Evaluation of dysregulation of the receptor tyrosine kinases Kit, Flt3, and Met in histiocytic sarcomas of dogs

Regina Zavodovskaya Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616.

Search for other papers by Regina Zavodovskaya in
Current site
Google Scholar
PubMed
Close
 MS
,
Albert T. Liao Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616.

Search for other papers by Albert T. Liao in
Current site
Google Scholar
PubMed
Close
 DVM, PhD
,
Cameron L. R. Jones Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616.

Search for other papers by Cameron L. R. Jones in
Current site
Google Scholar
PubMed
Close
,
Becky Yip Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616.

Search for other papers by Becky Yip in
Current site
Google Scholar
PubMed
Close
,
May B. Chien Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616.

Search for other papers by May B. Chien in
Current site
Google Scholar
PubMed
Close
 MS
,
Peter F. Moore Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA 95616.

Search for other papers by Peter F. Moore in
Current site
Google Scholar
PubMed
Close
 DVM, PhD
, and
Cheryl A. London Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California, Davis, CA 95616.

Search for other papers by Cheryl A. London in
Current site
Google Scholar
PubMed
Close
 DVM, PhD

Abstract

Objective—To evaluate canine histiocytic sarcoma cell lines and tumor samples for dysregulation of the Kit/stem-cell factor (SCF), Flt3/Flt3 ligand (Flt3L), and Met/hepatocyte growth factor (HGF) receptor tyrosine kinase signaling pathways, as these are known to contribute to the differentiation and survival of normal dendritic cells as well as malignant transformation of dendritic cells in mouse models.

Sample Population—4 histiocytic sarcoma tumor cell lines and 35 formalin-fixed histiocytic sarcoma specimens obtained from dogs.

Procedure—Histiocytic sarcoma cell lines were evaluated for expression of Kit/SCF, Flt3/Flt3L, and Met/HGF by use of reverse transcriptase-PCR procedures. Histiocytic sarcoma cell lines and tumor samples were evaluated for mutations in Kit, Flt3, and Met by use of PCR analysis of genomic DNA, followed by both sequencing and fluorescent PAGE for deletions or internal tandem duplications. The ability of the multitargeted split-kinase inhibitor SU11654 to block proliferation and induce apoptosis of histiocytic sarcoma cell lines was also evaluated.

Results—No mutations in Kit, Flt3, and Met were identified in any of the cell lines or tumor samples evaluated. Furthermore, SU11654 did not induce cellcycle arrest or apoptosis of histiocytic sarcoma lines, even at supratherapeutic doses.

Conclusions and Clinical Relevance—These data suggest that dysregulation of Kit/SCF, Flt3/Flt3L, and Met/HGF signaling pathways is unlikely to occur in histiocytic sarcomas of dogs and that inhibitors of the Kit, Flt3, and Met pathways are unlikely to provide clinical benefit to dogs with histiocytic sarcomas.

Abstract

Objective—To evaluate canine histiocytic sarcoma cell lines and tumor samples for dysregulation of the Kit/stem-cell factor (SCF), Flt3/Flt3 ligand (Flt3L), and Met/hepatocyte growth factor (HGF) receptor tyrosine kinase signaling pathways, as these are known to contribute to the differentiation and survival of normal dendritic cells as well as malignant transformation of dendritic cells in mouse models.

Sample Population—4 histiocytic sarcoma tumor cell lines and 35 formalin-fixed histiocytic sarcoma specimens obtained from dogs.

Procedure—Histiocytic sarcoma cell lines were evaluated for expression of Kit/SCF, Flt3/Flt3L, and Met/HGF by use of reverse transcriptase-PCR procedures. Histiocytic sarcoma cell lines and tumor samples were evaluated for mutations in Kit, Flt3, and Met by use of PCR analysis of genomic DNA, followed by both sequencing and fluorescent PAGE for deletions or internal tandem duplications. The ability of the multitargeted split-kinase inhibitor SU11654 to block proliferation and induce apoptosis of histiocytic sarcoma cell lines was also evaluated.

Results—No mutations in Kit, Flt3, and Met were identified in any of the cell lines or tumor samples evaluated. Furthermore, SU11654 did not induce cellcycle arrest or apoptosis of histiocytic sarcoma lines, even at supratherapeutic doses.

Conclusions and Clinical Relevance—These data suggest that dysregulation of Kit/SCF, Flt3/Flt3L, and Met/HGF signaling pathways is unlikely to occur in histiocytic sarcomas of dogs and that inhibitors of the Kit, Flt3, and Met pathways are unlikely to provide clinical benefit to dogs with histiocytic sarcomas.

Histiocytic sarcoma (also known as malignant histiocytosis) is a neoplastic disorder arising from cells of the histiocytic lineage (macrophages and dendritic cells).1–8 The disease is characterized by infiltration of many organs including the spleen, liver, lungs, lymph nodes, and bone marrow. Breeds of dog known to be at increased risk for this disease include Rottweilers, Golden Retrievers, Flat-Coated Retrievers, and Bernese Mountain Dogs.3 Diagnosis of histiocytic sarcoma is usually achieved through a combination of histologic examination and immunohistochemical evaluation of tissue samples to rule out other potential tumors. Results of a recent study1 indicated that the histiocytic lineage of histiocytic sarcoma can be reliably identified by a panel of markers; these tumors are characterized as CD1+, CD4, CD11c+, CD11d, MHC II+, and ICAM-1+. Unfortunately, histiocytic sarcoma is a rapidly aggressive disorder, and in most instances, affected patients die within 2 to 4 months of diagnosis. Recent evidence in dogs suggests that the chemotherapy agent lomustine has activity against this disease, inducing a 50% response rate, although these responses were of short duration (96 days) with a median survival time of 4 months (128 days).a Surgery and radiation therapy are often not viable therapeutic options because of the widespread distribution of histiocytic sarcoma in patients.

