Juvenile myelomonocytic leukemia (JMML) is an early childhood disease for which there is no effective therapy. Therapy with 13-cis retinoic acid or low-dose chemotherapy can induce some responses, but neither mode is curative. Stem cell transplantation can produce lasting remissions but is hampered by high rates of relapse. The pathogenesis of JMML involves deregulated cytokine signal transduction through the Ras signaling pathway, with resultant selective hypersensitivity of JMML cells to granulocyte-macrophage colony-stimulating factor (GM-CSF). A JMML mouse model, achieved through homozygous deletion of the neurofibromatosis gene, confirmed the involvement of deregulated Ras in JMML pathogenesis. With this pathogenetic knowledge, mechanism-based treatments are now being developed and tested. Ras is critically dependent on a prenylation reaction for its signal transduction abilities. Farnesyltransferase inhibitors are compounds that were developed specifically to block the prenylation of Ras. Two of these compounds, L-739,749 and L-744,832, were tested for their ability to inhibit spontaneous JMML granulocyte-macrophage colony growth. Within a dose range of 1 to 10 μmol/L, each compound demonstrated dose-dependent inhibition of JMML colony growth. An age-matched patient with a different disease and GM-CSF–stimulated normal adult marrow cells also demonstrated dose-dependent inhibitory effects on colony growth, but they were far less sensitive to these compounds than JMML hematopoietic progenitors. Even if the addition of L-739,749 were delayed for 5 days, significant inhibitory effects would still show in JMML cultures. These results demonstrate that a putative Ras-blocking compound can have significant growth inhibitory effects in vitro, perhaps indicating a potential treatment for JMML.

Juvenile myelomonocytic leukemia (JMML), previously termed juvenile chronic myelogenous leukemia, is a rare, clonal myeloproliferative/myelodysplastic disorder of infancy and early childhood.1-5 It converts to an acute leukemia-type blast crisis in only a few patients. Nevertheless the mortality rate is high because of the infiltration of monocytic cells into nonhematopoietic organs such as the lungs and the intestines, leading to organ failure, bleeding, and infection. Intensive chemotherapeutic regimens have largely proved futile in inducing durable remissions.6-9Low-to-intermediate dose chemotherapy may be temporarily effective in a proportion of patients, but it has generally not been shown to result in long-term disease control.10,11 Although 13-cis retinoic acid has an overall response rate of 40% to 50% and minimal toxicity, it is associated with extended responses in <10% of patients.12,13 Stem cell transplantation is the only therapy capable of producing durable remissions.14-17Unfortunately, the relapse rate remains high, and the overall survival rate is approximately 25%. Clearly, more effective therapy is sorely needed for this disease.

The pathogenesis of JMML has been linked to deregulated signal transduction through the Ras signaling pathway. This deregulation results in JMML cells demonstrating hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF) in in vitro dose-response assays.18-20 This hypersensitivity is selective because the responsiveness of JMML cells to IL-3 and G-CSF is normal.18 The family of Ras proteins acts as the master switch in transducing signals from the cell surface to the nucleus.21-24 Activating mutations of the RAS gene are observed in 15% to 30% of patients with JMML.25-28One of the major inactivators of Ras within cells is the neurofibromin protein, encoded by the neurofibromatosis type 1 tumor suppressor gene (NF1).29-31 Neurofibromin is a GTPase-activating protein, and it serves to hydrolyze Ras from its active GTP-bound state to its inactive GDP-bound state. The incidence of clinically apparent neurofibromatosis in patients with JMML is a striking 10% to 15%,4,32-34 compared with a general incidence of 1 in 3500. Many patients with JMML and neurofibromatosis demonstrate loss of heterozygosity at the NF1 locus.35-38 In addition to the 10% to 15% of patients with clinical evidence of neurofibromatosis, another 15% with JMML harbor NF1 mutations within their leukemic cells but do not have outward clinical manifestations.39 Although a causal relationship between the activating RAS mutations and the pathogenesis of JMML has yet to be established, RAS mutations and NF1abnormalities do appear to be mutually exclusive.28,39Conversely, Nf1 mutations have been proven causal in a mouse model of JMML. Homozygous deletion of Nf1 in mice leads to embryonic death.40 However, the hematopoietic fetal liver cells from these embryos demonstrate the same selective hypersensitivity to GM-CSF as do JMML cells, and transplantation of these cells into irradiated recipient mice results in the development of a myeloproliferative disorder similar to the human JMML syndrome41,42 and characterized by activated Ras-MAP kinase signaling in hematopoietic cells.43 Furthermore, recent studies show that murine cells lacking Nf1 are critically dependent on GM-CSF for growth.44 

Given these compelling data linking JMML pathogenesis to deregulated GM-CSF signal transduction through the Ras intracellular pathway, it seems reasonable to begin to explore mechanism-based therapy for JMML. Because JMML hematopoietic progenitor cells do not produce sufficient GM-CSF themselves to sustain in vitro colony growth, JMML is not an autocrine-driven disease.45 Rather, because of their inherent hypersensitivity to GM-CSF, JMML progenitors are dependent on the basal production of GM-CSF from monocytes.45 IL-10 has been shown to inhibit the monocytic production of GM-CSF and specifically to inhibit JMML cell growth.46 The GM-CSF antagonist and apoptotic agent, E21R, has also been shown to inhibit JMML in vitro cell growth and JMML cell engraftment in immunodeficient mice.47-49 Finally, 1 of 2 recently developed50,51 GM-CSF/diphtheria toxin fusion proteins has been shown to inhibit JMML cell growth in vitro.52 It is hypothesized that most of these potential therapies interfere with GM-CSF–cell interactions at the JMML cell surface. Whether any of these potential therapies can actually abolish the entire malignant clone is a matter of ongoing investigation.

