol Oncol Clin North Am 18:671, 2004. [PMID: 15271399]
855. Honda H, Ushijima K, Oda H, et al: Acquired loss of p53 induces blast transformation in p210bcr/abl-expressing hematopoietic cells: A transgenic study for blast crisis in human CML. Blood 95:1144, 2000. [PMID: 10666183]
856. Yamaguchi H, Inokuchi K, Sakuma Y, Dan K: Mutation of p51/p63 gene is associated with blast crisis in chronic myelogenous leukemia. Leukemia 15:1729, 2001. [PMID: 11681414]
857. Coiffier B, Byron PA, Flere D, et al: Chronic granulocytic leukemia: Early detection of metamorphosis with “in vitro” culture of granulocytic progenitors. Biomedicine 33:96, 1980. [PMID: 6932975]
858. Todd MB, Waldron JA, Jennings TA, et al: Loss of myeloid differentiation antigens precedes blastic transformation in chronic myelogenous leukemia. Blood 70:122, 1987. [PMID: 2439147]
859. Grinesshammer M, Heinze B, Bangerter M, et al: Karyotype abnormalities and their clinical significance in blast crisis of chronic myeloid leukemia. J Mol Med 75:8836, 1997.
860. Spencer A, Vulliamy T, Kaeda J, et al: Clonal instability preceding lymphoid blastic transformation of chronic myeloid leukemia. Leukemia 11:195, 1997. [PMID: 9009080]
861. Anastasi J, Feng J, LeBeau MM, et al: The relationship between secondary chromosomal abnormalities and blast transformation in chronic myelogenous leukemia. Leukemia 9:628, 1995. [PMID: 7723396]
862. Bartram CR, de Klein A, Hagemeijer A, et al: Additional C-abl/bcr rearrangements in a CML patient exhibiting two Ph1 chromosomes during blast crisis. Leuk Res 10:221, 1986. [PMID: 3005778]
863. Collins SJ, Grudine MT: Chronic myelogenous leukemia: Amplification of a rearranged c-abl oncogene in both chronic phase and blast crisis. Blood 69:893, 1987. [PMID: 3545323]
864. Mughal TI, Goldman JM: Chronic myeloid leukemia: Why does it evolve from chronic phase to blast transformation? Front Biosci 11:198, 2006. [PMID: 16146725]
865. Schaefer-Rego K, Dudik H, Popenoe D, et al: CML patients in blast crisis have breakpoints localized to a specific region of the bcr. Blood 70:448, 1987. [PMID: 3038213]
866. Mills KI, Benn P, Birnie GD: Does the breakpoint within the major breakpoint region (M-bcr) influence the duration of the chronic phase in chronic myeloid leukemia? An analytical comparison of current literature. Blood 78:1155, 1991. [PMID: 1878582]
867. Bartram CR, Janssen JWG, Becher R, et al: Persistence of chronic myelocytic leukemia despite deletion of rearranged bcr/c-abl sequences in blast crisis. J Exp Med 164:1389, 1986. [PMID: 3464689]
868. Okabe M, Matsushima S: Philadelphia chromosome-positive leukemia: Molecular analysis of bcr and abl genes and transforming genes. Nippon Ketsueki Gakkai Zasshi 51:1471, 1988. [PMID: 3247820]
869. Ahuja H, Bar-Eli M, Arlin Z, et al: The spectrum of molecular alterations in the evolution of chronic myelocytic leukemia. J Clin Invest 87:2042, 1991. [PMID: 2040694]
870. Kelman Z, Prokocimer M, Peller S, et al: Rearrangements in the p53 gene in Philadelphia chromosome positive chronic myelogenous leukemia. Blood 74:2318, 1989. [PMID: 2572286]
871. Mashal R, Shtalrid M, Talpaz M, et al: Rearrangement and expression of p53 in the chronic phase and blast crisis of chronic myelocytic leukemia. Blood 75:180, 1990. [PMID: 1967214]
872. Guinn BA, Mello KI: P53 mutations, methylation and genomic instability in the progression of chronic myeloid leukemia. Leuk Lymphoma 26:241, 1997.
873. Malinen T, Palotie A, Pakkala S, et al: Acceleration of chronic myeloid leukemia correlates with calcitonin gene methylation. Blood 77:2435, 1991. [PMID: 2039824]
874. Asimakopoolos FA, Shteper PJ, Krichevsky S, et al: ABL1 methylation is a distinct molecular event associated with clonal evolution of chronic myeloid leukemia. Blood 94:2452, 1999.
875. Towatari M, Adachi K, Kato H, Saito H: Absence of the human retinoblastoma gene product in the megakaryoblastic crisis of chronic myelogenous leukemia. Blood 78:2178, 1991. [PMID: 1932738]
876. Sill H, Goldman JM, Cross NC: Homozygous deletions of the p16 tumor-suppressor gene are associated with lymphoid transformation of chronic myeloid leukemia. Blood 85:2013, 1995. [PMID: 7718873]
877. Hernandez-Boluda J-C, Cervantes F, Colomer D, et al: Genomic p16 abnormalities in the progression of chronic myeloid leukemia into blast crisis. Exp Hematol 31:204, 2003. [PMID: 12644017]
878. Serra A, Gottardi E, Della Ragione F, et al: Involvement at the cyclin-dependent kinase-4 inhibitor (CDKN2) gene in the pathogenesis of lymphoid blast crisis of chronic myelogenous leukaemia. Br J Haematol 91:625, 1995. [PMID: 8555065]
879. Menssen HD, Renki JMJ, Rodeck U, et al: Presence of Wilms’ tumor gene (wt1) transcripts and the WT1 nuclear protein in the majority of human acute leukemias. Leukemia 9:1060, 1995. [PMID: 7596170]
880. Mitarri K, Ogawa S, Tanaka T, et al: Generation of the AML1-EVI-1 fusion gene in the t(3;21) (q26;q22) causes blastic crisis in chronic myelocytic leukemia. EMBO J 13:504, 1994.