In contrast to the disease in dogs, histiocytic sarcoma is now considered to be an extremely rare entity in humans. This is because of the fact that many tumors in patients that were previously classified as histiocytic sarcoma have been reclassified as anaplastic large-cell lymphoma and are no longer considered to be of histiocytic origin.9–11 Virtually nothing is known regarding the etiopathology of histiocytic sarcoma in humans or dogs. As a consequence, it has been extremely difficult to identify novel therapeutic agents that may be effective in the treatment of this disease. Moreover, as investigations into the biology of canine neoplastic disorders are often based on findings in the human medical literature, the absence of information in humans has impaired progress in understanding molecular mechanisms that may be involved in development of histiocytic sarcoma.

Increasingly, it has been recognized that dysregulation of protein kinases, particularly among RTKs, is present in a number of human tumors and this phenomenon is just beginning to be investigated in regard to neoplasia in dogs. The growth factor receptors Kit and Flt3 are members of the so-called splitkinase family of RTKs.12–14 They share structural characteristics such as 5 immunoglobulinlike domains in the extracellular region, a juxtamembrane domain that negatively regulates receptor function, and 2 kinase domains separated by a kinase insert.15 The ligands for Kit and Flt3 are SCF and Flt3L, respectively.16–18 Both Kit and Flt3 play important roles in the survival and self-renewal of early hematopoietic progenitors and monocytic precursors (particularly Flt3) and in early lymphoid development; Flt3 signaling is critical in the development of dendritic cells from hematopoietic cells.16,18,19

Both Kit and Flt3 are proto-oncogenes, and when constitutively activated, they can cause malignant transformation. Indeed, expression of constitutively active Kit or Flt3 in murine hematopoietic progenitors results in the development of hematologic malignancies.20,21 In naturally occurring cancers, mutations in Kit and Flt3 are detected in a variety of different neoplastic disorders. Deletions in the juxtamembrane domain of Kit leading to receptor activation are detected in 30% to 60% of gastrointestinal stromal tumors in humans.20,21 Aggressive mastocytosis in humans is often associated with the presence of an activating point mutation in the kinase domain (catalytic domain) of Kit.22,23 In contrast, approximately 30% to 50% of high-grade canine mast cell tumors possess internal tandem duplications in the juxtamembrane domain, which also result in dysregulation of Kit function.24–27 Similar juxtamembrane domain duplications and catalytic domain point mutations have been identified in Flt3 from humans with acute myeloid leukemia.28,29 Lastly, both Kit and Flt3 are aberrantly expressed in several human neoplasms, including lung carcinoma, glioblastoma, ovarian tumors, leukemias, and lymphomas.30–34

Another RTK intimately associated with neoplasia is Met, which is normally expressed on a wide variety of epithelial cells; its ligand, HGF, is primarily produced by stromal cells.35,36 Interactions of Met and HGF promote an array of cellular responses, such as proliferation, scattering (motility), and branching morphogenesis, which are all important in normal processes such as wound healing, angiogenesis, and cell invasion and differentiation.35–41 In several human carcinomas, Met is aberrantly expressed or mutated and some of these mutations are known to be carried in the germline.42–47 Results of recent studies48,49 have indicated that mice carrying mutations in the Met kinase domain develop histiocytic sarcoma as well as other forms of neoplasia, suggesting a relationship between Met dysfunction and malignant transformation of dendritic cells. Moreover, aberrant Met expression has been described in cancers in dogs, indicating that it is likely that dysfunction of this pathway also contributes to cancer development in this species.50

Given the fact that Kit and Flt3 are important for normal histiocytic differentiation and that dysregulation of Kit, Flt3, or Met can lead to the development of neoplastic disease, it was logical to suspect that these genes may be mutated in canine histiocytic sarcoma. Therefore, the purpose of the study reported here was to evaluate canine histiocytic sarcoma cell lines and tumor samples for dysregulation in the Kit/SCF, Flt3/Flt3L, and Met/HGF signaling pathways in an effort to determine whether inhibitors of these RTKs would be useful for the treatment of this disease.