Another feasible way to block the GM-CSF hypersensitive growth of JMML cells is to block the intracellular signaling pathway. For Ras to be active as a master switch for signal transduction, it must localize to the inner surface of the plasma membrane; this occurs after a series of posttranslational modifications. The first obligatory step in this series, which is essential for Ras cell-transforming activity, is the addition of a 15-carbon isoprenyl (farnesyl) group to Ras through a covalent link.53-58 The addition of the farnesyl moiety to the cysteine residue of the COOH-terminal CAAX motif (C, cysteine; A, usually an aliphatic residue; X, any other amino acid) is catalyzed by the enzyme farnesyl-protein transferase (FPTase). Several inhibitors of FPTase, representing broad structural diversity, have been synthesized.59,60 Some of these compounds, now termed farnesyltransferase inhibitors (FTIs), have been evaluated in several different in vitro and in vivo preclinical systems and have demonstrated significant antitumor effects.61-68 They have demonstrated an ability to inhibit the Ras-induced transformation of tissue culture cells and several cancer cell lines (primarily solid tumor types) and to block the proliferation of Ras-activated xenografts in nude mice. Further, FTIs have shown efficacy in RAS-driven transgenic mouse models of mammary and salivary carcinomas in which theRAS expression was forced by a mammary tumor virus. Finally, FTIs have demonstrated efficacy in blocking some of the phenotypic changes in NF1-deficient cells.69 70 Given this background of the developmental design of the FTIs and the pathogenetic mechanisms involving RAS and NF1 in JMML, the goal of this study was to determine the effectiveness of the CAAX peptidomimetic FPTase inhibitors L-739,749 and L-744,832 to abrogate JMML cell growth in vitro.

Acquisition of donor samples

With the approval of the respective institutional review boards and after obtaining parental consent, peripheral blood samples, bone marrow samples, or both were obtained from children with JMML and from a child with another disorder to serve as an age-matched control. The diagnosis of JMML was based on uniform criteria as agreed on by the International JMML Working Group71 and confirmed by the demonstration of selective hypersensitivity to GM-CSF in all children.

Normal controls were volunteer adults who donated bone marrow samples after informed consent and with the approval of the Institutional Review Board of the University of Alabama at Birmingham.

Mononuclear cell isolation and colony assays

Peripheral blood or marrow mononuclear cells for patient and control samples were isolated by density gradient centrifugation as described previously.18 Soft agar assays for granulocyte-macrophage colonies (CFU-GM) were established in 1-mL cultures of 0.3% agarose with McCoys' 5A medium plus nutrients and 15% fetal bovine serum as described previously.18,45,72 73 Cultures were incubated for 14 days at 37°C in a 5% CO2 atmosphere. CFU-GM colonies (≥40 cells/colony) were scored at day 14 using a dissecting microscope. GM-CSF (R & D, Minneapolis, MN) was added to the control cultures from the normal adult volunteer samples to stimulate growth at final concentrations of either 0.32 ng/mL or 2 ng/mL.

Addition of farnesyltransferase inhibitor to CFU-GM assays

The FTIs, L-739,749 and L-744,832, were supplied by Drs Allen Oliff and Jackson Gibbs of Merck Research Laboratories (West Point, PA) and were dissolved in a stock solution of 50% methanol at a concentration of 100 mmol/L and stored at −20°C. Three methods of adding the FTI to the CFU-GM assays were evaluated: (1) L-739,749 or L-744,832 was added only once, 24 hours after the cultures were established, duplicating the type of in vitro assay that established the effectiveness of 13-cis retinoic acid12 13; (2) the one-time dosing of FTI was delayed and was added at either day 3, day 5, or day 7 after the cultures were established; or (3) the cells were exposed to FTI before the establishment of the semisolid agar cultures. In the latter experiment, the mononuclear cells were placed in liquid suspension in McCoys' 5A medium with nutrients and 15% fetal bovine serum and then the FTI inhibitor was mixed in. Cells were exposed to the FTI inhibitor for 1, 3, or 5 days, then washed twice and placed in agar assays without any further addition of FTI inhibitor. In all types of cultures, the appropriate methanol dilutions for the respective FTI concentrations were simultaneously established to control for any effects on CFU-GM growth imposed by the methanol itself. Appropriate dilutions of the FTI or methanol control were made such that, for each dose, 100 μL of volume was spread uniformly over the agarose surface.

Samples from 12 patients with JMML were evaluated. All patients fulfilled the diagnostic criteria for JMML71 and demonstrated selective GM-CSF hypersensitivity of hematopoietic progenitor cells in vitro. Only 5 of 12 patient samples have been fully evaluated for NF1 or RAS abnormalities. Of the 5 fully studied, 2 had RAS mutations (both KRAS point mutations) and the other 3 had NF1 abnormalities (unpublished observations, Snyder RC, Emanuel PD and ref. 39).

In the first series of experiments, the FTI was added once 24 hours after the establishment of the cultures. Numbers of CFU-GM (>40 cells/colony) were counted, and the amount of inhibition by the FTI was calculated as a percentage of the maximal colony growth. As depicted in Figures 1 and 2, there was inhibition of JMML spontaneous CFU-GM colony growth at all concentrations of either FTI, L-739,749, or L-744,832. At concentrations ≥10 μmol/L FTI, there was complete abrogation of growth, and virtually no colonies were present in any patient sample. At a concentration of 1 μmol/L FTI, significant inhibition of CFU-GM colony growth was noted, and the colonies were smaller (fewer cells) than in the no addition controls. Because the FTI was in a methanol stock solution, appropriate methanol controls were also established, and these showed no significant effect on CFU-GM growth (data not shown).