881. Carapeti M, Goldman JM, Cross NC: Overexpression of EV-l in blast crisis of chronic myeloid leukemia. Leukemia 10:1561, 1996. [PMID: 8751483]
882. Mori N, Takeuchi S, Tasaka T, et al: Absence of microsatellite instability during the progression of chronic myelocytic leukemia. Leukemia 11:151, 1997. [PMID: 9001431]
883. Handa H, Hegde UP, Kuteninikov VM, et al: Bcl-2 and c-myc expressions, cell cycle kinetics and apoptosis during the progression of chronic myelogenous leukemia from diagnosis to blastic phase. Leuk Res 21:479, 1997. [PMID: 9279359]
884. Daheron L, Salmeron S, Patri S, et al: Identification of several genes differentially expressed during progression of chronic myelogenous leukemia. Leukemia 12:326, 1998. [PMID: 9529126]
885. Foti A, Ahuja HG, Allen SL, et al: Correlation between molecular and clinical events in the evolution of chronic myelocytic leukemia to blast crisis. Blood 77:2441, 1991. [PMID: 2039825]
886. Mori N, Morosetti R, Loe S, et al: Allelotype analysis in the evolution of chronic myelocytic leukemia. Blood 90:2010, 1997. [PMID: 9292536]
887. Radich JP, Dai H, Mao M, et al: Gene expression changes associated with progression and response in chronic myeloid leukemia. Proc Natl Acad Sci U S A 103:2794, 2006. [PMID: 16477019]
888. Yong AS, Szydlo RM, Goldman JM, et al: Molecular profiling of CD34+ cells identifies low expression of CD7, along with high expression of proteinase 3 or elastase, as predictors of longer survival in patients with CML. Blood 107:205, 2006. [PMID: 16144796]
889. Spiers ASD: Metamorphosis of chronic granulocytic leukemia: Diagnosis, classification and management. Br J Haematol 49:1, 1979.
890. Grignani F: Chronic myelogenous leukemia. Crit Rev Oncol Hematol 4:31, 1985. [PMID: 3902263]
891. Matsuo T, Tomonaga M, Kuriyama K, et al: Prognostic significance of the morphological dysplastic changes in chronic myelogenous leukemia. Leuk Res 10:331, 1986. [PMID: 3456479]
892. Ishii N, Murakami H, Matsushima T, et al: Histamine excess symptoms in basophilic crisis of chronic myelogenous leukemia. J Med 26:235, 1995. [PMID: 8721900]
893. Specchia G, Palumbo G, Pastore D, et al: Extramedullary blast crisis in chronic myeloid leukemia. Leuk Res 20:905, 1996. [PMID: 9009247]
894. Inveradi D, Lazzarino M, Morra E, et al: Extramedullary disease in Ph-positive chronic myelogenous leukemia: Frequency, clinical features, prognostic significance. Haematologica 75:146, 1990.
895. Jacknow J, Fizzera G, Gajl-Peczalska K, et al: Extramedullary presentation of the blast crisis of chronic myelogenous leukemia. Br J Haematol 61:225, 1985. [PMID: 3862425]
896. Terjanian T, Kantarjian H, Keating M, et al: Clinical and prognostic features of patients with Philadelphia chromosome-positive chronic myelogenous leukemia and extramedullary disease. Cancer 59:297, 1987. [PMID: 3467820]
897. Miksanek T, Reyes CV, Semkuo Z, Molnar ZJ: Granulocytic sarcoma of the peritoneum. CA Cancer J Clin 33:40, 1983. [PMID: 6401575]
898. Jones TI: Pleural blast crisis in chronic myelogenous leukemia. Am J Hematol 44:75, 1993. [PMID: 8342572]
899. Pascoe HR: Tumors composed of immature granulocytes occurring in the breast in chronic granulocytic leukemia. Cancer 25:697, 1970. [PMID: 4906722]
900. Chabner BA, Haskell CM, Canellos GP: Destructive bone lesions in chronic granulocytic leukemia. Medicine (Baltimore) 48:401, 1969. [PMID: 5266365]
901. Licht A, Many N, Rachmilewitz EA: Myelofibrosis, osteolytic bone lesions and hypercalcemia in chronic myeloid leukemia. Acta Haematol 49:182, 1973. [PMID: 4198453]
902. Lee CH, Morris TCM: Bone marrow necrosis and extramedullary myeloid tumor necrosis in aggressive chronic myeloid leukemia. Pathology 11:551, 1979. [PMID: 293601]
903. Asarro S, Sato N, Ueshima Y, et al: Localized blastoma preceding blastic transformation in Ph1-positive chronic myelogenous leukemia. Scand J Haematol 25:251, 1980.
904. Ohyashiki K, Ito H: Characterization of extramedullary tumors in a case of Ph-positive chronic myelogenous leukemia. Cancer Genet Cytogenet 15:119, 1985. [PMID: 3871350]
905. Sun T, Susin M, Koduru P, et al: Extramedullary blast crisis in chronic myelogenous leukemia. Cancer 68:605, 1991. [PMID: 2065281]
906. Saikia TK, Dhabhar B, Iyer RS, et al: High incidence of meningeal leukemia in lymphoid blast crisis of chronic myelogenous leukemia. Am J Hematol 43:10, 1993. [PMID: 8317457]
907. Falini B, Tabilio A, Pelicci PG, et al: T-cell receptor B-chain gene rearrangement in a case of Ph1-positive chronic myeloid leukaemia blast crisis. Br J Haematol 62:776, 1986. [PMID: 3083860]
908. Giannone L, Whitlock JA, Kinney MC, et al: Use of the BCR probe to demonstrate extramedullary recurrence of CML with a T cell lymphoid phenotype following bone marrow transplantation. Bone Marrow Transplant 3:631, 1988. [PMID: 3063330]
909. Ohyashiki J, Ohyashiki K, Shimizu H, et al: Testicular tumor as the first manifestation of B-lymphoid blastic crisis in a case of Ph-positive chronic myelogenous leukemia. Am J Hematol 29:164, 1988. [PMID: 3263797]
910. Rosenthal S, Canellos GP, DeVita VT, Gralnick HR: Characteristics of blast crisis in chronic granulocytic leukemia. Blood 49:705, 1977. [PMID: 265737]
911. Barton JC, Conrad ME: Current status of blastic transformation in chronic myelogenous leukemia. Am J Hematol 4:281, 1978. [PMID: 362906]
912. Peterson LC, Bloomfield CD, Brunning RD: Blast crisis as an initial or terminal manifestation of chronic myeloid leukemia. Am J Med 60:209, 1976. [PMID: 1062162]
913. Bettelheim P, Lutz D, Majdic O, et al: Cell lineage heterogeneity in blast crisis of chronic myeloid leukaemia. Br J Haematol 59:395, 1985. [PMID: 3855651]
914. Nair C, Chopra M, Shinde S, et al: Immunophenotype and ultrastructural studies in blast crisis of chronic myeloid leukemia. Leuk Lymphoma 19:309, 1995. [PMID: 8535224]