Materials and Methods

Reagents and cell lines—Cell lines (provided by one of the authors)b used in this study included 030210,c DH82,d and Nikee (all of which are canine histiocytic sarcoma cell lines) and ML3 (a myeloid leukemia cell line). These cell lines were established and characterized previously in the laboratory of one of the authors (PFM). A C57 mouse mast cell linef and C2 canine mast cell lineg were also used. The Madin-Darby canine kidney cell line and the mouse P815 mastocytoma line were purchased from American Type Culture Collection. All cell lines were maintained in RPMI 1640 medium supplemented with 10% fetal bovine serum, nonessential amino acids, sodium pyruvate, HEPES, penicillin, streptomycin, and L-glutamine.h A new histiocytic canine cell line (Taz) was established in our laboratory from a tumor histiocytic sarcoma resected from a dog. All of the histiocytic cell lines were confirmed to be of histiocytic origin by use of immunohistochemical staining (to identify CD1+, CD11c+, CD11d, MHC II+, and ICAM-1+). The splitkinase inhibitor SU11654i and Met inhibitor PHA665752i were also used.50,51 Fresh stock solutions of inhibitor (10mM) were prepared before each experiment by dissolving approximately 4 mg of inhibitor in 1 mL of dimethyl sulfoxide; this stock solution was then used for further dilution in dimethyl sulfoxide. For flow cytometry, the Ack45 anti-Kit monoclonal antibody conjugated to phycoerythrinj was used at a dilution of 1:200.

Histiocytic sarcoma tissue samples—Histiocytic sarcoma tissue samples were obtained from a large bank of previously characterized tumors.b Full records of each dog's history were reviewed for relevance to the screening. All of the tissues were formalin fixed and embedded in paraffin at the Veterinary Teaching Hospital of the University of California, Davis. Forty-one samples were available for use; 36 yielded suitable gDNA. Dog breeds represented included Bernese Mountain Dogs (n = 22), Rottweilers (4), Golden Retrievers (3), Flat-Coated Retrievers (3), Poodle (1), and Basset Hound (1); breed was unknown for 2 dogs. Within this population, there were 7 sexually intact males, 7 neutered males, 7 sexually intact females, and 14 neutered females; sex was unknown for 1 dog. The median age was 7 years (age range, 3 to 12 years).

Cloning of canine Flt3—Canine Flt3 cDNA was cloned from the canine myeloid leukemia cell line ML3. Starting primers were placed in regions of high homology between human and mouse sequences. The resultant products were sequenced and then used as templates for further primer design (Appendix). Once the full sequence was obtained, the full-length product was generated and cloned into the pGEM-T vectork and directly sequenced.

RT-PCR assays for receptor tyrosine kinases and ligands—Cell lines were evaluated for expression of the Kit/SCF, Flt3/Flt3L, and Met/HGF by use of the following primer pairs: SCF (sense 5′-GAGCATATGAAAGGGA TCTGCGGGAA-3′ and antisense 5′-CACCCGCGGTTATGCAACAGGGGGTAACATA AA-3′); Kit (sense 5′-CCCATGTATGAAGTACAGTGGAAG-3′ and antisense 5′-CTGATTAAGTCGGATGCGGCCATG-3′); Flt3L (sense 5′-GTGCTGGCGCCAGCCT GGACCCAA-3′ and antisense 5′-CCTGCAGGTGGAGGCGACAGTGAC-3′); Flt3 (sense 5′CCAGTCGGGCGTTGGCGCGCCGCGGCGGCCGGCT-3′ and antisense 5′-TCAGGG AACTGTGTTTAAAGACCCAGAG-3′); HGF (sense 5′-GGTGCTATACTCTTGAC CCTGC-3′ and antisense 5′-CACCAGGGTGACTCAGCCCCATCTG-3′); Met (sense 5′-GGACTTTTCCTGTGGCTGAAAAGG-3′ and antisense 5′-GGTAGTCTACAGATTCAT TTG-3′); and GAPDH (sense 5′GCTGCTTTTAACTCTGGCAAAG-3′ and antisense 5′-GAATCATACTGGAACATGTACAG-3′). All primer pairs were designed to span at least 1 intron, thereby making identification of gDNA contamination in the reaction assay easy to identify. For RT-PCR assay of cell lines to detect mRNA for Met, HGF, and HGFA, total RNA was extracted from 20 × 106 cells by use of standard phenol and guanidine isothiocyanate purificationl and cDNA was synthesized according to the manufacturer's specifications. Polymerase chain reaction analysis was performed for 40 cycles at 94°C for 30 seconds, 60°C for 30 seconds, and 72°C for 90 seconds. For all PCR analyses, GAPDH was used as a positive control.

Strand analysis of RTKs—Isolation of the gDNA from the paraffin-embedded tissue blocks was performed following an established protocol.27 In brief, two 25-μm slices of the block were placed in DNase-free tubes. Two washes with xylene and 100% ethanol were completed, and the pellets were air-dried. Digestion was undertaken with proteinase K bufferm for approximately 12 hours at 55°C. The gDNA was isolated by use of a standard isolation column according to the manufacturer's specificationsn with 150 µL of nuclease-free water.h The resultant DNA samples were quantitated via spectrophotometry. The primers used for PCR amplification of gDNA for fluorescent PAGE were as follows: Kit exon 11 (sense 5′-CCCATGTAT-GAAGTACAGTGGAAG-3′ and antisense 5′-GTTCCC-TAAAGTCATTGTT ACACG-3′); Kit exon 17 (sense 5′-TGTATC ACAGAGACTTGGCTG-3′ and antisense 5′-GTTTC-CTTTGACCACAT-3′); Flt3 exon 11 (sense 5′GCAATTTAG-GTATG AAAGCCAG-3′ and antisense 5′-GAAACA TTGGGAC-CATTCGAGTC-3′); and Flt3 exon 17 (sense 5′-GAGACCTG-GCAGCCAGGAATGTGC-3′ and antisense 5′-GCCATCCACTT TACAGGCAGAC-3′). To generate PCR products that could be visualized via fluorescent detection, the forward primers were labeled with 6-carboxyfluorescein.h Approximately 20 μL of the PCR product was separated via electrophoresis on a 4% agarose gel, stained with ethidium bromide, and examined via UV light. For PAGE, 0.1 μL of PCR product was denatured for 2 minutes at 95°C in 2 μL of loading buffer (1 mL of deionized formamide, 100 μL of carboxy-X-rhodamine-labeled size standard,o and 200 µL of loading dye). This product underwent electrophoresis for 2 hours on a standard DNA sequencero with a 6% polyacrylamide gel. The resultant fluorescent output was used by a specific nucleic acid analysis software packagep to determine precise fragment lengths.52

