ABOUT 40 YEARS ago two forms of chronic myelocytic leukemia (CML) were recognized in children. One had the typical features of CML of adulthood and usually appeared in children older than 4 years; the other affected younger children and presented as a myelomonocytic proliferation associated with hemorrhage, infection, lymphadenopathy, and skin rash. The prognosis of the latter group of patients was invariably poor.1-3 In subsequent attempts to identify clinico-biological features that would further discriminate between these leukemias, Reisman and Trujillo4 found the absence of the Philadelphia (Ph) chromosome to be a hallmark of the juvenile-type entity (JCML). Signs of disturbed erythropoiesis were also reported to be typical of JCML, including low levels of hemoglobin (Hb) A2 and erythrocyte carbonic anhydrase,5 together with a marked increase of HbF,6 and glucose-6-phosphate dehydrogenase activity.7 Maurer and others concluded that JCML is accompanied by a reversion to fetal-like erythropoiesis.8-10 The observation of chromosomal translocations in leukemic bone marrow cells of different lineages suggested that the pathologic process might involve a multipotent stem cell.7,11 We review here the salient clinical and biological features of this disease, emphasizing recent advances in its treatment and in understanding its pathological origin.

The classification of chronic childhood myeloproliferative disorders remains controversial. Although the proposed categories of the French-American-British (FAB) system do not always correspond to the clinical features of such cases,12 they still constitute the standard for comparative evaluation of different series of patients. Patients who present with peripheral blood monocytosis and bone marrow dysplasia (often in association with monosomy 7) with a morphological picture of chronic myelomonocytic leukemia (CMML) in the absence of the t(9; 22) translocation are generally considered to have the juvenile form of CML. However, in different reports, patients sharing most of the above features are variously described as having JCML,7,8,10,13-15 monosomy 7 syndrome,16,17 or CMML in accord with the FAB system.12,18,19 Using features such as cytogenetic abnormalities and pretreatment HbF level, Passmore et al20 recently reclassified their cases as (1) JCML if there was CMML cellular morphology and raised HbF (>10%), and no monosomy 7; or (2) infantile monosomy 7 syndrome if the presenting age was less than 4 years, with any type of myelodysplasia and monosomy 7. All other patients, including older children with monosomy 7, were classified according to FAB criteria. The revised classification led the authors to propose a new prognostic scoring system.20 Although the aim of providing an improved system of risk assessment is laudable, the utility of the scoring system proposed by Passmore et al20 may be compromised by the criteria they used to reclassify cases of childhood myelodysplastic syndrome (MDS). In particular, some categories, such as the monosomy 7 syndrome, are widely debated or not accepted by some investigators.18,19 Thus, the uncertainty of diagnostic criteria in this group of disorders can be expected to limit the utility of the proposed scoring system.

In an effort to resolve most of the above described discrepancies, the International Juvenile Myelomonocytic Leukemia Working Group has recently proposed the term juvenile myelomonocytic leukemia (JMML; R. Castleberry, personal communication, December 1996), which has been accepted and widely used in some recent publications.21-23 It will be used throughout the remainder of this review.

The exact incidence of JMML is not yet known. In the only population-based study reported to date,18,24 JMML accounted for 18% of all cases of myelodysplastic syndrome in children less than 15 years old and 1.62% of all hematologic malignancies (0.61 new cases per year per million children at risk). The disease predominates in children younger than 4 years with 40% of cases occurring before the age of 1 year and 60% before the age of 2; however, 26% of patients with JMML are 3 years of age or older. Boys are affected more often than girls (M/F ratio, 2.5). Associated conditions include neurofibromatosis type 1 (NF1)25-27 in 7%, and a variety of occasional clinical abnormalities in an additional 9% of cases. At least three pairs of affected twins have been described.15,20,28 In another study, an identical twin was healthy at the time his brother presented with JMML and remained free of leukemia during preparation of the written report.29 

The onset of JMML is often heralded by acute or subacute symptoms of a recent infection. Table 1 summarizes clinical and laboratory features of patients variably referred to as juvenile chronic granulocytic leukemia (JCGL),11,29 JCML,13,15,20,30-35,36,37-42 CMML20,28,43 or monosomy 7.20,40 As shown, more than half of the patients have fever and signs of respiratory involvement, such as pharyngo-tonsillitis or bronchitis; pulmonary infection is infrequent. Bleeding symptoms are observed in about 50% of cases. Prominent findings are those common to most proliferative diseases: enlarged spleen, hepatomegaly, and lymphadenopathy. Skin involvement is apparent in about one half of the patients, with maculo-papular rash (often starting from the face) and xanthomas, or cafe-au-lait spots, seen in patients with NF1.

Table 1.

Presenting Features of Children With JMML

CharacteristicCategoryNo.%
Clinical 
Sex Male 146 71 
 Female 60 29 
Age <1 yr 72 40 
 1 to <2 yr 36 20 
 2 to <3 yr 25 14 
 ≥3 yr 46 26 
Hepatomegaly  165 79 
Splenomegaly  206 97 
Lymphadenopathy  89 52 
Skin involvement  78 48 
Associated conditions NF1 14 
 Other conditions 17 
Laboratory 
Leukocyte count (×109/L) <50 144 70 
 50-99 39 22 
 ≥100 14 
Monocyte count (×109/L) <5 67 35 
 5-9.9 45 32 
 ≥10 46 33 
Percentage of blasts in peripheral blood 0-2 92 56 
 >2 73 44 
Hb level (g/dL) >8-12 100 66 
 <8 53 34 
HbF level (%) 0-9 71 38 
 ≥10 117 62 
Platelets count (×109/L) ≥100 40 23 
 99-50 54 30 
 <50,000 80 47 
Bone marrow blasts (%) 0-9 67 64 
 10-20 27 26 
 >20 12 10 
Alkaline phosphatase score Low 47 41 
Cytogenetic analysis Normal 132 68 
 Monosomy 7 31 16 
 Other abnormalities 32 16 
CharacteristicCategoryNo.%
Clinical 
Sex Male 146 71 
 Female 60 29 
Age <1 yr 72 40 
 1 to <2 yr 36 20 
 2 to <3 yr 25 14 
 ≥3 yr 46 26 
Hepatomegaly  165 79 
Splenomegaly  206 97 
Lymphadenopathy  89 52 
Skin involvement  78 48 
Associated conditions NF1 14 
 Other conditions 17 
Laboratory 
Leukocyte count (×109/L) <50 144 70 
 50-99 39 22 
 ≥100 14 
Monocyte count (×109/L) <5 67 35 
 5-9.9 45 32 
 ≥10 46 33 
Percentage of blasts in peripheral blood 0-2 92 56 
 >2 73 44 
Hb level (g/dL) >8-12 100 66 
 <8 53 34 
HbF level (%) 0-9 71 38 
 ≥10 117 62 
Platelets count (×109/L) ≥100 40 23 
 99-50 54 30 
 <50,000 80 47 
Bone marrow blasts (%) 0-9 67 64 
 10-20 27 26 
 >20 12 10 
Alkaline phosphatase score Low 47 41 
Cytogenetic analysis Normal 132 68 
 Monosomy 7 31 16 
 Other abnormalities 32 16 

Data from 11,13,15,20,29-43. Certain features are not reported for all patients.