915. Rosenthal S, Canellos GP, Gralnick HR: Erythroblastic transformation of chronic granulocytic leukemia. Am J Med 63:116, 1977. [PMID: 267430]
916. Ekblom M, Borgstrom G, von Willebrand E, et al: Erythroid blast crisis in chronic myelogenous leukemia. Blood 62:591, 1983. [PMID: 6576813]
917. Lingg G, Schmalzl F, Breton-Gorius J, et al: Megakaryoblastic micro-megakaryocytic crisis in chronic myeloid leukemia. Blut 51:275, 1985. [PMID: 3863678]
918. Castaigne S, Berger R, Jolly V, et al: Promyelocytic blast crisis of chronic myelocytic leukemia with both t(9;22) and t(15;17) in M3 cells. Cancer 54:2409, 1984. [PMID: 6594186]
919. Berger R, Bernheim A, Daniel MT, Flandrin G: T(15;17) in a promyelocytic form of chronic myeloid leukemia blastic crisis. Cancer Genet Cytogenet 8:149, 1983. [PMID: 6572090]
920. Misawa S, Lee E, Schiffer CA, et al: Association of translocation (15;17) with malignant proliferation of promyelocytes in acute leukemia and chronic myelogenous leukemia in blast crisis. Blood 67:270, 1986. [PMID: 3455826]
921. Marinone G, Rossi G, Verzura P: Eosinophilic blast crisis in a case of chronic myeloid leukaemia. Br J Haematol 55:251, 1983. [PMID: 6577909]
922. Goh K-O, Anderson FW: Cytogenetic studies in basophilic chronic myelocytic leukemia. Arch Pathol Lab Med 103:288, 1979. [PMID: 287415]
923. Rosenthal NS, Knapp D, Farhi DC: Promyelocytic blast crisis of chronic myelogenous leukemia. A rare subtype associated with disseminated intravascular coagulation. Am J Clin Pathol 103:185, 1995. [PMID: 7856560]
924. Lemes A, Gomez Casares MT, de la Iglesia S, et al: P190 BCR-ABL rearrangement in chronic myeloid leukemia and acute lymphoblastic leukemia. Cancer Genet Cytogenet 113:100, 1999. [PMID: 10459357]
925. Bertazzoni U, Brusamolino E, Isernia P, et al: Diagnostic significance of terminal transferase and adenosine deaminase in acute and chronic myeloid leukemia. Blood 60:685, 1982. [PMID: 7049266]
926. Schuh AC, Sutherland DR, Horsfall W, et al: Chronic myeloid leukemia arising in a progenitor common to T cells and myeloid cells. Leukemia 4:631, 1990. [PMID: 2168506]
927. Uike N, Takeichi N, Kimura N, et al: Dual arrangement of immunoglobulin and T-cell receptor genes in blast crisis of CML. Eur J Haematol 42:460, 1989. [PMID: 2543592]
928. Greaves MF, Verbi W, Reeves, BR, et al: “Pre-B” phenotypes in blast crisis of Ph1 positive CML: Evidence for a pluripotential stem cell “target.” Leuk Res 3:181, 1979. [PMID: 316485]
929. Bakhshi A, Minowada J, Arnold A, et al: Lymphoid blast crisis of chronic myelogenous leukemia represents stages in the development of B-cell precursors. N Engl J Med 309:826, 1983. [PMID: 6412140]
930. Griffin JD, Todd RF, Ritz J, et al: Differentiation patterns in the blastic phase of chronic myeloid leukemia. Blood 61:85, 1983. [PMID: 6571717]
931. Bollum FJ: Terminal deoxynucleotidyl transferase, in The Enzymes, edited by RD Boyer, p 145. Academic, New York, 1974.
932. Dorfman DM, Longtine JA, Fox EA, et al: T-cell blast crisis in chronic myelogenous leukemia. Am J Clin Pathol 107:168, 1997. [PMID: 9024065]
933. Allouche M, Bourinbaiar A, Georgoulias V, et al: T-cell lineage involvement in lymphoid blast crisis of chronic myeloid leukemia. Blood 66:1155, 1985. [PMID: 3876856]
934. Gramatzki M, Bartram CR, Muller D, et al: Early T-cell differentiated chronic myeloid leukemia blast crisis with rearrangement of the breakpoint cluster region but not of the T-cell receptor beta chain genes. Blood 69:1082, 1987. [PMID: 3103711]
935. Dastugue N, Kuhlein E, Duchayne E, et al: T(14:14)(q11;q32) in biphenotypic blastic phase of chronic myeloid leukemia. Blood 68:949, 1986. [PMID: 3489495]
936. Kuriyama K, Tomonaga M, Yao E, et al: Dual expression of lymphoid/basophil markers on single blast cells transformed from chronic myeloid leukemia. Leuk Res 10:1015, 1986. [PMID: 3462436]
937. Yasukawa M, Iwamasa K, Kawamura S, et al: Phenotypic and genotypic analysis of chronic myelogenous leukaemia with T lymphoblastic and megakaryoblastic mixed crisis. Br J Haematol 66:331, 1987. [PMID: 3497665]
938. Spencer A, Vulliamy T, Chase A, et al: Myeloid to lymphoid clonal suppression following autologous transplantation in second chronic phase of chronic myeloid leukemia. Leukemia 9:2138, 1995. [PMID: 8609730]
939. Cervantes F, Villamor N, Esteve J, et al: “Lymphoid” blast crisis of chronic myeloid leukaemia is associated with distinct clinicohaematological features. Br J Haematol 100:123, 1998. [PMID: 9450800]
940. Stoll C, Oberline F: Non-random clonal evolution in 45 cases of chronic myeloid leukemia. Leuk Res 46:61, 1980.
941. Sandberg AA: The cytogenetics of chronic myelocytic leukemia (CML): Chronic phase and blastic crisis. Cancer Genet Cytogenet 1:217, 1980.
942. Myint H, Ross FM, Hall JL, et al: Early transformation to acute myeloblastic leukaemia with the acquisition of inv(16) in Ph positive chronic granulocytic leukaemia. Leuk Res 21:473, 1997. [PMID: 9225078]
943. Johansson B, Fioretos T, Mitelman F: Cytogenetic and molecular genetic evolution of chronic myeloid leukemia. Acta Haematol 107:76, 2002. [PMID: 11919388]
944. Sandberg AA: Chronic myelocytic leukemia, in The Chromosomes in Human Cancer and Leukemia, 2nd ed, p 465. Elsevier North Holland, New York, 1990.
945. Mitani K, Miyazono K, Urabe A, Takaku F: Karyotypic changes during the course of blastic crisis of chronic myelogenous leukemia. Cancer Genet Cytogenet 39:299, 1989. [PMID: 2752380]
946. Diez-Martin JL, DeWald GW, Pierre RV, et al: Possible cytogenetic distinction between lymphoid and myeloid blast crisis in chronic granulocytic leukemia. Am J Hematol 27:194, 1988. [PMID: 3279762]
947. Feinstein E, Cimino G, Gale RP, Canaani E: Initiation and progression of chronic myelogenous leukemia. Leukemia 6(Suppl 1):37, 1992.