Met mutation analysis—Primers used for PCR amplification of gDNA from histiocytic sarcoma cell lines and tumor samples to analyze the Met kinase domain were as follows: Exon 16 (sense 5′-GAAGTTAATGTCTCTCCTGCTGG-3′ and antisense 5′-CTATGTTCTCACCAGGATA AG-3′); Exon 17 (sense 5′-CTGCTGTTATCAACAAGGCAC-3′ and antisense 5′-GTCACT TAATTTGGACTGTGGC-3′); Exon 18 (sense 5′-GATCAGTAGAGGCCAGA TGAAG-3′ and antisense 5′-CTCATCACCTTTCATGTTCATACG-3′); and Exon 19/20 (sense 5′-CACAGTCCTCACCTGGATTC-3′ and antisense 5′-TCATGTGT CCCCCTCGCCATCAA TGTTATCTTGTG-3′). The PCR products were evaluated via agarose gel electrophoresis and then directly sequenced at the University of California-Davis Sequence Facility.

Cell proliferation assay—To evaluate the effect of Kit and Flt3 inhibition, cells were plated at 1 to 3 × 104 cells in 100 μL in 96-well plates; differences in cell number were a result of established differences in growth rates of the various cell lines. The cells were left untreated, or SU11654 was added to the wells at a final concentration of 0.01μM (sub-therapeutic dose) or 0.1μM (therapeutic dose). After 72 hours, plates were collected and the WST-1 (tetrazolium salt that is cleaved to formazan dye by metabolically active cells) assayq was performed (according to the manufacturer's specifications) to determine the relative cell number in each well. Absorbency readings were performed on an ELISA plate readerr after 5 minutes of incubation. To evaluate the effect of Met inhibition, cells were plated in the aforementioned manner and then left untreated, cultured with rhHGF (50 ng/mL), or cultured with rhHGF and PHA665752 (0.1μM) together. After 72 hours, plates were collected and the WST-1 assay was performed according to the manufacturer's specifications to determine the relative cell number in each well. Absorbency readings were performed on an ELISA plate reader after 5 minutes of incubation. These experiments were performed in triplicate. The P815 and MDCK cell lines were used as positive control samples for these experiments.

Flow cytometry—Cell lines were counted and then resuspended in 24-well plates at a concentration of 0.25 to 0.5 × 106 cells/well (depending on growth rates) in complete medium. Cells were left untreated, or SU11654 was added to a final concentration of 0.01 to 5μM. Cells were collected after 72 hours of culture, washed twice with 0.1% glucose in PBS solution, fixed in 1 mL of cold 70% ethanol, and then stored at 4°C. Prior to flow cytometric analysis, the cells were centrifuged and 0.5 mL of propidium iodide staining solution (50 μg/mL) and RNAse in 0.1% glucose-PBS solution (10 μg/mL) were added. A flow cytometers was used, and at least 10,000 counts were obtained for each sample. Flow cytometry analysis softwaret was used for analysis, which was performed by gating on live cells and evaluating histograms by use of a linear scale. These experiments were performed in triplicate. The P815 and C2 cell lines were used as positive control samples for these experiments.

Results

Cloning of canine Flt3—To begin to assess the roles of Kit/SCF, Flt3/Flt3L, and Met/HGF signal transduction in histiocytic sarcomas in dogs, it was first necessary to determine whether these genes are expressed in the relevant cell lines. Although sequences for canine Kit, SCF, Flt3L, Met, and HGF were available in GenBank (AF099030, S53329, AF155148, AY543631, and AY543632, respectively), canine Flt3 had not yet been cloned and the canine genome was not yet available. Therefore, we cloned the gene from the canine acute myeloid leukemia cell line ML3, initially using primers derived from regions of homology between the mouse and human sequences to generate PCR products, and then using primers designed from theses PCR products to complete the entire sequencing. Canine Flt3 is composed of 994 amino acids (GenBank No. DQ007998) and is located on canine chromosome 25 (CanFam 1.0). It has 90% amino acid homology with human Flt3 and 80% amino acid homology with mouse Flt3. As expected, critical domains of Flt3, such as the juxtamembrane and kinase domains, have 100% homology with both species.