Leukocyte counts tend to remain less than 50 × 109/L; in fact, fewer than 8% of cases have counts greater than 100 × 109/L. Monocytosis exceeding 5 × 109/L with circulating immature myeloid cells, erythroblasts, and a few blasts (<3% in 56% cases) are typical findings. A decreased leukocyte alkaline phosphatase score is not characteristic, occurring in less than 50% of the patients. The Hb level is usually low and is accompanied by erythrocyte abnormalities and low serum iron concentrations. An altered Hb pattern affords a readily accessible and reliable marker of JMML; at diagnosis the HbA2 concentration is reduced, whereas HbF level greater than 10% is found in two-thirds of patients. Thrombocytopenia is common and often severe. Elevated levels of serum vitamin B12 are frequently observed and appear related to active protein synthesis by granulocytes.44 Polyclonal hypergammaglobulinemia is also common, but its significance remains unclear.45 Increased serum levels of lysozyme are reported in two thirds of the patients in association with a higher number of immature circulating granulocytic precursors.19 Autoantibodies with different reactivities (antinuclear type or positive Coombs test) and no apparent clinical relevance have been observed in fewer than one fourth of the patients.19 

An analysis of peripheral blood usually provides more reliable diagnostic information than does an examination of the bone marrow smear. Typically, marrow cellularity is increased, with the vast majority of cells belonging to the myeloid series in all stages of maturation. Monocytes account for 5% to 10% of all myeloid cells, with the blast cell population far below the level seen in acute leukemia. Follow-up marrow evaluations rarely yield informative results. In contrast to other types of childhood leukemia, JMML has few discernible constitutional or clonal chromosome abnormalities, partly explaining the slow progress in understanding the pathobiology of this disease. In one study, 68% of the reported cases of JMML lacked cytogenetic abnormalities, the remainder having either dissimilar lesions (16%) or monosomy 7 (16%).46-48 

More recently in a retrospective analysis of 110 patients defined by entry criteria and treated in five European countries,19 the European Working Group on Myelodysplastic Syndromes in childhood (EWOG-MDS) found a constellation of features to have diagnostic relevance: hepatosplenomegaly in greater than 90%, lymphadenopathy in 76%, pallor in 64%, fever in 54%, skin rash in 36%, absence of the 9; 22 translocation, bone marrow blasts less than 20%, and peripheral blood monocytosis greater than 1 cell × 109/L. Additional criteria required for definite diagnosis included at least two of the following: spontaneous in vitro growth of granulocyte-macrophage progenitors (CFU-GM), HbF elevated for age, peripheral blood myeloid precursors, leukocyte count greater than 10 × 109/L, and chromosomal abnormalities.

The observation of patients who either were not treated or did not respond to treatment has provided useful information on the natural history of JMML. Approximately one third of the patients present with a rapidly progressive disease in which cachexia, organomegaly, and complications of marrow failure may lead to early death, whether or not treatment is instigated. About another third of the patients show a more indolent disease course characterized by clinical improvement with partial or even complete normalization of the blood count after minimal or no treatment. Such patients may enjoy a stable clinical remission, even when there is persistence of a variable pattern of disease markers, including splenomegaly, moderate leukocytosis, or monocytosis. During this true chronic phase of the disease, they may occasionally experience reactivation of their leukemia, sometimes in association with infectious episodes. Because these episodes are often self-limiting, introduction of chemotherapy at this time may produce what appears to be a clinical response. The ultimate outcome of leukemia is difficult to predict, but a significant proportion of patients eventually develop massive disease reactivation evolving into a rapid progression, often referred to as blastic crisis.15,29,49 The remaining patients have an intermediate prognosis.

Because the course of JMML is highly variable and the therapeutic choices wide, some investigators have attempted to identify factors that predict outcome or at least time to progression. The major role in defining prognosis seems to be played by age at the time of disease onset. Most published series support the generalization that children who develop the disease when younger than 1 or 2 years have a longer survival.15,20,30,50 In our review, infants younger than 1 year of age had a significantly higher probability of long-term survival than did older patients; there was no significant difference between infants presenting at the age of less than 6 months or 6 to 12 months. These relationships were valid whether patients treated with bone marrow transplantation were excluded (Fig 1) or included (not shown) in the analysis. This analysis should be regarded with caution both because we can only refer to published data and because data were taken from sources without uniform entry criteria and do not represent randomized clinical trials.

Fig. 1.

Kaplan-Meier survival estimates of 177 children with JMML according to age at the diagnosis. The curves were generated by pooling the data from the following references, by including only cases for which both informations (ie, age at diagnosis and duration of survival) were available.8,11,13,27,28,30-37,41,47,82,85,97-99,101,103 

Fig. 1.

Kaplan-Meier survival estimates of 177 children with JMML according to age at the diagnosis. The curves were generated by pooling the data from the following references, by including only cases for which both informations (ie, age at diagnosis and duration of survival) were available.8,11,13,27,28,30-37,41,47,82,85,97-99,101,103 

Close modal

Among the proposed unfavorable risk factors are a low platelet count and an increased HbF level15,20 (Table 2). In a recent review of their single-center series of childhood MDS patients, Passmore et al20 advocated a pediatric scoring system based on HbF level, platelet count, and cytogenetic results. The so-called FPC score was predictive of a good prognosis (60% survival at 5 years) when HbF was greater than 10%, platelets greater than 40 × 109/L, and clonal cytogenetic abnormalities were absent.

Table 2.

Unfavorable Prognostic Factors in JMML

FeatureStudy (reference)FeatureStudy (reference)
Age (mo)  HBF level 
>48 Niemeyer19  Increased Castro15  
>24 Castro,15 Passmore20   >15% Niemeyer19  
>12 Aricò,30 Pui50   >10% Owen,29 Passmore20  
>6 Owen29  Thrombocytopenia (×109/L) 
Male sex Owen29   <100 Castro15  
   <50 Aricò30  
Bleeding Castro,15 Owen29   <40 Passmore20  
 Niemeyer19   <33 Niemeyer19  
Hepatomegaly (>3 cm) Castro15  Blasts in peripheral blood 
   ≥2 × 109/L Castro15  
Hb level (<10 g/dL) Niemeyer19   >4% Niemeyer19  
Reticulocytes (<10%) Niemeyer19  Normoblasts in peripheral blood (>1 × 109/L) Castro15  
FPC score (≥2)* Passmore20  Blasts in the BM myeloid compartment (>5%) Niemeyer19  
FeatureStudy (reference)FeatureStudy (reference)
Age (mo)  HBF level 
>48 Niemeyer19  Increased Castro15  
>24 Castro,15 Passmore20   >15% Niemeyer19  
>12 Aricò,30 Pui50   >10% Owen,29 Passmore20  
>6 Owen29  Thrombocytopenia (×109/L) 
Male sex Owen29   <100 Castro15  
   <50 Aricò30  
Bleeding Castro,15 Owen29   <40 Passmore20  
 Niemeyer19   <33 Niemeyer19  
Hepatomegaly (>3 cm) Castro15  Blasts in peripheral blood 
   ≥2 × 109/L Castro15  
Hb level (<10 g/dL) Niemeyer19   >4% Niemeyer19  
Reticulocytes (<10%) Niemeyer19  Normoblasts in peripheral blood (>1 × 109/L) Castro15  
FPC score (≥2)* Passmore20  Blasts in the BM myeloid compartment (>5%) Niemeyer19  
*

Relevant in all cases of childhood MDS, including JMML.