Because of the nature of JMML, finding suitable controls for these experiments was problematic. We have demonstrated that JMML peripheral blood-derived or marrow-derived progenitor cells are essentially equivalent with regard to spontaneous growth patterns and to GM-CSF hypersensitivity patterns.18,45 Normal peripheral blood mononuclear cells did not show spontaneous colony growth, and normal marrow mononuclear cells did so only sporadically. Numerous normal age-matched marrow samples were not obtainable because of ethical concerns. Therefore, as controls for these experiments, we used normal adult marrow mononuclear cells stimulated with GM-CSF to simulate colony growth similar to that observed in JMML. We examined the inhibitory effects of the FTI on normal CFU-GM colony growth under several conditions: freshly obtained specimens stimulated with maximal concentrations of GM-CSF (2 ng/mL), freshly obtained specimens stimulated with threshold concentrations of GM-CSF (0.32 ng/mL), and specimens that had been stored in a liquid nitrogen environment for a period of time. The latter 2 conditions were attempted to simulate JMML conditions as much as possible. Because JMML cells are hypersensitive to GM-CSF, we stimulated normal cells with a GM-CSF concentration obtained from our dose-response curve data that represents a threshold concentration (0.32 ng/mL GM-CSF) at which JMML cells show an initial growth response.3 This was compared with effects at 2 ng/mL, which represents a maximal stimulation situation. Therefore, both physiologic and pharmacologic situations were simulated. No significant difference was noted in normal adult marrow cultures between the 2 GM-CSF concentrations (Figure1). In each situation the amount of colony growth inhibition by the FTI was always less than that seen in the JMML samples. The third situation simulated the JMML conditions in that several of our JMML samples had been stored in liquid nitrogen for various periods of time before evaluation. The storage conditions did have some increased effects on the amount of colony growth inhibition in the normal adult control samples (Figure 1), but these effects were still less than those seen in JMML samples. Taking into account the hypersensitivity to GM-CSF in JMML cells, these results are predictable. Finally, we were able to evaluate an age-matched marrow sample from a patient whose hematopoietic progenitor cells did in fact show spontaneous CFU-GM colony growth. This patient was ultimately diagnosed with thrombocytopenia with absent radii (TAR) syndrome and did not meet the clinical or culture criteria for JMML. The effect of L-739,749 on this patient's spontaneous colony growth was more similar to the effect of the FTI on the normal adult controls than to the effect of the FTI on JMML cells (Figure 1).

Fig. 1.

Inhibition of CFU-GM colony growth by the addition of L-739,749.

L-739,749 was a one-time addition 24 hours after establishment of the cultures, with respective concentrations of L-739,749 as indicated. Results in all cases are the mean of experiments performed in triplicate and are expressed as the percentage inhibition of maximal spontaneous CFU-GM colony growth. Error bars indicate the standard error of margin. Solid bars: Inhibition of JMML spontaneous CFU-GM colony growth. Results represent the mean of experiments performed in triplicate from 10 different patients with JMML. Hatched bars: Inhibition of GM-CSF–stimulated normal donor CFU-GM colony growth from marrow mononuclear cells obtained fresh from 5 different normal adult marrow donors. Results are expressed as the percentage inhibition of maximal CFU-GM colony growth stimulated by GM-CSF (either 2 ng/mL or 0.32 ng/mL). Diagonal bars: Inhibition of GM-CSF stimulated normal donor CFU-GM colony growth from marrow mononuclear cells that had been previously obtained from 3 different normal adult marrow donors and had been put in liquid nitrogen for long-term storage. Results are expressed as the percentage inhibition of maximal CFU-GM colony growth stimulated by GM-CSF (2 ng/mL). Open bars: Inhibition of spontaneous CFU-GM colony growth from the bone marrow mononuclear cells of an age-matched patient eventually diagnosed with thrombocytopenia with absent radii (TAR syndrome). The child did not meet the diagnostic criteria for JMML. CFU, colony-forming unit; CSF, colony-stimulating factor; GM, granulocyte macrophage; JMML, juvenile myelomonocytic leukemia.

Fig. 1.

Inhibition of CFU-GM colony growth by the addition of L-739,749.

L-739,749 was a one-time addition 24 hours after establishment of the cultures, with respective concentrations of L-739,749 as indicated. Results in all cases are the mean of experiments performed in triplicate and are expressed as the percentage inhibition of maximal spontaneous CFU-GM colony growth. Error bars indicate the standard error of margin. Solid bars: Inhibition of JMML spontaneous CFU-GM colony growth. Results represent the mean of experiments performed in triplicate from 10 different patients with JMML. Hatched bars: Inhibition of GM-CSF–stimulated normal donor CFU-GM colony growth from marrow mononuclear cells obtained fresh from 5 different normal adult marrow donors. Results are expressed as the percentage inhibition of maximal CFU-GM colony growth stimulated by GM-CSF (either 2 ng/mL or 0.32 ng/mL). Diagonal bars: Inhibition of GM-CSF stimulated normal donor CFU-GM colony growth from marrow mononuclear cells that had been previously obtained from 3 different normal adult marrow donors and had been put in liquid nitrogen for long-term storage. Results are expressed as the percentage inhibition of maximal CFU-GM colony growth stimulated by GM-CSF (2 ng/mL). Open bars: Inhibition of spontaneous CFU-GM colony growth from the bone marrow mononuclear cells of an age-matched patient eventually diagnosed with thrombocytopenia with absent radii (TAR syndrome). The child did not meet the diagnostic criteria for JMML. CFU, colony-forming unit; CSF, colony-stimulating factor; GM, granulocyte macrophage; JMML, juvenile myelomonocytic leukemia.

Close modal

Because the inhibitory effect of L-739,749 could be a nonspecific toxicity, we next sought to determine whether a second FTI would produce similar inhibitory effects on JMML spontaneous CFU-GM growth. L-744,832, which has been explored in the Nf1 mouse model of JMML, as discussed below, was also used in these human JMML sample experiments. As shown in Figure 2, L-744-832 had inhibitory effects on JMML CFU-GM growth similar to those of L-739,749. Samples from 4 patients were tested with this compound. One was tested with both L-739,749 and L-744,832, and the inhibitory results were almost identical (Figures 1 and 2).

Fig. 2.

Inhibition of CFU-GM colony growth by addition of L-744,832.

L-744,832 was a one-time addition 24 hours after establishment of the cultures, with respective concentrations of L-744,832 as indicated. Results are the mean of experiments performed in triplicate and expressed as the percentage inhibition of maximal spontaneous CFU-GM colony growth. Error bars indicate the standard error of margin. The solid bars show the inhibition of JMML spontaneous CFU-GM colony growth. Results represent the mean of experiments performed in triplicate from 4 different patients with JMML. CFU, colony-forming unit; GM, granulocyte macrophage; JMML, juvenile myelomonocytic leukemia.

Fig. 2.

Inhibition of CFU-GM colony growth by addition of L-744,832.