Expression of Kit/SCF, Flt3/Flt3L, and Met/HGF in canine histiocytic sarcoma cell lines—Because of the possible role of Kit, Flt3, and Met dysregulation in the malignant transformation of dendritic cells, canine histiocytic sarcoma tumor cell lines were assessed for expression of these RTKs and their respective ligands. All cell lines expressed mRNA for Kit, Flt3, Met, SCF, and Flt3L (Figure 1). Additionally, all lines except the 0302 line also expressed mRNA for HGF. Incidentally, this was also the only cell line that expressed low levels of mRNA for Met. Antibodies were available for identification of Kit via flow cytometry, and protein could be readily detected on the cell surface of all histiocytic sarcoma lines (Figure 2). Interestingly, the cell lines had variable levels of Kit expression; the functional consequences of these differences are not currently known. Because of a lack of reagents with which to identify the other proteins via flow cytometry, protein expression of Flt or Met was not assessed. Nevertheless, data from the canine histiocytic tumor cell lines investigated indicate that Kit/SCF, Flt3/Flt3L, and Met/HGF pathways are present in histiocytic sarcomas in dogs.

Figure 1—
Figure 1—

Expression of mRNA for receptor tyrosine kinases Kit, Flt3, and Met and SCF, Flt3L, and HGF in canine histiocytic sarcoma cell lines. The RNA was isolated from the cell lines, cDNA was generated from the RNA, and PCR procedures were performed with primers specific for Kit, SCF, Flt3, Flt3L, Met, and HGF; a PCR procedure for GAPDH was used as a positive control for cDNA quality. Cell lines evaluated were as follows: lane 1 = Taz (a histiocytic canine cell line established in the authors' laboratory from a tumor histiocytic sarcoma resected from a dog); lane 2 = 030210; lane 3 = DH82; lane 4 = Nike; and lane 5 = the positive control sample (ie, the C2 line for Kit and SCF and the ML3 line for all other assessments). The larger-sized Kit PCR product produced by the C2 positive control sample is a result of the presence of an internal tandem duplication in Kit in this cell line.

Citation: American Journal of Veterinary Research 67, 4; 10.2460/ajvr.67.4.633

Figure 2—
Figure 2—

Cell surface expression of Kit in canine histiocytic sarcoma cell lines 030210, Nike, and DH82. Cells were collected, washed, and then incubated with a phycoerythrin labeled anti-Kit antibody (Ack 45) or control isotype antibody. Flow cytometry was performed to assess levels of Kit surface expression. The C57 cell line served as a positive control specimen for these experiments.

Citation: American Journal of Veterinary Research 67, 4; 10.2460/ajvr.67.4.633

Analysis of cell lines and tumor samples for Kit, Flt3, and Met mutation—To begin to assess the potential role of Kit, Flt3, or Met dysregulation in canine histiocytic sarcoma, we evaluated these genes for evidence of mutation in regions known to frequently undergo mutation in human and canine neoplasms. For Kit and Flt3, a previously described technique termed strand analysis was used to evaluate these genes for evidence of duplications or deletions in exons 11 and 17. Briefly, this analysis involves PAGE of fluorescently labeled PCR products that can detect insertions or deletions of 1 to 2 or more base pairs and provides rapid screening of several samples simultaneously. We have recently used this technique to screen canine mast cell tumors for Kit internal tandem duplications in exon 11 and have found it to be reliable, accurate, and efficient.52 None of the tumor samples or cell lines evaluated had evidence of insertions or deletions in exon 11 or exon 17 of either gene. Although it is possible that there are point mutations in the juxtamembrane and catalytic domains that were not detected by this screening process, dysregulation of Kit or Flt3 through deletions and insertions in these regions is extremely unlikely to occur in canine histiocytic sarcoma.

As the known mutations in Met occur over the entire kinase domain, we elected to directly sequence most of this domain including exons 16 through 19. To facilitate this process, primers were designed on the basis of the corresponding introns because gDNA was used and the intervening introns were prohibitively long for PCR procedures. The PCR products for exons 18 through 20 were directly sequenced from the 4 histiocytic sarcoma lines and 34 representative histiocytic sarcoma tumor samples because most mutations in Met are known to occur in these coding regions. No evidence of mutation was found in any of the exons analyzed, although a polymorphism at nucleotide 3711 (C to T, amino acid 1237) was identified. However, this did not change the amino acid sequence. The PCR products for exons 16 to 17 were also directly sequenced from the 4 histiocytic sarcoma lines and 23 representative histiocytic sarcoma tumor samples because rare mutations are found in these coding regions. Again, no mutations were identified. Therefore, our data suggest that mutations in kinase domain of Met are unlikely to occur in canine histiocytic sarcoma.

Inhibition of Kit, Flt3, and Met signal transduction in histiocytic sarcoma cell lines—Although there was no obvious evidence of mutations in Kit, Flt3, and Met, it was still possible that these signal transduction pathways were active secondary to autocrine stimulation through coexpression of ligand or through point mutations that were not detected in the initial screenings. To test this possibility, the histiocytic tumor cell lines were treated with 2 inhibitors of these pathways. First, we used the previously described multitargeted kinase inhibitor SU11654 that blocks phosphorylation of several split-kinase RTKs, including Kit, Flt3, PDGFR, and VEGFR. It is known that SU11654 inhibits proliferation of murine, canine, and human tumor cell lines that carry mutations in Kit and Flt3.53–55 Additionally, many of those lines underwent apoptosis after 2 to 3 days of culture in the presence of SU11654. In the present study, there was no evidence of apoptosis in any of the histiocytic sarcoma lines evaluated, even when doses far above the known relevant concentrations were used (Figure 3). We then investigated whether SU11654 was capable of slowing or inhibiting cell proliferation. There was no effect on cell numbers after 72 hours of culture in the presence of SU11654 (Figure 4). These experiments suggest that Kit/SCF and Flt3/Flt3L signal transduction are not essential for survival and proliferation of histiocytic sarcoma. Moreover, because both PDGFR and VEGFR signal transduction were also blocked by SU11654, it is unlikely that these pathways are critical for histiocytic sarcoma cells.