Two consistent abnormalities have been shown by in vitro studies of JMML patients: the exuberant spontaneous growth of colony forming units-granulocyte/macrophage (CFU-GM) in the absence of exogenous growth factors,51-54 and the impaired growth of normal hemopoietic progenitors. Spontaneous cell growth has been ascribed to the peculiar hypersensitivity of the JMML bone marrow or peripheral blood progenitors to very low levels of cytokines and growth factors probably produced by adherent cells.55 Removal of monocytes by adherence to plastic before culture of bone marrow or peripheral blood progenitor cells abrogates spontaneous growth.55 The observed hypersensitivity response appears to be quite selective, as it occurs in tests with GM-CSF, but not other growth factors regulating early steps of myelopoiesis, such as inteleukin-3 (IL-3) or granulocyte colony-stimulating factor (G-CSF ).52 To date, binding studies and molecular analysis of the GM-CSF receptor have shown no abnormality,56 although several provocative clues have emerged (Fig 2). During neoplastic transformation of myeloid progenitors, RAS genes often acquire activating point mutations that lead to elevated levels of Ras-GTP, resulting in constitutive activation of this common signal transduction pathway.57 In this regard, mutated RAS genes have been found in 30% of cases of JMML.34,58-60 In one series most of the patients with mutated RAS were in the older age group with poor prognosis.34 Loss of the normal NF1 allele is a common finding in JMML cells from patients with NF1.61,62 As a tumor suppressor, the NF1 protein acts as GTPase, which in turn downregulates Ras-GTP. Primary leukemic cells from children with NF1 show a selective decrease in NF1-like GTPase activating protein (GAP) activity for RAS, but retain normal cellular GAP activity. Leukemic cells also show an elevated percentage of Ras in the GTP-bound conformation.63 These genetic and biochemical data from studies of primary JMML cells strongly support the hypothesis that inactivation of NF1 with consequent Ras deregulation plays an important role in leukemogenesis in children with NF1 who develop JMML. This idea is reinforced by a number of observations in mice with a targeted disruption of Nf1. First, heterozygous Nf1 mice are predisposed to myeloid leukemia and these leukemias delete the wild-type Nf1 allele.64 Second, fetal hematopoietic cells from embryos with homozygous inactivation of Nf1 show a pattern of selective in vitro GM-CSF hypersensitivity that is reminiscent of that seen in JMML.62,63 Finally, transplanting these Nf1 −/− fetal liver cells into irradiated recipients consistently induces a myeloproliferative disorder with clinical and pathologic features of JMML62 (K.M. Shannon, personal communication, December 1996). Taken together these human and murine data point to deregulated signaling through the RAS pathway as the central event in the abnormal growth of JMML progenitor cells. Activation of the JAK-2 tyrosine kinase65 or overexpression of the Shc proteins57 might represent an additional mechanism(s) to be involved in GM-CSF hypersensitivity.

Fig. 2.

Schematic diagram of GM-CSF signal transduction from cell surface to nucleus. GM-CSF binds to the α and β subunits of its cell surface receptor. Two distinct signaling pathways are triggered by GM-CSF: the Ras signaling pathway and the one that involves activation of the Jak2 tyrosine kinase. The first one includes Shc phosphorylation, increases in GTP-bound Ras and activation of mitogen activated (MAP) kinases. The presence of RAS points mutations or the inactivation of neurofibromin (the protein encoded by the neurofibromatosis type 1 gene, NF1 ), constitutively activate the Ras pathway by increasing intracellular levels of Ras-GTP. Modified from Emanuel et al.23

Fig. 2.

Schematic diagram of GM-CSF signal transduction from cell surface to nucleus. GM-CSF binds to the α and β subunits of its cell surface receptor. Two distinct signaling pathways are triggered by GM-CSF: the Ras signaling pathway and the one that involves activation of the Jak2 tyrosine kinase. The first one includes Shc phosphorylation, increases in GTP-bound Ras and activation of mitogen activated (MAP) kinases. The presence of RAS points mutations or the inactivation of neurofibromin (the protein encoded by the neurofibromatosis type 1 gene, NF1 ), constitutively activate the Ras pathway by increasing intracellular levels of Ras-GTP. Modified from Emanuel et al.23

Close modal

Several other cytokines secreted by malignant JMML cells have been implicated in autocrine or paracrine pathways of growth stimulation in JMML54 or in suppression of normal hematopoiesis.53 Bagby et al54 reported that monocytes from these patients secrete high levels of IL-1, which in turn stimulate the release of another growth factor(s) by normal accessory cells. In recent studies, an IL-1 receptor antagonist was reported to inhibit spontaneous JMML proliferation in vitro.66 Tumor necrosis factor (TNF ) α has been suggested to inhibit normal hematopoietic progenitors,53 resulting in anemia and thrombocytopenia, both clinical hallmarks of JMML.

The involvement of the erythroid lineage is a common feature of JMML, as indicated by the presence of mature but morphologically aberrant erythroid cells, the conversion to a fetal-type erythropoiesis, and the abnormal in vitro growth properties of peripheral erythroid progenitors.52,67 Cytogenetic analysis of burst-forming unit-erythroid (BFU-E) colonies from patients with a chromosome marker suggested that JMML may arise from early progenitors capable to differentiate into monocytic-macrophage–erythroid cells.38,39 The involvement of the erythroid lineage in this clonal disorder has been also confirmed by the observation of increased expression of GATA-1, erythropoietin receptor, and α and γ globin genes in JMML.68 More recently, loss of the normal NF1 allele in erythroblasts harvested from the BFU-E colonies of JMML patients has added support to this hypothesis.69 

Several lines of evidence indicate the clonal nature of JMML. First, clonality was shown by the presence of a cytogenetic marker in the bone marrow of rare patients with JMML,38 and more recently by a clonal pattern of X chromosome inactivation in girls with this disease.56 Loss of the normal NF1 allele in CD34+ cells in some JMML patients represents a third independent line of evidence.69 Cell separation studies have also traced the origin of JMML stem cells to the level of the most primitive myeloid progenitor.56,69 Highly purified CD34+ CD38 early progenitor cells (as well as the reticulocytes, platelets, monocytes, and granulocytes) from a JMML patient have been shown to be clonally derived when examined by HUMARA (human androgen receptor assay).56 Loss of the normal NF1 allele in unfractionated bone marrow samples and in CD34+ cells lends further support to this view. The chronic and rapidly progressive (blast crisis) phase of JMML15 is likely to have the same clonal origin. In one report, a child with JMML and cytogenetic evidence of monosomy 7 developed pre-B acute lymphoblastic leukemia49; the karyotypic analysis of the lymphoblasts in this case confirmed the presence of monosomy 7, as typically seen in pure stem cell disorders such as the Ph-chromosome–positive CML, in which involvement of both lymphocytes and myeloid cells is well recognized.70 Whether JMML represents a true stem cell disorder or the consequence of malignant transformation occurring in hematopoietic cells restricted to the myeloid differentiation remains controversial. Miles et al69 recently reported the retention of the normal NF1 allele in cell lines derived from pre-B lymphocytes of a JMML patient carrying an NF1 gene mutation. However, Nf1 −/− cells can reconstitute both myeloid and lymphoid cells in mice, but abnormal proliferation is observed only in the myeloid compartment63 (K.M. Shannon, personal communication, December 1996). The recent successful engraftment of the human JMML stem cells in primary and secondary SCID mice should provide a model with which to further delineate the stem-cell properties of JMML.71 