L-744,832 was a one-time addition 24 hours after establishment of the cultures, with respective concentrations of L-744,832 as indicated. Results are the mean of experiments performed in triplicate and expressed as the percentage inhibition of maximal spontaneous CFU-GM colony growth. Error bars indicate the standard error of margin. The solid bars show the inhibition of JMML spontaneous CFU-GM colony growth. Results represent the mean of experiments performed in triplicate from 4 different patients with JMML. CFU, colony-forming unit; GM, granulocyte macrophage; JMML, juvenile myelomonocytic leukemia.

Close modal

Given that either FTI was able to inhibit 100% of spontaneous CFU-GM growth in virtually all patients with JMML at concentrations of 10 μmol/L, in the next series of experiments we sought to determine whether variations in time of exposure to the FTI would affect the degree of growth inhibition. Therefore, L-739,749 was added at either days 1, 3, 5, or 7 after the establishment of cultures. Because of limited supplies of L-739,749, this type of multiple-dosing experiment could be performed on only 1 patient sample. Figure3 demonstrates a clear-cut, time-dependent loss of inhibitory effect of L-739,749 on spontaneous JMML CFU-GM colony growth.

Fig. 3.

Inhibition of JMML spontaneous CFU-GM colony growth by addition of L-739,749.

L-739,749 was a one-time addition at either 1, 3, 5, or 7 days after establishment of the cultures. Results, expressed as the percentage inhibition of maximal spontaneous CFU-GM colony growth, are the mean of experiments performed in triplicate from patient J97 with JMML. Concentrations of L-739,749 added are 1μmol/L (solid bars) and 10μmol/L (hatched bars). CFU, colony-forming unit; GM, granulocyte macrophage; JMML, juvenile myelomonocytic leukemia.

Fig. 3.

Inhibition of JMML spontaneous CFU-GM colony growth by addition of L-739,749.

L-739,749 was a one-time addition at either 1, 3, 5, or 7 days after establishment of the cultures. Results, expressed as the percentage inhibition of maximal spontaneous CFU-GM colony growth, are the mean of experiments performed in triplicate from patient J97 with JMML. Concentrations of L-739,749 added are 1μmol/L (solid bars) and 10μmol/L (hatched bars). CFU, colony-forming unit; GM, granulocyte macrophage; JMML, juvenile myelomonocytic leukemia.

Close modal

In a final series of experiments to determine the effectiveness of L-739,749 at inhibiting JMML CFU-GM growth, the JMML mononuclear cells from 1 patient were placed in liquid suspension in the presence of L-739,749 and in McCoys' 5A medium with nutrients and 15% fetal bovine serum. After either 1, 3, or 5 days of liquid suspension culture incubation with exposure to the FTI, the cells were removed, washed twice to remove any extracellular FTI, and placed in soft agar colony assay for 14 days. Trypan blue exclusion viability assays performed at all time points showed 95% to 100% viability, indicating that L-739,749 was not exerting any effect of cell necrosis on the cells (data not shown). Figure 4 shows that even as little as 24 hours of exposure to the FTI before a 14-day colony assay was sufficient to produce some minimal inhibition. More significant inhibition was obtained with 3 days of exposure before washout, and 5 days of exposure at doses of either 1 or 10 μmol/L of L-739,749 was sufficient to inhibit virtually all spontaneous CFU-GM colony growth in JMML.

Fig. 4.

Inhibition of JMML spontaneous CFU-GM colony growth by exposure of JMML mononuclear cells to L-739,749.

Exposure was in liquid suspension for either 1, 3, or 5 days before the establishment of colony assays. Cells were washed twice before the establishment of soft agar cultures to remove any extracellular L-739,749 in the liquid suspension. Results expressed are the percentage inhibition of maximal spontaneous CFU-GM colony growth, and they are the mean of experiments performed in triplicate from patient J49 with JMML. Concentrations of L-739,749 added are 1μmol/L (solid bars) and 10μmol/L (hatched bars).

Fig. 4.

Inhibition of JMML spontaneous CFU-GM colony growth by exposure of JMML mononuclear cells to L-739,749.

Exposure was in liquid suspension for either 1, 3, or 5 days before the establishment of colony assays. Cells were washed twice before the establishment of soft agar cultures to remove any extracellular L-739,749 in the liquid suspension. Results expressed are the percentage inhibition of maximal spontaneous CFU-GM colony growth, and they are the mean of experiments performed in triplicate from patient J49 with JMML. Concentrations of L-739,749 added are 1μmol/L (solid bars) and 10μmol/L (hatched bars).

Close modal

These experiments demonstrate that farnesyl-protein transferase inhibitors, initially developed for their potential to block Ras signal transduction, profoundly inhibited JMML cell growth in vitro. The value of this study stems from the fact that these cell culture studies were performed using primary cells from patient samples obtained from 12 different patients with confirmed diagnoses of JMML. These patients accurately represented a spectrum of JMML; some had NF1abnormalities, some had RAS abnormalities, and some probably had neither. The pathogenesis of JMML is intricately linked with the deregulation of signal transduction through the Ras signaling pathway.3,41,42 This deregulation, regardless of where in the Ras pathway the causative mutation(s) may occur, ultimately results in JMML cells demonstrating in vitro selective hypersensitivity to GM-CSF in granulocyte-macrophage colony-forming proliferation assays (CFU-GM).18-20 Activating RAS mutations are found in 15% to 30% of patients with JMML,25-28,39 andNF1 gene abnormalities are found in as many as 30% more.35-39 NF1 is a tumor-suppressor gene that encodes for the protein neurofibromin, which serves to inactivate Ras by hydrolyzing it from its active guanosine triphosphate (GTP)-bound state to an inactive guanosine diphosphate (GDP)-bound state.29-31 Loss of heterozygosity or other loss of function mutations of NF1, therefore, are essentially equivalent to activating RAS point mutations. All JMML samples examined in this study demonstrated similar growth inhibition by FTIs regardless of the identification within individual samples ofRAS mutations, NF1 abnormalities, or other as yet undefined mutations.