Figure 3—
Figure 3—

Effects of inhibition of Kit and Flt3 signal transduction on apoptosis in canine histiocytic sarcoma cell lines 030210, Nike, and DH82 and the murine mast cell line P815 (positive control sample). Cell lines were plated in 24-well plates then left untreated (thin line) or incubated with the split-kinase inhibitor SU11654 (thick line) at 0.01μM (P815) or 1μM (histiocytic sarcoma lines). The plates were then collected after 72 hours of culture and analyzed by use of propidium iodide staining to assess extent of apoptosis (M1 region). After treatment, apoptosis was detected only in the P815 cell line.

Citation: American Journal of Veterinary Research 67, 4; 10.2460/ajvr.67.4.633

Figure 4—
Figure 4—

Effect of blocking Kit, Flt3, or Met signal transduction on cell proliferation in histiocytic sarcoma cell lines. A—Cell lines 030210, Nike, and DH82 and the canine mast cell line C2 (positive control sample) were plated in 96-well plates and then left untreated or incubated with SU11654 at 0.01μM (white bars) or 0.1μM (black bars). After 72 hours of culture, plates were analyzed by use of the WST-1 colorimetric assay to assess cell numbers; cell proliferation was calculated as a percentage of the control wells (fluorescence of sample/fluorescence of untreated cells). The horizontal dotted line indicates 100% of control cells. B—CPHA665752 at 0.1μM (black bars). After 72 hours of culture, plates were analyzed by use of the WST-1 a colorimetric assay to assess cell numbers; cell proliferation was calculated as a percentage of the control wells (fluorescence of sample/fluorescence of untreated cells). The horizontal dotted line indicates 100% of control cells.

Citation: American Journal of Veterinary Research 67, 4; 10.2460/ajvr.67.4.633

To evaluate the ability of Met inhibition to affect cell proliferation, the Met inhibitor PHA665752 was used in a similar assay.50 In this setting, HGF was added to the tissue culture medium to promote Met signal transduction and potentially induce cycling in the tumor cells. There was no evidence that HGF had a biological effect on the histiocytic sarcoma cells, and PHA665752 did not inhibit cell proliferation (Figure 4). Therefore, our data suggest that Met/HGF signaling is unlikely to play a role in the survival and proliferation of histiocytic sarcoma.

Discussion

Receptor tyrosine kinase dysregulation is now known to play an important role in a variety of human cancers and has recently been identified in naturally occurring tumors in dogs and cats. Identification and characterization of kinase dysregulation are important because these abnormal pathways can be targeted for therapeutic intervention. Indeed, this approach has already proven successful for the treatment of chronic myelogenous leukemias and primary lung cancer in humans by use of drugs such as imatinib mesylate and gefitinib, as well as for treatment of mast cell tumors in dogs via administration of SU11654.56 However, application of such targeted agents requires careful molecular characterization of particular tumors because simple expression of a specific kinase does not translate into dysfunction of that kinase. This was proven to be the case in human primary lung cancer; nearly all affected individuals overexpressed EGFR, but only those persons whose tumors carried EGFR mutations responded to treatment.57

The purpose of the present study was to evaluate kinase dysregulation in canine histiocytic sarcoma cell lines because this type of tumor is extremely aggressive and there is currently no effective treatment, resulting in death of all affected dogs. Identification of mutated kinases may therefore provide an opportunity for the development of novel agents to treat this disease. For several reasons, we chose to examine 3 particular pathways: Kit/SCF, Flt3/Flt3L, and Met/HGF. Both Kit and Flt3 signaling are important in normal dendritic cell maturation, promoting both survival and cell proliferation and differentiation.16,19 Therefore, it was reasonable to hypothesize that inappropriate function of either of these pathways may contribute to the malignant transformation of dendritic cells. Furthermore, mutation of Kit and Flt3 that results in uncontrolled signaling is known to occur in several neoplasms, including gastrointestinal stromal tumors, acute myelogenous leukemia, and mast cell tumors, and small molecule inhibitors designed to block these pathways have been shown to provide a therapeutic benefit.56,58,59 Lastly, because Kit and Flt3 inhibitors are currently being investigated in clinical trials in humans and dogs, such agents would be readily available for clinical application.

Aberrant Met signaling has been identified in a variety of human cancers, and overexpression of this RTK is often associated with a poor long-term prognosis.42,48 More importantly, mice expressing Met that has particular mutations in the kinase domain develop histiocytic sarcoma as well as other tumors such as lymphoma and hemangiosarcoma.49 Our group has previously evaluated the effects of a novel Met inhibitor and shown that this inhibitor was capable of blocking HGF-induced cell cycling as well as migration and scattering of tumor cells in vitro.50 Several small molecule inhibitors of Met are presently being investigated in clinical trials, thereby providing a potential opportunity for future use in dogs.