948. Brizard F, Cividin M, Villalva C, et al: Comparison of M-FISH and conventional cytogenetic analysis in accelerated and acute phases of CML. Leuk Res 28:345, 2004. [PMID: 15109532]
949. Heim S, Christensen EB, Fioretos T, et al: Acute myelomonocytic leukemia with inv(16) (p13q22) complicating Philadelphia chromosome positive chronic myeloid leukemia. Cancer Genet Cytogenet 59:35, 1992. [PMID: 1555189]
950. Hogge DE, Misawa S, Testa JR, et al: Unusual karyotypic changes and B-cell involvement in a case of lymph node blast crisis of chronic myelogenous leukemia. Blood 64:123, 1984. [PMID: 6610445]
951. Kantarjian H, Talpaz M, O’Brien S, et al: Survival benefit with imatinib mesylate therapy in patients with accelerated-phase chronic myelogenous leukemia—Comparison with historic experience. Cancer 103:2099, 2005. [PMID: 15830345]
952. Shah NP: Advanced CML: Therapeutic options for patients in accelerated and blast phases. J Natl Compr Canc Netw. 6:S31, 2008.
953. Oki Y, Kantarjian HM, Gharibyan V, et al: Phase II study of low-dose decitabine in combination with imatinib mesylate in patients with accelerated or myeloid blastic phase of chronic Myelogenous leukemia. Cancer 109:899, 2007. [PMID: 17236224]
954. Fruehauf S, Topaly J, Buss EC, et al: Imatinib combined with mitoxantrone/etoposide and cytarabine is an effective induction therapy for patients with chronic myeloid leukemia in myeloid blast crisis. Cancer 109:1543, 2007. [PMID: 17340589]
955. Cortes J, Kantarjian H: Advanced-phase chronic myeloid leukemia. Semin Hematol 40:79, 2003. [PMID: 12563614]
956. Druker BJ, Sawyers CL, Kantarjian H, et al: Activity of a specific inhibitor of the BCR-ABL tyrosine kinase in the blast crisis of chronic myeloid leukemia and acute lymphoblastic leukemia with the Philadelphia chromosome. N Engl J Med 344:1038, 2001. [PMID: 11287973]
957. Altintas A, Cil T, Kilinc I, et al: Central nervous system blastic crisis in chronic myeloid leukemia on imatinib mesylate therapy: A case report. J Neurooncol 84:103, 2007. [PMID: 17318411]
958. Simpson E, O’Brien SG, Reilly JT: Extramedullary blast crises in CML patients in complete hematological remission treated with imatinib mesylate. Clin Lab Haematol 28:215, 2006. [PMID: 16706941]
959. Gozzetti A, Bocchia M, Calabrese S, et al: Promyelocytic blast crisis of chronic myelogenous leukemia during imatinib treatment. Acta Haematol 117:236, 2007. [PMID: 17308370]
960. Kantarjian HM, Cortes J, O’Brien S, et al: Imatinib mesylate (STI571) therapy for Philadelphia chromosome–positive chronic myelogenous leukemia in blast phase. Blood 99:3547, 2002. [PMID: 11986206]
961. Giles FJ, Larson RA, Kantarjian HM, et al: Nilotinib in patients with Philadelphia chromosome-positive chronic myelogenous leukemia in blast crisis (CML-BC) who are resistant or intolerant to imatinib. J Clin Oncol 26:376, 2008.
962. Cortes J, Rousselot P, Kin DW, et al: Dasatinib induces complete hematologic and cytogenetic responses in patients with imatinib-resistant or intolerant chronic myeloid leukemia in blast crisis. Blood 109:3207, 2007. [PMID: 17185463]
963. Barone S, Baer MR, Sait SNJ, et al: High-dose cytosine arabinoside and idarubicin treatment of chronic myeloid leukemia in myeloid blast crisis. Am J Hematol 67:119, 2001. [PMID: 11343384]
964. Hirase C, Maeda Y, Takai S, Kanamaru A: Hypersensitivity of Ph-positive lymphoid cell lines to rapamycin: Possible clinical application of mTOR inhibitor. Leuk Res 33:450, 2009. [PMID: 18783828]
965. Champlain R, Ho W, Arenson E, Gale RP: Allogeneic bone marrow transplantation for chronic myelogenous leukemia in chronic or accelerated phase. Blood 60:1038, 1982.
966. McGlave PB, Kim TH, Hard DD, et al: Successful allogeneic bone-marrow transplantation for patients in the accelerated phase of chronic granulocytic leukaemia. Lancet 2:625, 1982. [PMID: 6125774]
967. Martin PJ, Clift RA, Fisher LD, et al: HLA-identical marrow transplantation during accelerated-phase chronic myelogenous leukemia: Analysis of survival and remission duration. Blood 77:1978, 1988.
968. Falkenberg JHF, Wafelman AR, Joosten P, et al: Complete remission of accelerated phase chronic myeloid leukemia by treatment with leukemia-reactive cytotoxic T lymphocytes. Blood 94:1201, 1999.
969. Weisser M, Schleuning M, Haferlach C, et al: Allogeneic stem-cell transplantation provides excellent results in advanced stage chronic myeloid leukemia with major cytogenetic response to pre-transplant imatinib therapy. Leuk Lymphoma 48:295, 2007. [PMID: 17325889]
970. Carella AM, Gaozza E, Raffo MR, et al: Therapy of acute phase chronic myelogenous leukemia with intensive chemotherapy, blood cell autotransplant and cyclosporin A. Leukemia 5:517, 1991. [PMID: 1676080]
971. Bouvet M, Babiera GV, Termuhlen PM, et al: Splenectomy in the accelerated or blastic phase of chronic myelogenous leukemia: A single-institution 25-year experience. Surgery 122:20, 1997. [PMID: 9225910]
972. Wadhwa J, Szydio RM, Apperley J, et al: Factors affecting duration of survival after onset of blastic transformation of chronic myeloid leukemia. Blood 99:2304, 2002. [PMID: 11895760]
973. Majiis A, Smith TL, Talpaz M, et al: Signficance of cytogenetic clonal evolution in chronic myelogenous leukemia. J Clin Oncol 14:196, 1996.