FTIs were developed to exploit Ras signal transduction physiology. For Ras proteins to serve as molecular master switches in mitogenic signal transduction, they must be in a membrane-bound, GTP-bound state. Obligatory for Ras cell-transforming activity is the prenylation reaction that attaches a farnesyl group (a 15-carbon isoprenyl group) to Ras to allow its association to the outer membrane of the cell, thus becoming a fully mature protein. Prenylated proteins share characteristic C-terminal sequences such as the CAAX motif. Farnesyl-protein transferase is 1 of 3 enzymes that catalyze protein prenylation. The others are geranylgeranyl protein transferase (GGPTase) types I and II. Selective inhibition of FPTase was explored and developed because geranylgeranylation of normal cellular proteins is 5 to 10 times more prevalent than farnesylation. Several farnesylated proteins play important roles in normal cells, including nuclear lamins essential for nuclear structural integrity, proteins of the retinal visual signal transduction system, the human homologue of the yeast molecular chaperone YDJ1, the skeletal muscle phosphorylase kinase, and others. The question that arises is how can the FTIs exert selective effects on tumor cells when so many normal cells also depend on farnesylation. The answer is likely not simple, but it may in part relate to the sensitivity of particular proteins to the FTIs, the ability of some proteins also to use GGPTase-I, and the dependency of the tumor cell on the Ras signal pathway rather than on the other redundant pathways in normal cells. Reports published recently74-77 indicate that KRAS-transformed cells may not be nearly as sensitive to FTIs as HRAS-transformed cells in which much of the preliminary testing of the FTIs was performed.60,61These reports demonstrate that KRAS proteins can be prenylated by geranylgeranyl protein transferase if the farnesylation pathway is blocked by an FTI.75,76 KRAS is the gene form most often mutated in human tumors. However, the specific RAS mutational status of human tumor cells may not necessarily correlate with their sensitivity to FTIs.63 In these cases the sensitivity to FTIs may lie in the dependency of the tumor cells on the Ras pathway, or it may raise the possibility that other farnesylated proteins besides Ras contribute to their biologic phenotypes. In this regard, recent studies78-81 implicate RhoB as a potential alternative or an additional target for the block of farnesylation. Therefore, though FTIs were developed to be specific compounds to block Ras signal transduction, it is becoming evident that inhibiting the farnesylation of proteins other than Ras may play a major role in their mechanism of action. In addition, since the initial development of the FTIs, more knowledge is emerging about the trafficking of Ras. A recent report82 shows that prenylated CAAX proteins do not, in fact, associate directly with the plasma membrane; rather, they associate with the endomembrane and are subsequently transported to the plasma membrane. Therefore, it is clear that the full function and mechanism of action of the FTIs remain to be elucidated.

The first FTI used in this study, L-739,749, is the methyl ester of the prodrug L-739,750. In the initial report61 on the development of these compounds, L-739,750 was shown to have profound effects on blocking prenylation in vitro, and this was highly selective for farnesylation over geranylgeranylation, as studied by prenylation assays. However, though L-739,750 could block prenylation on a subcellular level, it demonstrated minimal effectiveness in inhibiting whole-cell growth. Conversely, the methyl ester L-739,749 demonstrated less effectiveness in prenylation experiments (though selectively was maintained), but it did have profound effects specifically blocking Ras-transformed growth in whole-cell experiments. The inhibition of Ras-transformed Rat1 cell growth displayed a clear dose dependency of L-739,749 in the range from 1 μmol/L to 20 μmol/L.61Similarly, in the current study, we observed a dose-dependent inhibition of JMML spontaneous CFU-GM growth over the same drug concentration range of 1 to 20 μmol/L. The second FTI used in this study was L-744,832, the isopropyl ester of the prodrug L-739,750.62 It was employed to demonstrate that the effect of L-739,749 on JMML hematopoietic progenitor cells was not simply the result of a nonspecific toxicity event. The isopropyl ester and the methyl ester are similar compounds, and it is not unexpected that the results obtained in the JMML cell cultures showed similar levels of inhibition. L-744,832 has also been evaluated recently in theNf1-deficient mouse model by Shannon et al.83 They demonstrated dose-dependent inhibition of CFU-GM colony growth in the murine system at dose ranges of 1 to 20 μmol/L L-744,832, similar to the human system in this study and in the cell line testing previously reported. They were also able to demonstrate that L-744,832 could block H-Ras but not N-Ras farnesylation. GM-CSF-induced MAP kinase activation was blunted as well. Disappointingly, L-744,832 did not produce responses when used in the in vivo whole mouse with the myeloproliferative syndrome. This potential discrepancy between the in vitro human cell testing and the in vivo whole mouse testing is as yet unexplained. The answer may ultimately lie in the effect of FTIs on cellular events other than activating mutations of RAS, such as the recently reported FTI-induced activation of suppressed apoptotic pathways.67 In this regard, a report84describes the enhanced cell survival of JMML cells with a reversal of this effect using the GM-CSF antagonist analogue E21R. We recently described altered levels of the p85 regulatory subunit of phosphatidylinositol 3′-kinase in JMML cells,85 which has been linked to apoptotic pathways in other hematopoietic cells. Similar to the apoptosis-inducing effects seen with the GM-CSF antagonist analogue E21R, the possibility exists that some of the effectiveness of FTIs in JMML may result from their influences on the cell survival pathways. The possibility exists that theNf1-deficient mouse may not harbor the same alterations in the cell survival pathways as in the primary human JMML cells.

In summary, farnesyltransferase inhibitors demonstrate profound in vitro inhibitory effects on the cell growth of primary cells obtained from humans with JMML. Determining whether this is because of a specific Ras-related inhibition, other cellular effects, or multifactorial events will be the subject of ongoing investigations as the search for effective FTIs, and potentially other new therapeutic strategies for JMML, continues.

The authors thank Allen Oliff and Jackson Gibbs of Merck Research Laboratories for supplying the farnesyltransferase inhibitors evaluated in this study and for their helpful discussions and criticisms.

Supported in part by Public Health Service grants CA60407, CA25408, and CA80916; the J. L. Griffin Foundation; and the Cancer Research and Youth Outreach Network.

Reprints:Peter D. Emanuel, Division of Hematology/Oncology, Wallace Tumor Institute, Suite 520, University of Alabama at Birmingham, Birmingham, AL 35294-3300; e-mail:peter.emanuel@ccc.uab.edu.