To begin evaluating these pathways in histiocytic sarcomas, it was first necessary to develop appropriate reagents. Our group had previously cloned canine Kit, Met, and HGF, and sequences for canine SCF and Flt3L were present in GenBank. We therefore cloned canine Flt3 using a canine myelogenous leukemia cell line as a template. As expected, canine Flt3 had high homology with both human and mouse proteins (90% and 80%, respectively). The sequence was then used to search the canine genome database, and Flt3 was located on canine chromosome 25 (CanFam1.0). These canine sequences were used to generate appropriate PCR primers, and 4 histiocytic sarcoma tumor cell lines were screened for expression of the RTKs and their ligands. All tumor cell lines expressed mRNA for Kit/SCF, Flt3/Flt3L, and Met, and only 1 cell line did not express mRNA for HGF. Protein expression of Kit was confirmed via flow cytometry; appropriate reagents for assessment of canine Flt3 and Met expression are not currently available.

In the present study, histiocytic sarcoma cell lines as well as tumors were evaluated for evidence of mutation in Kit, Flt3, and Met. For the first 2 RTKs, we focused on regions of the gene known to commonly possess activating mutations: the juxtamembrane domain and the catalytic domain. With respect to the juxtamembrane domain, these mutations usually consist of deletions or insertions, whereas point mutations most commonly occur in the catalytic domain and involve codon 814. To facilitate screening of the cell lines and tumor specimens, a previously described technique involving fluorescent PAGE that is capable of identifying PCR products that differ in size by as little as 1 to 2 bp was used. As previously discussed, none of the samples analyzed had evidence of insertions or deletions in exon 11 (encoding the juxtamembrane domain) of Kit or Flt 3. Moreover, insertions or deletions in exon 18 (catalytic domain) of either RTK were not detected. Although it was possible that an undetected point mutation in this region may have resulted in constitutive activation, the lack of response to SU11654 even at extremely high doses indicated that such mutations are not likely to be driving cell survival and proliferation in the histiocytic sarcoma cells. Given that SU11654 also effectively inhibited VEGFR and PDGFR signal transduction, it is probable that dysfunction of these pathways is also absent in histiocytic sarcoma. Overall, these data suggest that the split-kinase RTK family is unlikely to be involved in malignant transformation of dendritic cells and, thus, does not serve as a relevant target for therapeutic intervention.

As previously stated, Met mutation is known to result in the development of histiocytic sarcoma in mouse models. Given the potential link between this RTK and malignant transformation of dendritic cells, we decided to evaluate canine histiocytic sarcoma cells for evidence of Met dysregulation. The expression of mRNA for Met was detected in the histiocytic sarcoma cell lines; 3 of the 4 lines also expressed mRNA for HGF. The cell lines and tumor specimens were also evaluated for mutation in Met. Unlike Kit and Flt3, mutations in Met occur primarily in the kinase domain; the mutations are not localized to 1 particular codon but occur throughout this domain spanning exons 16 to 20. It was therefore necessary to develop primers that would enable direct sequencing of each exon. Given the enormity of this task, we elected to sequence exons 16 to 20 from all of the cell lines and only 20 of the tumors, as this would likely provide a representative sample from which to determine whether mutations occur with any regular frequency. No mutations were identified in the kinase domain of Met in any of the samples evaluated. The absence of dysfunction was further supported by a lack of response to inhibition of Met signaling with PHA66752. Therefore, as with Kit and Flt3, dysregulation of Met is considered unlikely to play an important role in malignant transformation of canine dendritic cells.

Although the present study did not reveal any evidence of Kit, Flt3, or Met dysregulation in canine histiocytic sarcoma cells, it did provide important information with respect to the application of targeted agents to treat this disease. As endeavors in human oncology have revealed, a comprehensive understanding of the molecular biology of individual cancers is necessary for the successful application of targeted inhibitors. This was clearly shown to be the case for human lung cancer, in which most tumors overexpressed EGFR, but only 10% to 20% responded to EGFR inhibition as a result of mutations in EGFR that led to inappropriate signal transduction.57 Our data suggest that dysregulation of Kit, Flt3, and Met is unlikely to play an important role in canine histiocytic sarcomas; therefore, treatments intended to block these pathways will probably yield little therapeutic benefit in affected dogs. As such, the effective use of targeted agents to treat histiocytic sarcoma in dogs will likely require a more detailed characterization of the molecular biology of this tumor type.

ABBREVIATIONS

RTK

Receptor tyrosine kinase

SCF

Stem-cell factor

Flt3L

Flt3 ligand

HGF

Hepatocyte growth factor

HGFA

HGF activator

rhHGF

Recombinant human hepatocyte growth factor

VEGFR

Vascular endothelial growth factor receptor

PDGFR

Platelet-derived growth factor receptor

EGFR

Epithelial growth factor receptor

RT-PCR

Reverse transcriptase-PCR

gDNA

Genomic DNA

GAPDH

Glyeraldehyde-3-phosphate dehydrogenase

a.

Skorupski KA, Clifford CA, Paoloni MC, et al. CCNU for the treatment of dogs with metastatic or disseminated histiocytic sarcoma (abstr), in Proceedings. Conf Vet Cancer Soc 2003;23:36.

b.

Provided by Dr. Peter Moore, Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, Calif.

c.