The relationship between JMML and childhood monosomy 7 syndrome is uncertain. Both disorders share several clinical features including a young age at diagnosis, a preponderance of male patients, prominent hepatosplenomegaly, myelomonocytic proliferation, increased frequency of NF1, and a poor prognosis when treatment consists of chemotherapy without marrow rescue.16,72-74 However, JMML patients without demonstrable monosomy 7 tend to have higher HbF levels,19 more pronounced lymphadenopathy, and more severe skin rashes than do patients with monosomy 7, who show a tendency to develop leukopenia and bacterial infections.16,72-75 The majority of patients with JMML do not have monosomy 7 at diagnosis, whereas others acquire the cytogenetic abnormality only at the time of the disease progression.28,41 In a study that failed to detect the loss of heterozygosity (LOH) of chromosome 7 in patients with JMML, Butcher et al76 speculated that JMML and monosomy 7 are distinct disorders that share some, but not all, of the multiple steps commonly seen in malignant transformation. This idea has gained further support from a recent study in which molecular analysis failed to show microscopic deletions of 7q in 22 cases of JMML.77 

The treatment of JMML continues to generate controversy. Early therapeutic attempts by Lilleymann, using sequential subcutaneous cytarabine and oral mercaptopurine, provided some improvement in the condition of patients, but not in survival time.13 Castro-Malaspina et al15 reported the failure of various treatment modalities, including chemotherapy with 6-mercaptopurine (with or without prednisone) in 27 patients and more intensive chemotherapy in four patients. Regardless of the type of regimen, chemotherapy never resulted in a complete remission (CR) in this large series. These investigators concluded that neither splenectomy nor radiotherapy nor moderate or intensive chemotherapy was of value in the treatment of JMML. Thus, during a period when vastly improved results were being reported for most childhood leukemias, the prognosis for JMML remained poor.

Experience over the past decade suggests that a subset of patients with JMML will become long-term survivors when treated exclusively with chemotherapy. Chan et al32 treated four children with an intensive AML-type regimen, three of whom relapsed after 11 to 21 months with one remaining in CR at 27 months follow-up. The outcome was superior to that in five children, who received less intensive therapy, all of whom died within 1 to 29 months. Despite obtaining hematological remission, ie, reversal of the blood counts to normal within 4 to 8 weeks of initiating therapy, with intensive chemotherapy, usually directed to AML, Festa et al33 were unable to eradicate extramedullary disease in their six patients, who all showed persistent hepatosplenomegaly and thus failure to achieve complete remission; all of the patients experienced hematologic relapse and died of disease at a median of 8 months. In his recent report on the use of intensive chemotherapy for childhood MDS, Hasle et al78 described 20 patients with this syndrome, including 8 with CMML who were treated with AML-type therapy; only 2 entered remission.

In our literature review we identified 171 children treated for JMML, with or without bone marrow rescue, for whom there was adequate information to correlate the duration of survival with the type of therapy. Fourteen of the 50 nonintensively treated patients (28%) were still alive after 1 to 307 months of follow-up (median 9 months), compared with only 3 of 30 (10%) who received intensive chemotherapy (surviving at 32, 36, and 120 months). Comparison of estimated survival probabilities suggests that intensive chemotherapy is not superior to nonintensive chemotherapy or even to no chemotherapy (Fig 3). These findings should be interpreted with caution because again we can only refer to published data, which were taken from sources without uniform entry criteria, did not consider proposed risk factors (Table 2) and do not represent randomized clinical trials.

Fig. 3.

Kaplan-Meier survival estimates of 171 children with JMML according to treatment applied as reviewed from the literature. The curves were generated by pooling the data from the following references, by including only cases for which both informations (ie, treatment applied and duration of survival) were available.13,15,19,20,27-29,31-35,37,40,43,47,79,82,83,85,97-103 

Fig. 3.

Kaplan-Meier survival estimates of 171 children with JMML according to treatment applied as reviewed from the literature. The curves were generated by pooling the data from the following references, by including only cases for which both informations (ie, treatment applied and duration of survival) were available.13,15,19,20,27-29,31-35,37,40,43,47,79,82,83,85,97-103 

Close modal

The poor results of chemotherapy for JMML, with or without other modalities, have led to the evaluation of alternative agents, including cytokines and biological response modifiers. Castleberry et al79 recently described the efficacy of isotretinoin in producing durable clinical and laboratory responses in 10 children with this disease, two of whom had complete remissions, three partial remissions, and one a minimal response; the remaining four patients had progressive disease. Of the seven patients who did not undergo BMT, one was in complete remission 83 months after diagnosis, and two others had stable disease at 36 and 37 months while still receiving isotretinoin. Although clearly warranting further study, these data should be evaluated cautiously. First, a clinical response, defined by the authors as a reduction of leukocytosis, does not include the correction of cytopenia. Moreover, it is well known that some patients with JMML can have long periods of stable disease (up to 10 years), even when untreated. Additionally, the very young age of 10 patients who received isotretinoin (median, 10 months), probably had a favorable influence on outcome (Table 2).

The apparently increased sensitivity of the JMML cells to interferon-α,14 together with successful use of interferon for CML,80 prompted some investigators to administer this cytokine to patients with the juvenile form of the disease.15,29 The results are difficult to evaluate because of the limited sample size, the heterogeneity of the presenting clinical features and the complex history of previous treatment. Nonetheless, in one series of 10 previously untreated patients, high dose interferon therapy (30 millions units/m2) appeared excessively toxic with no evidence of improved disease control.81 

Currently, bone marrow transplantation (BMT) is the only therapy that clearly improves outcome in the clinical management of JMML. Of 91 patients treated with this modality in 16 different reports, 38 (41%) were still alive, including 30 of the 60 patients who received grafts from HLA-matched or one antigen mismatched familial donor, 2 of 12 with mismatched donors, and 6 of 19 with matched unrelated donors. The estimated survival for patients treated with grafts from HLA-matched familial donors appears significantly better than results achieved with any other treatment modalities (Fig 3). Attempts to decrease the leukemic blast cell infiltrate or to ameliorate the degree of cytopenia, such as aggressive cytoreduction or splenectomy, may be helpful in some cases as a means to reduce the risk of peritransplant complications, but there is no evidence that such procedures may actually improve the survival rate or even reduce the risk of post-transplant leukemic relapse.83 The largest study of allogeneic BMT in JMML reported to date83,84 indicates that a preparative regimen consisting of busulfan and other cytotoxic drugs offers a higher likelihood of event-free-survival (due to lower rate of leukemic relapse) in children with an HLA-compatible relative donor than does any other modality including total-body-irradiation.85 As a result, this regimen is currently being used in the EWOG-BMT cooperative study.83 

For patients lacking access to BMT, novel therapeutic approaches should be considered. For instance, the significant pathogenic role of IL-1 in JMML and the inhibitory effect of its neutralization on spontaneous cell proliferation in vitro66 provide a compelling rationale for experimental treatment with an IL-1 receptor antagonist. Additionally, evidence of a significant in vitro inhibitory effect of IL-4 on the JMML growth pattern and GM-CSF, TNF, and IL-1 secretion86 would appear to support therapeutic strategies based on this cytokine as well. Recently, IL-10 was demonstrated to inhibit the autonomous in vitro growth of marrow and peripheral blood mononuclear cells from JMML patients, most likely through suppression of endogenous GM-CSF release.87 This observation suggests therapeutic evaluation of IL-10 in JMML patients.