The publication costs of this article were defrayed in part by page charge payment. Therefore, and solely to indicate this fact, this article is hereby marked “advertisement” in accordance with 18 U.S.C. section 1734.

1
Altman
AJ
Palmer
CG
Baehner
RL
Juvenile “chronic granulocytic leukemia”: a panmyelopathy with prominent monocytic involvement and circulating monocyte colony-forming cells.
Blood.
43
1974
341
350
2
Castro-Malaspina
H
Schaison
G
Passe
S
et al
Subacute and chronic myelomonocytic leukemia in children (juvenile CML): clinical and hematologic observations, and identification of prognostic factors.
Cancer.
54
1983
675
686
3
Emanuel
PD
Shannon
KM
Castleberry
RP
Juvenile myelomonocytic leukemia: molecular understanding and prospects for therapy.
Mol Med Today.
2
1996
468
475
4
Niemeyer
CM
Arico
M
Basso
G
et al
Chronic myelomonocytic leukemia in childhood: a retrospective analysis of 110 cases.
Blood.
89
1997
3534
3543
5
Arico
M
Biondi
A
Pui
C-H
Juvenile myelomonocytic leukemia.
Blood.
90
1997
479
488
6
Chan
HS
Estrov
Z
Weitzman
SS
Freedman
MH
The value of intensive combination chemotherapy for juvenile chronic myelogenous leukemia.
J Clin Oncol.
5
1987
1960
1967
7
Festa
RS
Shende
A
Lanzkowsky
P
Juvenile chronic myelocytic leukemia: experience with intensive combination chemotherapy.
Med Pediatr Oncol.
18
1990
311
316
8
Passmore
SJ
Hann
IM
Stiller
CA
et al
Pediatric myelodysplasia: a study of 68 children and a new prognostic scoring system.
Blood.
85
1995
1742
1750
9
Hasle
H
Kerndrup
G
Yssing
M
et al
Intensive chemotherapy in childhood myelodysplastic syndrome: a comparison with results in acute myeloid leukemia.
Leukemia.
10
1996
1269
1273
10
Lilleyman
JS
Harrison
JF
Black
JA
Treatment of juvenile chronic myeloid leukemia with sequential subcutaneous cytarabine and oral mercaptopurine.
Blood.
49
1977
559
562
11
Thomas
WJ
North
RB
Poplack
DG
Slease
RB
Duval-Arnold
B
Chronic myelomonocytic leukemia in childhood.
Am J Hematol.
10
1981
181
194
12
Castleberry
RP
Emanuel
PD
Zuckerman
KS
et al
A pilot study of isotretinoin in the treatment of juvenile chronic myelogenous leukemia.
N Engl J Med.
331
1994
1680
1684
13
Castleberry
RP
Chang
M
Maybee
D
Emanuel
PD
A phase II study of 13-cis retinoic acid (CRA) in juvenile myelomonocytic leukemia (JMML): a Pediatric Oncology Group (POG) study [abstract].
Blood.
90
1997
346a
14
Sanders
JE
Buckner
CD
Thomas
ED
et al
Allogeneic marrow transplantation for children with juvenile chronic myelogenous leukemia.
Blood.
71
1988
1144
1146
15
Smith
FO
Sanders
JE
Robertson
KA
Gooley
T
Sievers
EL
Allogeneic marrow transplantation for children with juvenile chronic myelogenous leukemia [abstract].
Blood.
84
1994
201a
16
Wagner
JE
Broxmeyer
HE
Byrd
RL
et al
Transplantation of umbilical cord blood after myeloablative therapy: analysis of engraftment.
Blood
79
1992
1874
1881
17
Locatelli
F
Niemeyer
C
Angelucci
E
et al
Allogeneic bone marrow transplantation for chronic myelomonocytic leukemia in childhood: a report from the European working group on myelodysplastic syndrome in childhood.
J Clin Oncol.
15
1997
566
573
18
Emanuel
PD
Bates
LJ
Castleberry
RP
Gualtieri
RJ
Zuckerman
KS
Selective hypersensitivity to granulocyte-macrophage colony-stimulating factor by juvenile chronic myeloid leukemia hematopoietic progenitors.
Blood.
77
1991
925
929
19
Lapidot
T
Cohen
A
Grunberger
T
Dick
J
Freedman
MH
Aberrant growth properties of juvenile chronic myelogenous leukemia (JCML) CD34+ cells in vitro and in vivo using SCID mouse assays [abstract].
Blood.
82
1993
197a
20
Cambier
N
Menot
ML
Fenaux
P
Wattel
E
Baruchel
A
Chomienne
C
GM-CSF hypersensitivity in CD34+ purified cells in juvenile and adult chronic myelomonocytic leukemia: effect of retinoids [abstract].
Blood.
86
1995
791a
21
Bos
JL
Ras oncogenes in human cancer: a review.
Cancer Res.
49
1989
4682
4689
22
Satoh
T
Nakafuku
M
Miyajima
A
Kaziro
Y
Involvement of ras p21 protein in signal transduction pathways from interleukin 2, interleukin 3, and granulocyte/macrophage colony-stimulating factor, but not from interleukin 4.
Proc Natl Acad Sci U S A.
88
1991
3314
3318
23
Rodenhuis
S
Ras and human tumors.
Semin Cancer Biol.
3
1992
241
247
24
Boguski
MS
McCormick
F
Proteins regulating Ras and its relatives.
Nature.
366
1993
643
654
25
Neubauer
A
Shannon
KM
Liu
E
Mutations of the ras proto-oncogenes in childhood monosomy 7.
Blood.
77
1991
594
598
26
Lubbert
M
Mirro
J
Jr
Kitchingman
G
et al
Prevalence of N-ras mutations in children with myelodysplastic syndromes and acute myeloid leukemia.
Oncogene.
7
1992
263
268
27
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
2254
28
Kalra
R
Paderanga
D
Olson
K
Shannon
KM
Genetic analysis is consistent with the hypothesis that NF1 limits myeloid cell growth through p21ras.
Blood.
84
1994
3435
3439
29
Bourne
HR
Sanders
DA
McCormick
F
The GTPase superfamily: a conserved switch for diverse cell functions.
Nature.
348
1990
125
129
30
Bourne