974. Fialkow PJ, Jacobsen RJ, Singer JW, et al: Philadelphia chromosome (Ph1)-negative chronic myelogenous leukemia (CML): A clonal disease with origin in a multipotent stem cell. Blood 56:70, 1980. [PMID: 6930309]
975. Cortes J: CMML: A biologically distinct disease. Curr Hematol Rep 2:202, 2003. [PMID: 12901341]
976. Onida F, Beran M: Chronic myelomonocytic leukemia: Myeloproliferative variant. Curr Hematol Rep 3:218, 2004. [PMID: 15087071]
977. McCollum A, Bigelow CL, Elkins SL, et al: Unusual skin lesions in chronic myelomonocytic leukemia. South Med J 96:681, 2003. [PMID: 12940320]
978. Saif MW, Hopkins JL, Gore SD: Autoimmune phenomena in patients with myelodysplastic syndromes and chronic myelomonocytic leukemia. Leuk Lymphoma 43:2083, 2002. [PMID: 12533032]
979. Cambier N, Baruchel A, Schlageter MH, et al: Chronic myelomonocytic leukemia: From biology to therapy. Hematol Cell Ther 39:41, 1997. [PMID: 9168299]
980. Stemmler J, Wittman GW, Hacker U, Heinemann V: Leukapheresis in chronic myelomonocytic leukemia with leukostasis syndrome: Elevated serum lactate levels as an early sign of microcirculation failure. Leuk Lymphoma 43:1427, 2002. [PMID: 12389624]
981. Bain BJ: Hypereosinophilia. Curr Opin Hematol 7:21, 2000. [PMID: 10608500]
982. Aguayo A, Kantarjian H, Manshouri T, et al: Angiogenesis in acute and chronic leukemias and myelodysplastic syndromes. Blood 96:2240, 2000. [PMID: 10979972]
983. Bellemy WI, Richter L, Sirjani D, et al: Vascular endothelial cell growth factor in autocrine promoter of abnormal localized precursors and leukemia progenitor formation in myelodysplastic syndromes. Blood 97:1427, 2001.
984. Ramshaw HS, Bardy PG, Lee MA, Lopez AQF: Chronic myelomonocytic leukemia requires granulocytic-macrophage colony-stimulating factor for growth in vitro and in vivo. Exp Hematol 30:1124, 2002. [PMID: 12384142]
985. Tessema M, Länger F, Dingemann J, et al: Aberrant methylation and impaired expression of the p14INK4B cell cycle regulatory gene in chronic myelomonocytic leukemia (CMML). Leukemia 17:910, 2003. [PMID: 12750705]
986. Magnusson MK, Meade KE, Nakamura R, et al: Activity of STI571 in chronic myelomonocytic leukemia with a platelet-derived growth factor B receptor fusion oncogene. Blood 100:1088, 2002. [PMID: 12130532]
987. Apperley JF, Gardembas M, Melo JV, et al: Response to imatinib mesylate in patients with chronic myeloproliferative diseases with rearrangements of the platelet-derived growth factor receptor beta. N Engl J Med 347:481, 2002. [PMID: 12181402]
988. Wessels JW, Fibbe WE, van der Keur D, et al: T(5;12)(q31;p12): A clinical entity with features of both myeloid leukemia and chronic myelomonocytic leukemia. Cancer Genet Cytogenet 65:7, 1993. [PMID: 8431918]
989. Golub TR, Barker GF, Love HM, Gilliland DG: Fusion of PDGF receptor to a novel ets-like gen, tel, in chronic myelomonocytic leukemia with t(5;12) chromosomal translocation. Cell 77:307, 1994. [PMID: 8168137]
990. Cross NCP, Reiter A: Tyrosine kinase genes in chronic myeloproliferative diseases. Leukemia 16:1207, 2002. [PMID: 12094244]
991. Gunby RH, Cazzaniga G, Tassi E, et al: Sensitivity to imatinib but low frequency of the TEL/PDGFR fusion protein in chronic myelomonocytic leukemia. Haematologica 88:408, 2003. [PMID: 12681968]
992. Pitini V, Arrigo C, Teti D, et al: Response to STI571 in chronic myelomonocytic leukemia with platelet derived growth factor beta receptor involvement: A new case report. Haematologica 88:ECR18, 2003.
993. Kröger N, Zabelina T, Guardiola P, et al: Allogeneic stem cell transplantation of adult chronic myelomonocytic leukemia. Br J Haematol 118:67, 2002. [PMID: 20120461]
994. Onida F, Kantarjian HM, Smith TL, et al: Prognostic factors and scoring systems in chronic myelomonocytic leukemia: A prospective analysis of 213 patients. Blood 99:840, 2002. [PMID: 11806985]
995. Germing U, Strupp C, Alvado M, Gattermann N: New prognostic parameters for chronic myelomonocytic leukemia? Blood 100:731, 2002. [PMID: 12123232]
996. Sagaster V, Ohler L, Berer A, et al: High spontaneous colony growth in chronic myelomonocytic leukemia correlates with increased disease activity and is a novel prognostic factor for predicting short survival. Ann Hematol 83:9, 2004. [PMID: 13680175]