The potent effects of the human GM-CSF analogue E21R, which completely blocks effects of the growth factor on cell proliferation and induces apoptosis, might also offer a novel therapeutic approach.22 It may also be possible to use mutant Ras peptides as targets for specific immunotherapy.88 Finally, the observation that oncogenic Ras proteins are unable to transform tissue culture cells unless they are farnesylated suggests that FPTase inhibitors may be useful in the treatment of JMML or other neoplastic disorders in which RAS contributes to dysregulated cell growth.89,90 This concept is now under investigation in model systems.91-94 

The diagnosis of JMML is based on evidence of splenomegaly, leukocytosis in the range of 20 to 30 cells × 109/L, monocytosis greater than 1.0 × 109/L, circulating myeloid precursors, and bone marrow hypercellularity with less than 20% myeloid blasts. Evidence of spontaneous growth of myelomonocytic cells in vitro, together with the verification of the absence of the t(9; 22) or the BCR/ABL fusion protein, should be obtained in all cases. High levels of HbF (usually >10%) are readily documented and strongly support a suspected diagnosis of JMML. Although JMML and monosomy 7 are probably two distinct entities, the presence of this cytogenetic abnormality should not be considered a diagnostic contraindication of JMML; patients with well-documented evidence of JMML and age greater than 1 year at disease onset should be considered at higher risk for rapidly progressive disease and thus possible candidates for early BMT from an HLA-identical sibling or an unrelated donor. Younger patients may benefit from nonintensive treatment, such as 6-mercaptopurine, 6-thioguanine, or isotretinoin. In general, patients should not be subjected to intensive chemotherapy or BMT with poorly matched donors unless their clinical course during the first few weeks or months is unusually aggressive or strongly suggestive of a blastic crisis. Experimental agents are justifiable in children with unfavorable risk features who lack an acceptable marrow donor. Meanwhile, it will be important to clarify the origin of fetal erythropoiesis in patients with JMML, the relationship between monosomy 796 and presenting clinical features, and the mechanisms that allow the disease to progress to blastic crisis.

The authors are indebted to Catherine Klersy, MD (Biometric Unit, Scientific Direction, IRCCS Policlinico S. Matteo, Pavia, Italy) for statistical analysis; Franco Locatelli, MD, Kevin Shannon, MD, Robert Castleberry, MD, and Charlotte Niemeyer, MD, for their critical comments and for sharing unpublished data; and John Gilbert for editorial review.

Supported in part by Grants No. PO1-CA-20180 and P30 CA-21765 from the National Cancer Institute (Bethesda, MD) (C.H.P.); by the American Lebanese Syrian Associated Charities (Memphis, TN) (C.H.P.); by Grant No. 390RCR91/01 from the IRCCS Policlinico S. Matteo, Pavia, Italy (M.A.); by Associazione Italiana per la ricerca sul Cancro (AIRC) (A.B.); by Consiglio Nazionale delle Ricerche (PF ACRO) No. 92.02140.PF.39 (A.B.); and by Fondazione Tettamanti (A.B.).

Address reprint requests to Maurizio Aricò, MD, Department of Pediatrics, University of Pavia, IRCCS Policlinico S. Matteo, 27100 Pavia, Italy.