HR
Sanders
DA
McCormick
F
The GTPase superfamily: conserved structure and molecular mechanism.
Nature.
349
1991
117
127
31
Hall
A
The cellular functions of small GTP-binding proteins.
Science.
249
1990
635
640
32
Bader
JL
Miller
RW
Neurofibromatosis and childhood leukemias.
J Pediatr.
92
1978
925
929
33
Gadner
H
Haas
OA
Experience in pediatric myelodysplastic syndromes.
Hematol Clin North Am.
6
1992
655
672
34
Shannon
KM
Watteson
J
Johnson
P
et al
Monosomy 7 myeloproliferative disease in children with neurofibromatosis, type 1: epidemiology and molecular analysis.
Blood.
79
1992
1311
1318
35
Shannon
KM
O'Connell
P
Martin
GA
et al
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
601
36
Brodeur
GM
The NF1 gene in myelopoiesis and childhood myelodysplastic syndromes [editorial].
N Engl J Med.
330
1994
637
639
37
Miles
DK
Freedman
MH
Stephens
K
et al
Patterns of hematopoietic lineage involvement in children with neurofibromatosis type 1 and malignant myeloid disorders.
Blood.
88
1996
4314
4320
38
Side
L
Tayor
B
Cayouette
M
et al
Homozygous inactivation of the NF1 gene in bone marrow cells from children with neurofibromatosis type 1 and malignant myeloid disorders.
N Engl J Med.
336
1997
1713
1720
39
Side
LE
Emanuel
PD
Taylor
B
et al
Mutations of the NF1 gene in children with juvenile myelomonocytic leukemia without clinical evidence of neurofibromatosis, type 1.
Blood.
92
1998
267
272
40
Jacks
T
Shih
TS
Schmitt
EM
Bronson
RT
Bernards
A
Weinberg
RA
Tumour predisposition in mice heterozygous for a targeted mutation in Nf1.
Nat Genet.
7
1994
353
361
41
Largaespada
DA
Brannan
CI
Jenkins
NA
Copeland
NG
Nf1 deficiency causes Ras-mediated granulocyte/macrophage colony stimluating factor hypersensitivity and chronic myeloid leukaemia.
Nat Genet.
12
1996
137
143
42
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.
2
1996
144
148
43
Zhang
Y
Vik
TA
Ryder
JW
et al
Nf1 regulates hematopoietic progenitor cell growth and Ras signaling in response to multiple cytokines.
J Exp Med.
87
1998
1893
1902
44
Birnbaum
R
O'Marcaigh
A
Wardak
Z
et al
Interaction between NF1 and GMCSF in leukemogenesis and hematopoietic engraftment [abstract].
Blood.
90
1997
411a
45
Emanuel
PD
Bates
LJ
Zhu
S-W
Castleberry
RP
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
1024
46
Iversen
PO
Hart
PH
Bonder
CS
Lopez
AF
Interleukin (IL)-10, but not IL-4 or IL-13, inhibits cytokine production and growth in juvenile myelomonocytic leukemia cells.
Cancer Res.
57
1997
476
480
47
Hercus
TR
Bagley
CJ
Cambareri
B
et al
Specific human granulocyte-macrophage colony-stimulating factor antagonists.
Proc Natl Acad Sci U S A.
91
1994
5838
5842
48
Iversen
PO
To
LB
Lopez
AF
Apoptosis of hemopoietic cells by the human granulocyte-macrophage colony-stimulating factor mutant E21R.
Proc Natl Acad Sci U S A.
93
1996
2785
2789
49
Iversen
PO
Lewis
ID
Turczynowicz
S
et al
Inhibition of granulocyte-macrophage colony-stimulating factor prevents dissemination and induces remission of juvenile myelomonocytic leukemia in engrafted immunodeficient mice.
Blood.
90
1997
4910
4917
50
Perentesis
JP
Waddick
KG
Bendel
AE
et al
Induction of apoptosis in multidrug-resistant and radiation-resistant acute myeloid leukemia cells by a recombinant fusion toxin directed against the human granulocyte macrophage colony-stimulating factor receptor.
Clin Cancer Res.
3
1997
347
355
51
Frankel
AE
Hall
PD
Burbage
C
et al
Modulation of the apoptotic response of human myeloid leukemia cells to a diphtheria toxin granulocyte-macrophage colony-stimulating factor fusion protein.
Blood.
90
1997
3654
3661
52
Frankel
AE
Lilly
M
Kreitman
R
et al
Diphtheria toxin fused to granulocyte-macrophage colony-stimulating factor is toxic to blasts from patients with juvenile myelomonocytic leukemia and chronic myelomonocytic leukemia.
Blood.
92
1998
4279
4286
53
Willumsen
BM
Norris
K
Papageorge
AG
Hubbert
NL
Lowy
DR
Harvey murine sarcoma virus p21 ras protein: biological and biochemical signficance of the cysteine nearest the carboxy terminus.
EMBO J.
3
1984
2581
2585
54
Hancock
JF
Magee
AI
Childs
JE
Marshall
CJ
All Ras proteins are polyisoprenylated but only some are palmitoylated.
Cell.
57
1989
1167
1177
55
Jackson
JH
Cochrane
CG
Bourne
JR
Solski
PA
Buss
JE
Der
CJ
Farnesol modification of Kirsten-ras exon 4B protein is essential for transformation.
Proc Natl Acad Sci U S A.
7
1990
3042
3046
56
Kato
K
Cox
AD
Hisaka
MM
Graham
SM
Buss
JE
Der
CJ
Isoprenoid addition to Ras protein is the critical modification for its membrane association and transforming activity.
Proc Natl Acad Sci U S A.
89
1992
6403
6407
57
Newman
CMH
Magee
AI
Posttranslational processing of the ras superfamily of small GTP-binding proteins.
Biochim Biophys Acta.
1155
1993
79
96
58
Gibbs
JB
Oliff
A
Kohl
NE
Farnesyltransferase inhibitors: Ras research yields a potential cancer therapeutic.
Cell.
77
1994
175
178
59
Kohl
NE
Mosser
SD
deSolams
SJ
et al