997. Stillman RG: A case of myeloid leukemia with predominance of eosinophil cells. Med Rec 81:594, 1912.
998. Benvenisti DS, Ultmann JE: Eosinophilic leukemia. Ann Intern Med 71:731, 1969. [PMID: 4391225]
999. Chusid MJ, Dale D, West BG, Wolff SM: The hypereosinophilic syndrome: Analysis of fourteen cases with a review of the literature. Medicine (Baltimore) 54:1, 1975. [PMID: 1090795]
1000. Brito-Babapulle F: The eosinophilias: Including the idiopathic hypereosinophilic syndrome. Br J Haematol 121:203, 2003. [PMID: 12694242]
1001. Robyn J, Lemery S, McCoy JP, et al: Multilineage involvement of the fusion gene in patients with FIP1L1/PDGFRA-positive hypereosinophilic syndrome. Br J Haematol 132:286, 2006. [PMID: 16409293]
1002. Crescenzi B, Chase A, Starza RL, et al: FIP1L1-PDGFRA in chronic eosinophilic leukemia and BCR-ABL1 in chronic myeloid leukemia affect different leukemic cells. Leukemia 21:397, 2007. [PMID: 17215855]
1003. Bain BJ: Cytogenetic and molecular genetic aspects of eosinophilic leukemia. Br J Haematol 122:173, 2003. [PMID: 12846884]
1004. Gotlib J, Cools J, Malone JM III, et al: The FIP1L1-PDGFRA fusion tyrosine kinase in hypereosinophilic syndrome and chronic eosinophilic leukemia: Implications for diagnosis, classification, and management. Blood 103:2879, 2004. [PMID: 15070659]
1005. Vandenberghe P, Wlodarska I, Michaux L, et al: Clinical and molecular features of FIP1L1-PDFGRA (+) chronic eosinophilic leukemia. Leukemia 18:734, 2004. [PMID: 14973504]
1006. Klion AD, Noel P, Akin C, et al: Elevated serum tryptase levels identify a subset of patients with a myeloproliferative variant of idiopathic hypereosinophilic syndrome associated with tissue fibrosis, poor prognosis, and imatinib responsiveness. Blood 101:4660, 2003. [PMID: 12676775]
1007. Florian S, Esterbauer H, Binder T, et al: Systemic mastocytosis (SM) associated with chronic eosinophilic leukemia (SM-CEL): Detection of FIP1L1/PDGFRalpha, classification by WHO criteria, and response to therapy with imatinib. Leuk Res 30:1201, 2006. [PMID: 16406018]
1008. Klion AD, Robyn J, Maric I, et al: Relapse following discontinuation of imatinib mesylate therapy for FIP1L1/PDGFRA-positive chronic eosinophilic leukemia: Implications for optimal dosing. Blood 110:3552, 2007. [PMID: 17709602]
1009. Verstovsek S, Tefferi A, Cortes J, et al: Phase II study of dasatinib in Philadelphia chromosome-negative acute and chronic myeloid disease, including systemic mastocytosis. Clin Cancer Res 14:3906, 2008. [PMID: 18559612]
1010. Esteva-Lorenzo FJ, Meehan KR, Spitzer TR, Mazumder A: Allogeneic bone marrow transplantation in a patient with hypereosinophilic syndrome. Am J Hematol 51:164, 1996. [PMID: 8579059]
1011. Juvonen E, Volin L, Koponen A, Ruutu T: Allogeneic blood stem cell transplantation following non-myeloablative conditioning for hypereosinophilic syndrome. Bone Marrow Transplant 29:457, 2002. [PMID: 11919738]
1012. Plotz S-G, Simon H-U, Darsow U, et al: Use of anti-interleukin-5 antibody in the hypereosinophilic syndrome with eosinophilic dermatitis. N Engl J Med 349:2334, 2003. [PMID: 14668459]
1013. Klion AD, Law MA, Noel P, et al: Safety and efficacy of the monoclonal anti-interleukin-5 antibody SCHJ55700 in the treatment of patients with hypereosinophilic syndrome. Blood 103:2939, 2004. [PMID: 15070668]
1014. Ardanani AD, Morice WG, Hoyer JD, Tefferi A: Chronic basophilic leukemia: A distinct clinical entity. Eur J Haematol 71:18, 2003.
1015. Lahortiga I, Akin C, Cools J, et al: Activity of imatinib in systemic mastocytosis with chronic basophilic leukemia and a PRKG2-PDGFRB fusion. Haematologica 93:49, 2008. [PMID: 18166785]
1016. Osgood EE: Monocytic leukemia. Report of six cases and review of one hundred and twenty-seven cases. Arch Intern Med 59:931, 1937.
1017. Bearman RM, Kjeldsberg CR, Pangalis GA, et al: Chronic monocytic leukemia in adults. Cancer 48:2239, 1981. [PMID: 6945897]
1018. Beattie JW, Seal RME, Crowther KV: Chronic monocytic leukemia. Q J Med 20:131, 1951.
1019. Sinn CW, Dick FW: Monocytic leukemia. Am J Med 20:588, 1956. [PMID: 13302235]
1020. Rodgers GM, Carrera CJ, Ries CA, Bainton DF: Blastic transformation of a well differentiated monocytic leukemia. Changes in cytochemical and cell surface markers. Leuk Res 6:613, 1982. [PMID: 6958932]
1021. Wahlin A, Nordenson I, Roos G: Chronic monocytic leukemia terminating in blastic transformation. Blut 53:405, 1986. [PMID: 3465380]
1022. Castro-Malaspina H, Schaison G, Brier J, et al: Philadelphia chromosome positive chronic myelocytic leukemia in children: Survival and prognostic factors. Cancer 51:721, 1983.
1023. Arico M, Biondi A, Pui C-H: Juvenile myelomonocytic leukemia. Blood 90:479, 1997. [PMID: 9226148]
1024. Neimeyer CM, Arico M, Basso A, et al: Chronic myelomonocytic leukemia in childhood. Blood 89:3535, 1997.
1025. Emanuel PD: Juvenile myelomonocytic leukemia and chronic myelomonocytic leukemia. Leukemia 22:1335, 2008. [PMID: 18548091]
1026. Niemeyer CM, Kratz C: Juvenile myelomonocytic leukemia. Curr Oncol Rep 5:510, 2003. [PMID: 14521811]
1027. Busque L, Gilliland DG, Prchal JT, et al: Clonality in juvenile chronic myelogenous leukemia. Blood 85:21, 1995. [PMID: 7803795]
1028. Cooper LJN, Shannon KM, Loken MR, et al: Evidence that juvenile chronic myelomonocytic leukemia can arise from a pluripotential stem cell. Blood 96:2310, 2000. [PMID: 10979983]
1029. Emanuel PD: RAS pathway mutations in juvenile myelomonocytic leukemia. Acta Haematol 119:207, 2008. [PMID: 18566538]
1030. Guilbert-Douet N, Morel F, Le Bris M-J, et al: Somatic PTPN11 mutation with a heterogeneous clonal origin in children with juvenile myelomonocytic leukemia. Leukemia 18:1142, 2004.
1031. Miyauchi J, Asada M, Sasaki M, et al: Mutations of the N-ras gene in juvenile chronic myelogenous leukemia. Blood 83:2248, 1994. [PMID: 8161790]
1032. Bader JL, Miller RW: Neurofibromatosis and childhood leukemia. J Pediatr 92:925, 1978. [PMID: 96239]
1033. Brodeur GM: The NF1 gene in myelopoiesis and childhood myelodysplastic syndrome. N Engl J Med 330:637, 1994. [PMID: 8302348]
1034. Shannon KM: Loss of normal NF1 allele from the bone marrow of children with type 1 neurofibromatosis and malignant myeloid disorders. N Engl J Med 330:597, 1994. [PMID: 8302341]
1035. Bollag G: Loss of NF1 results in activation of RAS signaling pathway and leads to aberrant growth in haematopoietic cells. Nat Genet 12:137, 1996.
1036. Tartaglia M, Niemeyer CM, Fragale A, et al: Somatic mutations in PTPN11 in juvenile myelomonocytic leukemia, myelodysplastic syndrome and acute myeloid leukemia. Nat Genet 34:148, 2003. [PMID: 12717436]
1037. Owen G, Lewis IJ, Morgan M, et al: Prognostic factors in juvenile chronic granulocytic leukaemia. Br J CancerSuppl 18:S68, 1992.