1
Eisenberg
AA
Wallerstein
H
Chronic myelosis in children.
J Lab Clin Med
19
1934
713
2
Cooke
JV
Chronic myelogenous leukemia in children.
J Pediatr
42
1953
537
3
Bernard
J
Mathe
G
Delorme
JC
Barnoud
O
La leucemie myeloide chronique de l'enfant.
Arch Fr Pediatr
19
1962
881
4
Reisman
LE
Trujillo
JM
Chronic granulocytic leukemia of childhood. Clinical and cytogenetic studies.
J Pediatr
62
1963
710
5
Weatherall
DJ
Edwards
JA
Donohoe
WTA
Hemoglobin and red cell enzymes changes in juvenile myeloid leukemia.
BMJ
1
1968
676
6
Hardisty
RM
Speed
DE
Till
M
Granulocytic leukemia in childhood.
Br J Haematol
10
1964
551
7
Cao
A
Juvenile chronic myeloid leukemia.
Lancet
1
1970
1002
8
Maurer
HS
Vida
LN
Honig
GR
Similarities of the erythrocytes in Juvenile Chronic Myelogenous Leukemia to fetal erythropoiesis.
Blood
39
1972
778
9
Wheatherall
DJ
Clegg
JB
Wood
WG
Foetal erythropoiesis in human leukemia.
Nature
257
1975
710
10
Weinberg
RS
Leibowitz
D
Weinblatt
ME
Kochen
J
Alter
BP
Juvenile Chronic Myelogenous Leukemia: The only example of truly fetal (not fetal-like) erythropoiesis.
Br J Haematol
76
1990
307
11
Altman
AJ
Palmer
CG
Baehner
RL
Juvenile “Chronic Granulocytic” Leukemia: A panmielopathy with prominent monocytic involvement and circulating monocyte colony-forming cells.
Blood
43
1974
341
12
Bennett
M
Catovski
D
Daniel
MT
Flandrin
G
Galton
BAG
Gralnick
H
Sultan
C
Cox
C
The chronic myeloid leukemias: Guidelines for distinguishing chronic granulocytic, atypical chronic myeloid, and chronic myelomonocytic leukemia. Proposals by the French-American-British Cooperative leukemia group.
Br J Haematol
87
1994
746
13
Lilleymann
JS
Harrison
JF
Black
JA
Treatment of juvenile chronic myeloid leukemia with sequential subcutaneous cytarabine and oral mercaptopurine.
Blood
49
1977
559
14
Estrov
Z
Grunberger
T
Chan
H
Freedman
M
Juvenile chronic myelogenous leukemia: Characterization of the disease using cell cultures.
Blood
67
1986
1382
15
Castro-Malaspina
H
Schaison
G
Passe
S
Pasquier
A
Berger
R
Bayle-Weisgerberg
C
Miller
D
Seligmann
M
Bernard
J
Subacute and chronic myelomonocytic leukemia in children (Juvenile CML). Clinical and hematologic observations, and identification of prognostic factors.
Cancer
54
1984
675
16
Sieff
CA
Chessels
JM
Harvey
BAM
Pickthall
VJ
Lawler
SD
Monosomy 7 in childhood: A myeloproliferative disorder.
Br J Haematol
49
1981
235
17
Gyger
M
Bonny
Y
Forest
L
Childhood monosomy 7 syndrome.
Am J Hematol
13
1982
329
18
Hasle
H
Myelodysplastic syndromes in childhood-Classification, epidemiology, and treatment.
Leuk Lymphoma
13
1994
11
19
Niemeyer C, Aricò M, Basso G, Biondi A, Cantù Rajnoldi A, Creutzig U, Haas O, Harbott J, Hasle H, Kerndrup G, Locatelli F, Mann G, Stollmann-Gibbels B, Th Van't Veer-Korthof E, van Weering E, Zimmermann M and Members of the European Working Group on Myelodysplastic Syndromes in Childhood (EWOG-MDS): Chronic Myelomonocytic Leukemia in childhood: A retrospective analysis of 110 cases. Blood 89:3534, 1997
20
Passmore
SJ
Hann
IM
Stiller
CA
Ramani
P
Swansbury
GJ
Gibbons
B
Reeves
BR
Chessels
JM
Pediatric myelodysplasia: A study of 68 children and a new prognostic scoring system.
Blood
85
1995
1742
21
Haas
OA
Gadner
H
Pathogenesis, biology, and management of myelodysplastic syndromes in children.
Semin Hematol
33
1996
225
22
Iversen
P
Rodwell
RL
Pitcher
L
Taylor
KM
Lopez
AF
Inhibition of proliferation and induction of apoptosis in juvenile myelomonocytic leukemic cells by the granulocyte-macrophage colony-stimulating factor analogue E21R.
Blood
86
1996
2634
23
Emanuel
PD
Shannon
KM
Castleberry
RP
Juvenile myelomonocytic leukemia: Molecular understanding and prospects for therapy.
Molec Med Today
00
1996
468
24
Hasle
H
Jacobsen
BB
Pedersen
NT
Myelodysplastic syndromes in childhood: A population based study of nine cases.
Br J Haematol
81
1992
495
25
Royer
B
Blondet
C
Guilhard
J
Xantholeucemie du nourisson et neurofibromatose de Recklinghausen.
Ann Pediatr
5
1958
260
26
Bader
JL
Miller
RW
Neurofibromatosis and childhood leukemia.
J Pediatr
92
1978
925
27
Stiller
CA
Chessels
JM
Fitchett
M
Neurofibromatosis and childhood leukemia/lymphoma: A population-based UKCCSG study.
Br J Cancer
70
1994
969
28
Brandwein
JM
Horsman
DE
Eaves
AC
Eaves
CJ
Massing
BG
Wadsworth
LD
Rogers
PCJ
Kalousek
DK
Childhood myelodysplasia: Suggested classification as myelodysplastic syndromes based on laboratory and clinical findings.
Am J Pediatr Hematol Oncol
12
1990
63
29
Owen G, Lewis IJ, Morgan M, Robinson A, Stevens RF: Prognostic factors in juvenile chronic granulocytic leukemia. Br J Cancer 66:S68, 1992 (suppl XVIII)
30
Aricò
M
Bossi
G
Schirò
R
Galimberti
M
Longoni
D
Macchia
P
Miniero
R
Natale
D
Pession
A
Pillon
M
Santoro
N
Tontoli
N
Rusca
MP
Biondi
A for the AIEOP
Juvenile chronic myelogenous leukemia: Report of the Italian Registry.
Hematologica
78
1993
264
31
Donadieu
J
Stephan
JL
Blanche
S
Cavazzana-Calvo
M
Baruchel
A
Herbelin
C
Benkerrou
M
Thomas
C
Girault
D
Fischer
A
Treatment of juvenile chronic myelomonocytic leukemia by allogeneic bone marrow transplantation.
Bone Marrow Transplant
13
1994
777
32
Chan
H
Estrov
Z
Weitzman
S
Freedman
MH
The value of intensive combination chemotherapy for juvenile chronic myelogenous leukemia.
J Clin Oncol
5
1987
1960
33
Festa
R
Shende
A
Lanzkowski
P
Juvenile chronic myelocytic leukemia: Experience with intensive chemotherapy.
Med Pediatr Oncol
18
1990
311
34
Miyauchi
J
Asada
M
Sasaki
M
Tsunematsu
Y
Kojima
S
Mizutani
S
Mutations of the N-ras gene in juvenile chronic myelogenous leukemia.
Blood
83
1994
2248
35
Toren
A
Mandel
M
Amariglio
N
Hakim
Y
Brok-Simoni
F
Rechavi
G
Neumann
Y
Ramot
B
Lack of bcr rearrangement in juvenile chronic myeloid leukemia.
Med Pediatr Oncol
19
1991
493
36
Whetherall DJ, Brown MJ: Juvenile chronic myeloid leukemia: Lancet i:526, 1970
37
Hazani
A
Barak
Y
Berant
M
Bar-Maor
A
Congenital juvenile chronic myelogenous leukemia: Therapeutic trial with interferon alpha-2.
Med Pediatr Oncol
21
1993
73
38
Amenomori
T
Tomonaga
M
Yoshida
Y
Kuriyama
K
Matsuo
T
Jinnai
I
Ichimaru
M
Omiya
A
Tsuji
Y
Cytogenetic evidence for partially committed myeloid progenitor cell origin of chronic myelomonocytic leukemia and juvenile chronic myeloid leukemia: Both granulocyte-macrophage precursors and erythroid precursors carry identical markers.
Br J Haematol
64
1986
539
39
Inoue
S
Shibata
T
Ravindranath
Y
Gohle
N
Clonal origin of erythroid cells in juvenile chronic myelogenous leukemia.
Blood
69
1987
975
40
Shannon
KM
Watterson
J
Johnson
P
O'Connell
P
Lange
B
Shah
N
Steinherz
P
Kan
YW
Priest
JR