Selective inhibition of ras-dependent transformation by a farnesyltransferase inhibitor.
Science.
260
1993
1934
1937
60
James
GL
Goldstein
JL
Brown
MS
et al
Benzodiazepine peptidomimetics: potent inhibitors of ras farnesylation in animal cells.
Science.
260
1993
1937
1942
61
Kohl
NE
Wilson
FR
Mosser
SD
et al
Protein farnesyltransferase inhibitors block the growth of ras-dependent tumors in nude mice.
Proc Natl Acad Sci U S A.
91
1994
9141
9145
62
Kohl
NE
Omer
CA
Conner
MW
et al
Inhibition of farnesyltransferase induces regression of mammary and salivary carcinomas in ras transgenic mice.
Nat Med.
1
1995
792
797
63
Sepp-Lorenzino
L
Ma
Z
Rands
E
et al
A peptidomimetic inhibitor of farnesyl: protein transferase blocks the anchorage-dependent and -independent growth of human tumor cell lines.
Cancer Res.
55
1995
5302
5309
64
Kohl
NE
Conner
MW
Gibbs
JB
Graham
SL
Hartman
GD
Oliff
A
Development of inhibitors of protein farnesylation as potential chemotherapeutic agents.
J Cell Biochem.
22(suppl)
1995
145
150
65
Gibbs
JB
Oliff
A
The potential of farnesyltransferase inhibitors as cancer chemotherapeutics.
Annu Rev Pharmacol Toxicol.
37
1997
143
166
66
Mangues
R
Corral
T
Kohl
NE
et al
Antitumor effect of a farnesyl protein transferase inhibitor in mammary and lymphoid tumors overexpressing N-ras in transgenic mice.
Cancer Res.
58
1998
1253
1259
67
Barrington
RE
Subler
MA
Rands
E
et al
A farnesyltransferase inhibitor induces tumor regression in transgenic mice harboring multiple oncogenic mutations by mediating alterations in both cell cycle control and apoptosis.
Mol Cell Biol.
18
1998
85
92
68
Nørgaard
P
Law
B
Joseph
H
et al
Treatment with farnesyl-protein transferase inhibitor induces regression of mammary tumors in transforming growth factor (TGF) α and TGFα/neu transgenic mice by inhibition of mitogenic activity and induction of apoptosis.
Clin Cancer Res.
5
1999
35
42
69
Yan
N
Ricca
C
Fletcher
J
Glover
T
Seizinger
BR
Manne
V
Farnesyltransferase inhibitors block the neurofibromatosis type I (NF1) malignant phenotype.
Cancer Res.
55
1995
3569
3575
70
Kim
HA
Ling
B
Ratner
N
Nf1-deficient mouse Schwann cells are angiogenic and invasive and can be induced to hyperproliferate: reversion of some phenotypes by an inhibitor of farnesyl protein transferase.
Mol Cell Biol.
17
1997
862
872
71
Niemeyer
CM
Fenu
S
Hasle
H
Mann
G
Stary
J
van Wering
E
Differentiating juvenile myelomonocytic leukemia from infectious disease [response].
Blood.
91
1998
365
72
Gualtieri
RJ
Castleberry
RP
Gibbons
J
et al
Cell culture studies and oncogene expression in juvenile chronic myelogenous leukemia.
Exp Hematol.
16
1988
613
619
73
Gualtieri
RJ
Emanuel
PD
Zuckerman
KS
et al
Granulocyte-macrophage colony-stimulating factor is an endogenous regulator of cell proliferation in juvenile chronic myelogenous leukemia.
Blood.
74
1989
2360
2367
74
James
GL
Goldstein
JL
Brown
MS
Polylysine and CVIM sequences of K-RasB dictate specificity of prenylation and confer resistance to benzodiazepine peptidomimetic in vitro.
J Biol Chem.
270
1995
6221
6226
75
James
G
Goldstein
JL
Brown
MS
Resistance of K-RasBV12 proteins to farnesyltransferase inhibitors in Rat1 cells.
Proc Natl Acad Sci U S A.
93
1996
4454
4458
76
Whyte
DB
Kirschmeieer
P
Hockenberry
TN
Nunez-Oliva
I
James
L
K- and N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors.
J Biol Chem.
272
1997
14,459
14,464
77
Rowell
CA
Kowalczyk
JJ
Lewis
MD
Garcia
AM
Direct demonstration of geranylgeranylation and farnesylation of Ki-Ras in vivo.
J Biol Chem.
272
1997
14,093
14,097
78
Prendergast
GC
Davide
JP
deSolms
SJ
et al
Farnesyltransferase inhibition causes morphological reversion of ras-transformed cells by a complex mechanism that involves regulation of the actin cytoskeleton.
Mol Cell Biol.
14
1994
4193
4202
79
Lebowitz
PF
Davide
JP
Prendergast
GC
Evi dence that farnesyl-transferase inhibitors suppress Ras transformation by interfering with Rho activity.
Mol Cell Biol.
15
1995
6613
6622
80
Lebowitz
PF
Casey
PJ
Prendergast
GC
Thissen
JA
Farnesyl-transferase inhibitors alter the prenylation and growth-stimulation function of RhoB.
J Biol Chem.
272
1997
15,591
15,594
81
Du
W
Lebowitz
PF
Prendergast
GC
Cell growth inhibition by farnesyltransferase inhibitors is mediated by gain of geranylgeranylated RhoB.
Mol Cell Biol.
19
1999
1831
1840
82
Choy
E
Chiu
VK
Silletti
J
et al
Endomembrane trafficking of Ras: the CAAX motif targets proteins to the ER and Golgi.
Cell.
98
1989
69
80
83
Mahgoub
N
Taylor
BR
Gratiot
M
et al
In vitro and in vivo effects of a farnesyltransferase inhibitor on Nf1-deficient hematopoietic cells.
Blood.
94
1999
2469
2476
84
Iversen
PO
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.
88
1996
2634
2639
85
Snyder
RC
Wiley
T
Castleberry
RC
Emanuel
PD
Increased levels of the p85α regulatory subunit of phosphatidylinisitol 3-OH kinase in cell lysates of juvenile myelomonocytic leukemia cells [abstract].
Blood.
90
1997
411a
Sign in via your Institution