1038. Estrov Z, Grunberger T, Chan HSL, Freedman MH: Juvenile chronic myelogenous leukemia. Characterization of the disease using cell cultures. Blood 67:1382, 1986. [PMID: 3486010]
1039. Estrov Z, Dube ID, Chan HSL, Freedman MH: Residual juvenile chronic myelogenous leukemia cells detected in peripheral blood during clinical remission. Blood 70:1466, 1987. [PMID: 3478101]
1040. Emanuel PD, Bates LJ, Zhu S-W, et al: The role of monocyte-derived hemopoietic growth factors in the regulation of myeloproliferation in juvenile chronic myelogenous leukemia. Exp Hematol 19:1017, 1991. [PMID: 1915702]
1041. Morerio C, Acquila M, Rosanda C, et al: HCMOGT-1 is a novel fusion partner to PDGFRB in juvenile myelomonocytic leukemia with t(5;17)(q33;p11.2). Cancer Res 64:2649, 2004. [PMID: 15087372]
1042. Inoue S, Ravindranath Y, Thompson RI, et al: Cytogenetics of juvenile type chronic granulocytic leukemia. Cancer 39:2017, 1977. [PMID: 265749]
1043. Brodeur GM, Dow LW, Williams DL: Cytogenetic features of juvenile chronic myelogenous leukemia. Blood 53:812, 1979. [PMID: 285733]
1044. Chan HSL, Estrov Z, Weitzman SS, Freedman MH: The value of intensive combination chemotherapy for juvenile chronic myelogenous leukemia. J Clin Oncol 5:1960, 1987. [PMID: 2445935]
1045. Kang HJ, Shin HY, Choi HS, Ahn HS: Novel regimen for the treatment of juvenile myelomonocytic leukemia (JMML). Leuk Res 28:167, 2004. [PMID: 14654081]
1046. Pui CH, Arico M: Isotretinoin for juvenile chronic myelogenous leukemia. N Engl J Med 332:1520, 1995. [PMID: 7739703]
1047. Bernard F, Thomas C, Emile JF, et al: Transient hematologic and clinical effects of E21R in a child with end-stage juvenile myelomonocytic leukemia. Blood 99:2615, 2002. [PMID: 11895804]
1048. Locatelli F, Niemeyer C, Angelucci E, et al: Allogeneic bone marrow transplantation for chronic myelomonocytic leukemia in childhood. J Clin Oncol 15:556, 1997.
1049. Manabe A, Okamura J, Yumura-Yagi K, et al: Allogeneic hematopoietic stem cell transplantation for 27 children with juvenile myelomonocytic leukemia diagnosed based on the criteria of the International JMML Working Group. Leukemia 16:645, 2002. [PMID: 11960345]
1050. Worth A, Rao K, Webb D, et al: Successful treatment of juvenile myelomonocytic leukemia relapsing after stem cell transplantation using donor lymphocyte infusion. Blood 101:1713, 2003. [PMID: 12393482]
1051. Scrideli CA, Baruffi MR, Rogatto SR, et al: B lineage acute lymphoblastic leukemia transformation in a child with juvenile myelomonocytic leukemia, type 1 neurofibromatosis and monosomy of chromosome 7. Possible implications in the leukemogenesis. Leuk Res 27:371, 2003. [PMID: 12531231]
1052. Tuohey EL: A case of splenomegaly with polymorphonuclear neutrophil hyperleukocytosis. Am J Med Sci 160:18, 1920.
1053. Froberg MK, Brunning RD, Dorion P, et al: Demonstration of clonality in neutrophils using FISH in a case of chronic neutrophilic leukemia. Leukemia 12:623, 1998. [PMID: 9557623]
1054. Böhm J, Schaefer HE: Chronic neutrophilic leukemia:14 new cases of an uncommon myeloproliferative disorder. J Clin Pathol 55:862, 2002. [PMID: 20120533]
1055. Standen GR, Steers FJ, Jones L: Clonality in chronic neutrophilic leukemia associated with myeloma: Analysis using the X-linked probe M27. J Clin Pathol 46:297, 1993. [PMID: 8098719]
1056. Bohm J, Kock S, Schaefer HE, Fisch P: Evidence of clonality in chronic neutrophilic leukaemia. J Clin Pathol 56:292, 2003. [PMID: 12663642]
1057. Yanagisawa K, Ohminami H, Sato M, et al: Neoplastic involvement of granulocytic lineage, not granulocytic-monocytic, monocytic, or erythrocytic lineage, in a patient with chronic neutrophilic leukemia. Am J Hematol 57:221, 1998. [PMID: 9495373]
1058. Hasegawa T, Suzuki K, Sakamoto C, et al: Expression of the inhibitor of apoptosis (IAP) family members in human neutrophils: Up-regulation of cIAP2 in chronic neutrophilic leukemia. Blood 101:1164, 2003. [PMID: 12393423]
1059. Hasle H, Olesen G, Kerndrup G, et al: Chronic neutrophilic leukaemia in adolescence and young adulthood. Br J Haematol 94:628, 1996. [PMID: 8826884]
1060. Elliott MA, Dewald GW, Tefferi A, Hanson CA: Chronic neutrophilic leukemia (CNL): A clinical and pathological entity. Leukemia 15:35, 2001. [PMID: 11243396]
1061. Reilly JT: Chronic neutrophilic leukaemia: A distinct clinical entity? Br J Haematol 116:10, 2002. [PMID: 11841395]
1062. Elliott MA: Chronic neutrophilic leukemia. Curr Hematol Rep 3:210, 2004. [PMID: 15087070]
1063. Piliotis E, Kutas G, Lipton JH: Allogeneic bone marrow transplantation in the management of chronic neutrophilic leukemia. Leuk Lymphoma 43:2051, 2002. [PMID: 12481908]
1064. Ito T, Kojima H, Otani K, et al: Chronic neutrophilic leukemia associated with monoclonal gammopathy of undetermined significance. Acta Haematol 95:140, 1996. [PMID: 8638444]
1065. Vorobiof DA, Benjamin A, Kaplan H, Dvilansky A: Chronic granulocytic leukemia, neutrophilic type with paraproteinemia (IgA type K). Acta Haematol 60:316, 1978. [PMID: 101017]
1066. Carcassonne Y, Gastaut JA, Sebahoun G, Gratecos N: Découverte simultanée chez un même malade d’un myélome, d’une leucémie granuleuse (à polynucléaires neutrophils) et d’une maladie de Paget. Nouv Rev Fr Hematol 18:240, 1977. [PMID: 268608]
1067. Franchi F, Seminara P, Gruinchi G: Chronic neutrophilic leukemia and myeloma. Report on long survival. Tumori 70:105, 1984. [PMID: 6585081]
1068. Lewis MJ, Oelbaum MH, Coleman M, Allen S: An association between chronic neutrophilic leukaemia and multiple myeloma with a study of cobalamin-binding proteins. Br J Haematol 63:173, 1986. [PMID: 3458500]
1069. Rovira M, Cervantes F, Namdedeu B, Rozman C: Chronic neutrophilic leukaemia preceding for seven years the development of multiple myeloma. Acta Haematol 3:94, 1990.