Monosomy 7 myeloproliferative disease in children with neurofibromatosis type 1: Epidemiology and molecular analysis.
Blood
79
1992
1311
41
Kaneko
Y
Maseki
N
Sakurai
M
Shibuya
A
Shinohara
T
Fujimoto
T
Kanno
H
Nishikawa
A
Chromosome pattern in juvenile chronic myelogenous leukemia, myelodysplastic syndrome, and acute leukemia associated with neurofibromatosis.
Leukemia
3
1989
36
42
Case records of the Massachusetts General Hospital
Case 37-1994.
N Engl J Med
331
1994
1005
43
Massaad
L
Prieur
M
Leonard
C
Dutrillaux
B
Biclonal chromosome evolution of chronic myelomonocytic leukemia in a child.
Cancer Genet Cytogenet
44
1990
131
44
Rachmilevitz
B
Rachmilevitz
M
The clinical significance of serum transcobalamin in myeloid leukemia and other myeloproliferative disorders.
Isr J Med Sci
13
1977
710
45
Cannat
A
Seligmann
M
Immunoglobulin abnormalities in juvenile myelomonocytic leukemia.
Br J Med
1
1973
71
46
Puchkova
GP
Prigogina
EL
Fleishmann
EV
Drosdova
TS
Mayakova
SA
Peterson
IS
Chromosome abnormalities in chronic myeloid leukemia in children.
Hum Genet
64
1983
257
47
Ghione
F
Mecucci
C
Symann
M
Michaux
J-L
Casteels-Van
Daele M
Van Den Berghe
H
Cytogenetic investigations in childhood chronic myelocytic leukemia.
Cancer Genet Cytogenet
20
1986
317
48
Brodeur
GM
Dow
LW
Williams
DL
Cytogenetic features of juvenile chronic myelogenous leukemia.
Blood
53
1979
812
49
Lau
RC
Squire
J
Brisson
L
Kamel-Reid
S
Grunberger
T
Dubé
I
Letarte
M
Shannon
K
Freedman
M
Lymphoid blast crisis of B-lineage phenotype with monosomy 7 in a patient with juvenile chronic myelogenous leukemia (JCML).
Leukemia
8
1994
903
50
Pui
CH
Aricò
M
Isotretinoin for juvenile chronic myelogenous leukemia.
N Engl J Med
332
1995
1520
51
Suda
T
Miura
H
Mizoguchi
H
Characterization of hemopoietic precursor cells in juvenile-type chronic myelocytic leukemia.
Leuk Res
6
1982
43
52
Emanuel
P
Bates
LJ
Castleberry
RP
Gualtieri
RJ
Zuckerman
KS
Selective hypersensitivity to granulocyte macrophage colony stimulating factor by juvenile chronic myeloid leukemia hemopoietic progenitors.
Blood
77
1991
925
53
Freedman
M
Cohen
A
Grunberger
T
Bunin
N
Luddy
RE
Saunders
EF
Shahidi
N
Lau
A
Estrov
Z
Central role of tumor necrosis factor, GM-CSF, and interleukin-1 in the pathogenesis of juvenile chronic myelogenous leukemia.
Br J Haematol
80
1992
40
54
Bagby
GC
Dinarello
CA
Neerhout
RC
Ridgway
D
McCall
E
Interleukin 1-dependent paracrine granulopoiesis in chronic granulocytic leukemia of the juvenile type.
J Clin Invest
82
1988
1430
55
Emanuel
PD
Bates
LJ
Zu
S-W
Castlebery
R
Gualtieri
RJ
Zuckerman
KS
The role of monocyte-derived hemopoietic growth factors in the regulation of myeloproliferation in juvenile chronic myelogenous leukemia.
Exp Hematol
19
1991
1017
56
Busque
L
Gililand
DG
Prchal
JT
Sieff
CA
Weinstein
HJ
Sokol
JM
Belickova
M
Wayne
AS
Zuckerman
KS
Sokol
L
Castleberry
RP
Emanuel
PD
Clonality in Juvenile chronic myelogenous leukemia.
Blood
85
1995
21
57
Lanfrancone
L
Pelicci
G
Brizzi
MF
Arouica
MG
Casciari
C
Giuli
S
Pegoraro
L
Pawson
T
Pelicci
PG
Overexpression of the Shc proteins potentiates the proliferative response to the granulocyte-macropahge colony-stimulating factor and recruitment of Grb2/SoS and Grb2/p140 complexes to the B receptor subunit.
Oncogene
10
1995
907
58
Hirsch-Ginberg
C
LeMaistre
AC
Kantarjian
H
Talpaz
M
Cork
A
Frereich
EJ
Trujillo
JM
Lee
MS
Stass
SA
RAS mutations are rare events in Philadelphia chromosome-negative/bcr gene rearrangement-negative chronic myelogenous leukemia, but are prevalent in chronic myelomonocytic leukemia.
Blood
76
1990
1214
59
Tien
H
Wang
C-H
Chuang
S-M
Chow
J-M
Lee
F-Y
Liu
M-C
Chen
Y-C
Shen
M-C
Lin
D-T
Lin
K-H
Cytogenetic studies, ras mutation and clinical characteristics in primary myelodysplastic syndrome. A study of 68 Chinese patients in Taiwan.
Cancer Genet Cytogenet
74
1994
40
60
Kalra
R
Paderanga
DC
Olson
K
Shannon
KM
Genetic analysis is consistent with the hypothesis that NF1 limits myeloid cell growth through p21ras.
Blood
84
1994
3435
61
Shannon
KM
O'Connell
P
Martin
G
Paderanga
D
Olson
K
Dinndorf
P
McCormick
F
Loss of the normal NF1 allele from the bone marrow of children with type 1 neurofibromatosis and malignant myeloid disorders.
N Engl J Med
330
1994
597
62
Bollag
G
Clapp
DW
Shih
S
Adler
F
Zhang
YY
Thompson
P
Lange
B
Freedman
M
McCormick
F
Jacks
T
Shannon
K
Loss of NF1 results in activation of the Ras signaling pathway and leads to aberrant growth in haematopoietic cells.
Nat Genet
12
1996
144
63
Largaespada
DA
Brannan
CI
Jenkins
NA
Copeland
NG
NF1 deficiency causes Ras-mediated granulocyte/macrophage colony stimulating factor hypersensitivity and chronic myeloid leukemia.
Nat Genet
12
1996
137
64
Jack
T
Shih
S
Schmitt
EM
Bronson
RT
Bernards
A
Weinberg
RA
Tumorigenic and development consequences of a targeted nf1 in the mouse.
Nat Genet
7
1994
353
65
Quelle
FW
Sato
N
Witthuhun
BA
Inhorn
RC
Eder
M
Miyajima
A
Griffin
JD
Ihle
JN
JAK2 associates with the beta chain of the receptor for granulocyte-macrophage colony stimulating factor, and its activation requires the membrane-proximal region.
Mol Cell Biol
14
1994
4335
66
Schirò
R
Longoni
D
Rossi
V
Maglia
O
Doni
A
Arsura
M
Carrara
G
Masera
G
Vannier
O
Dinarello
CA
Biondi
A
Suppression of juvenile chronic myelogenous leukemia colony growth factor by interleukin-1 receptor antagonist.
Blood
83
1994
460
67
Papayannopolou
T
Nakamoto
B
Anagnou
NP
Chui
D
Dow
L
Sanders
J
Expression of embryonic globins by erythroid cells in juvenile chronic myelocytic leukemia.
Blood
77
1991
2569
68
Privitera
E
Schirò
R
Longoni
D
Ronchi
A
Rambaldi
A
Bernasconi
S
Ottolenghi
S
Masera
G
Biondi
A
Constitutive expression of GATA-1, EPOR, alfa-globin, and gamma-globin genes in myeloid clonogenic cells from juvenile chronic myelocytic leukemia.
Blood
86
1995
323
69
Miles
DK
Feedman
MH
Stephens
K
Pallavicini
M
Sievers
EL
Weaver
M
Grunberger
T
Thompson
P
Shannon
KM
Pattern of hematopoietic lineage involvement in children with neurofibromatosis, type 1 and malignant myeloid disorders.
Blood
88
1996
4314
70
Fialkow
PJ
Denman
AM
Jacobson
RJ
Lowenthal
MN
Chronic myelocytic leukemia: Origin of some lymphocytes from leukemic stem cells.
J Clin Invest
62
1978
815
71
Lapidot
T
Gruneberger
T
Vormoor
J
Estrov
Z
Kollet
O
Bunin
N
Zaizov
R
Williams
DE
Freedman
MH
Identification of human juvenile chronic myelogenous leukemic stem cells capable of initiating the disease in primary and secondary SCID mice.
Blood
88
1996
2655
72
Hutter
SJ
Hecht
F
Kaiser-McCaw
B
Hays
T
Baranko
P
Cohen
J
Durie
B
Bone marrow monosomy 7: Hematologic and clinical manifestations in childhood and adolescence.
Hematol Oncol
2
1984
5
73
Freedman
M
Estrov
Z
Chan
E
Juvenile chronic myelogenous leukemia.
Am J Ped Hematol Oncol
10
1988
261
74
Gadner
H
Haas
OA
Experience in pediatric myelodysplastic syndromes.
Hematol Oncol North Am
6
1992
655
75
Luna-Finemann
S
Shannon
K
Ortega
J
Steinharz
P
Davis
J
Sanders
J
Masterson
M
Lange
BJ
De novo childhood preleukemic conditions: Preliminary data from a retrospective study of 172 cases.
Blood
86
1995
51a
76
Butcher
M
Frenck
R
Emperor
J
Paderanga
D
Maybee
D
Shannon
K
Molecular evidence that childhood monosomy 7 syndrome is distinct from juvenile chronic myelogenous leukemia and other childhood myeloproliferative disorders.
Genes Chromosom Cancer
12
1995
50
77
Le Beau MM, Larson RA, Green ED, Shannon KM: Analysis of juvenile chronic myelogenous leukemia (JCML) bone marrows for submicroscopic deletions on 7q. Blood (in press)
78
Hasle
H
Kerndrup
G
Yssing
M
Clausen
N
Ostergaard
E
Jacobsen
N
Brock
Jacobsen B
Intensive chemotherapy in childhood myelodysplastic syndrome. A comparison with results in acute myeloid leukemia.
Leukemia
10
1996
1269
79
Castleberry
RP
Emanuel
PD
Zuckerman
KS
Cohn
S
Strauss
L
Byrd
R
Homans
A
Chaffee
S
Nitschke
R
Gualtieri
RJ
A pilot study of isotretinoin in the treatment of juvenile chronic myelogenous leukemia.
N Engl J Med
331
1994
1680
80
Talpaz
M
Kantarjian
HM
McCredie
K
Trujillo
JM
Keating
M
Gutterman
JU
Hematologic remission and cytogenetic improvement induced by recombinant human interferon alpha in chronic myelogenous leukemia.
N Engl J Med
314
1986
1065
81
Maybee D, Dubow Krischer J, Buchnan G, Bowman W, Dickerman J, Hakami N, Laver J, Steuber P, Toledano S, Vietti T, Wilkinson R: Unusual toxicity of high dose alpha interferon in the treatment of juvenile chronic myelogenous leukemia. Proc Am Soc Clin Oncol 1:285, 1992 (abstr 950)
82
Shaw
NJ
Eden
OB
Juvenile chronic myelogenous leukemia and neurofibromatosis infancy presenting as ocular hemorrhage.
Pediatr Hematol Oncol
6
1989
23
83
Locatelli
F
Niemeyer
C
Angelucci
E
Bender-Gotze
C
Burdach
S
Ebell
W
Frederich
W
Hasle
H
Herman
J
Jacobsen
N
Klingebiel
T
Kremens
B
Mann
G
Pession
A
Peters
C
Schmitd
HJ
Stary
J
Suttorp
M
Uderzo
C
Van 'T Veer-Korthof
ET
Vosse
J
Zecca
M
Zimmermann
M and members of the EWOG-MDS
Allogeneic bone marrow transplantation for chronic myelomonocytic leukemia in childhood: a report from the European Working Group on myelodysplastic Syndrome in Childhood (EWOG-MDS).
J Clin Oncol
15
1997
556
84
Locatelli
F
Pession
A
Bonetti
F
Maserati
E
Prete
L
Pedrazzoli
P
Zecca
M
Prete
A
Paolucci
P
Cazzola
M
Busulfan cyclophosphamide and melphalan as conditioning regimen for bone marrow transplantation in children with myelodysplastic syndromes.
Leukemia
8
1994
844
85
Rubie
H
Attal
M
Demur
C
Brousset
P
Duchayne
E
Rigal-Huguet
F
Dastugue
N
Robert
A
Intensified conditioning regimen with busulfan followed by allogeneic BMT in children with myelodysplasia.
Bone Marrow Transplant
13
1994
759
86
Akashi
K
The role of interleukin-4 in the negative regulation of leukemia cell growth.
Leuk Lymphoma
9
1993
205
87
Geissler
K
Öhler
L
Födinger
M
Virgolini
I
Leimer
M
Kabrna
E
Kollars
M
Skoupy
S
Bohle
B
Rogy
M
Lechner
K
Interleukin 10 inhibits growth and granulocyte macrophage colony-stimulating factor production in chronic myelomonocytic leukemia cells.
J Exp Med
184
1996
1
88
Gjeritsen
MK
Bakka
A
Breivik
J
Saeterdal
I
Solheim
BG
Soereide
O
Thorsby
E
Gaudernack
G
Vaccination with mutant ras peptides and induction of T-cell responsiveness in pancreatic carcinoma patients carrying the corresponding RAS mutation.
Lancet
346
1995
1399
89
Gibb
JB
Oliff
A
Kohl
NE
Farnesyltransferase inhibitors: Ras research yields a potential cancer therapeutic.
Cell
77
1994
175
90
Hancock
JF
Anti-Ras drugs come of age.
Curr Biol
3
1993
770
91
Kohl
N
Mosser
S
deSolms
S
Giuliani
E
Pompliano
D
Graham
S
Smith
R
Scolnick
E
Oliff
A
Gibbs
JB
Selective inhibition of ras-dependent transformation by a farnesyltransferase inhibitor.
Science
260
1993
1934
92
James
G
Goldstein
J
Brown
M
Rawson
T
Somers
T
McDowell
R
Crowley
C
Lucas
B
Levinson
A
Marsters
J
Benzodiazepine peptidomimetics: Potent inhibitors of ras farnesylation in animal cells.
Science
260
1993
1937
93
Kohl
NE
Wilson
FR
Mosser
SD
Giuliani
E
DeSolms
SJ
Conner
MW
Anthony
NJ
Holtz
WJ
Gomez
RP
Lee
TJ
Smith
RL
Graham
SL
Hartman
GD
Gibbs
JB
Oliff
A
Protein farnesyltransferase inhibitors block the growth of ras-dependent tumors in nude mice.
Proc Natl Acad Sci USA
91
1994
9141
94
Sepp-Lorenzino
L
Ma
Z
Rands
E
Kohl
NE
Gibbs
JB
Oliff
A
Rosen
N
A peptidomimetic inhibitor of farnesyl protein transferase blocks the anchorage dependent and independent growth of human tumor cell lines.
Cancer Res
55
1995
5302
95
Casper
J
Camitta
B
Truitt
R
Baxter-Lowe
LA
Bunin
N
Lawton
C
Murray
K
Hunter
J
Pietryga
D
Garbrecht
F
Keever-Taylor
C
Drobyski
W
Horowitz
M
Flomenberg
N
Ash
R
Unrelated bone marrow donor transplant for children with leukemia or myelodysplasia.
Blood
85
1995
2354
96
Luna-Fineman
S
Shannon
KM
Lange
BJ
Childhood monosomy 7: Epidemiology, biology, and mechanistic implications.
Blood
85
1995
1985
97
Sanders
J
Buckner
CD
Thomas
ED
Fleischer
R
Sullivan
KM
Appelbaum
FA
Storb
R
Allogeneic marrow transplantation for children with juvenile chronic myelogenous leukemia.
Blood
71
1988
1144
98
Bunin
NJ
Casper
JT
Lawton
C
Murray
K
Camitta
B
Greenwood
M
Geil
J
Ash
RC
Allogeneic bone marrow transplantation using T cell depletion for patients with juvenile chronic myelogenous leukemia without HLA-identical siblings.
Bone Marrow Transplant
9
1992
119
99
Katz
F
Hann
I
Kinsey
S
Ball
S
Morgan
G
Chessels
J
Assessment of graft status following allogeneic bone marrow transplantation for haematological disorders in children using locus-specific minisatellite probes.
Br J Haematol
83
1993
473
100
Urban
C
Schwinger
W
Slavc
I
Schmid
C
Gamillscheg
A
Lackner
H
Hauer
C
Pakisch
B
Busulfan/cyclophosphamide plus bone marrow transplantation is not sufficient to eradicate the malignant clone in juvenile chronic myelogenous leukemia.
Bone Marrow Transplant
5
1990
353
101
Peltier
JY
Giralut
D
Debre
M
Galacteros
F
Fischer
A
Girot
R
Donor for BMT with hemoglobin H disease.
Bone Marrow Transplant
12
1993
81
102
Jacobson
BK
Kalayoglu
M
Effective early treatment of hepatic venocclusive disease with a central splenorenal shunt in an infant.
J Pediatr Surg
27
1992
531
103
Rassam
SMB
Katz
F
Chessels
JM
Morgan
G
Successful allogeneic bone marrow transplantation in juvenile CML: Conditioning or graft-versus-leukemia effect?
Bone Marrow Transplant
11
1993
247
Sign in via your Institution