1070. Standen GR, Jasani B, Wagstaff M, Wardrop CAJ: Chronic neutrophilic leukemia and multiple myeloma. Cancer 66:162, 1990. [PMID: 2112978]
1071. Nagai M, Oda S, Iwamoto M, et al: Granulocyte-colony stimulating factor concentrates in a patient with plasma cell dyscrasia and clinical features of chronic neutrophilic leukaemia. J Clin Pathol 49:858, 1996. [PMID: 8943758]
1072. Dinçol G, Nalçaci M, Dogan O, et al: Coexistence of chronic neutrophilic leukemia with multiple myeloma. Leuk Lymphoma 43:649, 2002. [PMID: 20116832]
1073. Masini L, Salvarani C, Macchioni P, et al: Chronic neutrophilic leukemia (CNL) with karyotype abnormalities associated with plasma cell dyscrasia. Haematologica 77:277, 1992. [PMID: 1427435]
1074. Pascucci M, Dorion P, Makary A, Froberg MK: Chronic neutrophilic leukemia evolving from the myelodysplastic syndrome. Acta Haematol 98:163, 1997. [PMID: 9352749]
1075. Takamatsu Y, Kondo S, Inoue M, Tamura K: Chronic neutrophilic leukemia with dysplastic features mimicking myelodysplastic syndrome. Int J Hematol 63:65, 1996. [PMID: 8713578]
1076. Higuchi T, Oba R, Endo M, et al: Transition of polycythemia vera to chronic neutrophilic leukemia. Leuk Lymphoma 33:203, 1999. [PMID: 10194140]
1077. Billio A, Venturi R, Morello E, et al: Chronic neutrophilic leukemia evolving from polycythemia vera with multiple chromosome rearrangements: A case report. Haematologica 86:1225, 2001. [PMID: 11694412]
1078. Foa P, Iurlo A, Saglio G, et al: Chronic neutrophilic leukemia associated with polycythemia vera. Br J Haematol 78:286, 1991. [PMID: 2064971]
1079. Higuchi T, Oba R, Endo M, et al: Transition of polycythemia vera to chronic neutrophilic leukemia. Leuk Lymphoma 33:203, 1999. [PMID: 10194140]
1080. Iurlo A, Foa P, Mailo AT, et al: Polycythemia vera terminating in chronic neutrophilic leukemia. Am J Hematol 35:139, 1990. [PMID: 2399908]
1081. Soda H, Kuriyama K, Tomonaga M, et al: Lymphoid crisis with T-cell phenotypes in a patient with Philadelphia chromosome negative chronic myeloid leukemia. Br J Haematol 59:671, 1985. [PMID: 3872678]
1082. Kessler JF, Grogan TM, Greenberg BR: Philadelphia-chromosome-negative chronic myelogenous leukemia with lymphoid stem cell blastic transformation. Am J Hematol 18:201, 1985. [PMID: 3855600]
1083. Dobrovic A, Morley AA, Seshadri R, Januszewicz EH: Molecular diagnosis of Philadelphia negative CML using the polymerase chain reaction and DNA analysis: Clinical features and course of M-bcr negative and M-bcr positive CML. Leukemia 5:187, 1990.
1084. Martiat P, Michaux JL, Rodhain J, et al: Philadelphia-negative (Ph–) chronic myeloid leukemia (CML): Comparison with Ph+ CML and chronic myelomonocytic leukemia. Blood 78:205, 1991. [PMID: 2070054]
1085. VanderPlas DC, Grosveld G, Hagemeijer A: Review of clinical, cytogenetic, and molecular aspects of Ph-negative CML. Cancer Genet Cytogenet 52:143, 1991.
1086. Galton DA: Haematological differences between chronic granulocytic leukemia, atypical chronic myeloid leukaemia and chronic myelomonocytic leukaemia. Leuk Lymphoma 7:343, 1992. [PMID: 1493435]
1087. Kato Y, Sawada H, Tashima M et al: Heterogeneous features of Ph-negative CML—Possible existence of Ph-negative, bcr-rearrangement-negative CML. Acta Haematol 52:1004, 1989. [PMID: 2588950]
1088. Selleri L, Emilia G, Luppi M, et al: Chronic myelogenous leukemia with typical clinical and morphological features can be Philadelphia chromosome negative and “bcr negative”. Hematol Pathol 4:67, 1990. [PMID: 2197265]
1089. Costello R, Sainty D, LaFage-Pochitaloff M, Gabert J: Clinical and biological aspects of Philadelphia-negative/BCR-negative chronic myeloid leukemia. Leuk Lymphoma 25:225, 1997. [PMID: 9168433]
1090. Kurzrock R, Bueso-Ramos CE, Kantarjian H, et al: BCR rearrangement-negative chronic myelogenous leukemia revisited. J Clin Oncol 19:2915, 2001. [PMID: 11387365]
1091. Onida F, Ball G, Kantarjian HM, et al: Characteristics and outcome of patients with Philadelphia chromosome negative, bcr/abl negative chronic myelogenous leukemia. Cancer 95:1673, 2002. [PMID: 12365015]
1092. O’Brien SG, Viera SA, Connors S, et al: Transient response to imatinib mesylate (STI571) in a patient with ETV6-ABL t(9;12) translocation. Blood 99:3465, 2002. [PMID: 11964320]
1093. Mauro MJ, Loriaux M, Deininger MW: Ph-positive and -negative myeloproliferative syndromes may coexist. Leukemia 18:1305, 2004. [PMID: 15103389]
1094. Raskind WH, Ferraris AM, Najfeld V, et al: Further evidence for the existence of a clonal Ph-negative stage in some cases of Ph-positive chronic myelocytic leukemia. Leukemia 18:1305, 2004.
1095. Chee YL, Vickers MA, Stevenson D, et al: Fatal myelodysplastic syndrome developing during therapy with imatinib mesylate and characterized by the emergence of complex Philadelphia negative clones. Leukemia 17:634, 2003. [PMID: 12646955]
1096. Meeus P, Demuynck H, Martiat P, et al: Sustained clonal karyotype abnormalities in the Philadelphia chromosome negative cells of CML patients successfully treated with imatinib. Leukemia 17:465, 2003. [PMID: 12592350]
1097. Bumm T, Muller C, Al Ali HK, et al: Emergence of clonal cytogenetic abnormalities in Ph-cells in some CML patients in cytogenetic remission to imatinib but restoration of polyclonal hematopoiesis in the majority. Blood 101:1941, 2001.