Originally from Dr. D. Gephardt, Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC.

d.

Originally from Dr. M. Wellman, Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio.

e.

Originally from Dr. B. Kitchell, Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Mich.

f.

Provided by Dr. Stephen Galli, Department of Pathology, School of Medicine, Stanford University, Stanford, Calif.

g.

Provided by Dr. Warren Gold, Cardiovascular Research Institute, University of California, San Francisco, Calif.

h.

Invitrogen, Carlsbad, Calif.

i.

Pfizer Inc, La Jolla, Calif.

j.

BD Biosciences, San Diego, Calif.

k.

Promega, Madison, Wis.

l.

TRIzol reagent, Invitrogen, Carlsbad, Calif.

m.

QIAGEN, Valencia, Calif.

n.

DNeasy column, QIAGEN, Valencia, Calif.

o.

Applied Biosystems, Foster City, Calif.

p.

STRand analysis software package. Available at: www.vgl.ucdavis.edu/STRand.

q.

Roche, Alameda, Calif.

r.

Molecular Devices, Sunnyvale, Calif.

s.

FACScan instrument, Becton-Dickinson, Franklin Lakes, NJ.

t.

CellQuest, Becton-Dickinson, Franklin Lakes, NJ.

References

  • 1

    Affolter VK, Moore PF. Localized and disseminated histiocytic sarcoma of dendritic cell origin in dogs. Vet Pathol 2002; 39: 7483.

  • 2

    Hayden DW, Waters DJ, Burke BA, et al. Disseminated malignant histiocytosis in a Golden Retriever: clinicopathologic, ultrastructural, and immunohistochemical findings. Vet Pathol 1993; 30: 256264.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 3

    Moore PF, Rosin A. Malignant histiocytosis of Bernese Mountain Dogs. Vet Pathol 1986; 23: 110.

  • 4

    Padgett GA, Madewell BR, Keller ET, et al. Inheritance of histiocytosis in Bernese Mountain Dogs. J Small Anim Pract 1995; 36: 9398.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 5

    Ramsey IK, McKay JS, Rudorf H, et al. Malignant histiocytosis in three Bernese Mountain Dogs. Vet Rec 1996; 138: 440444.

  • 6

    Rosin A, Moore P, Dubielzig R. Malignant histiocytosis in Bernese Mountain Dogs. J Am Vet Med Assoc 1986; 188: 10411045.

  • 7

    Schmidt ML, Rutteman GR, van Niel MH, et al. Clinical and radiographic manifestations of canine malignant histiocytosis. Vet Q 1993; 15: 117120.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 8

    Wellman ML, Davenport DJ, Morton D, et al. Malignant histiocytosis in four dogs. J Am Vet Med Assoc 1985; 187: 919921.

  • 9

    Favara BE, Feller AC, Pauli M, et al. Contemporary classification of histiocytic disorders. The WHO Committee On Histiocytic/Reticulum Cell Proliferations. Reclassification Working Group of the Histiocyte Society. Med Pediatr Oncol 1997; 29: 157166.

    • Search Google Scholar
    • Export Citation
  • 10

    Gogusev J, Nezelof C. Malignant histiocytosis. Histologic, cytochemical, chromosomal, and molecular data with a nosologic discussion. Hematol Oncol Clin North Am 1998; 12: 445463.

    • Search Google Scholar
    • Export Citation
  • 11

    Schmidt D. Malignant histiocytosis. Curr Opin Hematol 2001; 8: 14.

  • 12

    Rosnet O, Birnbaum D. Hematopoietic receptors of class III receptor-type tyrosine kinases. Crit Rev Oncog 1993; 4: 595613.

  • 13

    Rosnet O, Schiff C, Pebusque MJ, et al. Human FLT3/FLK2 gene: cDNA cloning and expression in hematopoietic cells. Blood 1993; 82: 11101119.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 14

    Ullrich A, Schlessinger J. Signal transduction by receptors with tyrosine kinase activity. Cell 1990; 61: 203.

  • 15

    Yarden Y, Ullrich A. Growth factor receptor tyrosine kinases. Annu Rev Biochem 1988; 57: 443478.

  • 16

    Lyman SD, Jacobsen SE. c-kit ligand and Flt3 ligand: stem/progenitor cell factors with overlapping yet distinct activities. Blood 1998; 91: 11011134.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 17

    Lyman SD. Biology of flt3 ligand and receptor. Int J Hematol 1995; 62: 6373.

  • 18

    Galli SJ, Zsebo KM, Geissler EN. The kit ligand, stem cell factor. Adv Immunol 1994; 55: 195.

  • 19

    McKenna HJ. Role of hematopoietic growth factors/flt3 lig- and in expansion and regulation of dendritic cells. Curr Opin Hematol 2001; 8: 149154.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 20

    Hawley TS, Fong AZ, Griesser H, et al. Leukemic predisposition of mice transplanted with gene-modified hematopoietic precursors expressing flt3 ligand. Blood 1998; 92: 20032011.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 21

    Kitayama H, Tsujimura T, Matsumura I, et al. Neoplastic transformation of normal hematopoietic cells by constitutively activating mutations of c-kit receptor tyrosine kinase. Blood 1996; 88: 9951004.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 22

    Nagata H, Okada T, Worobec AS, et al. c-kit mutation in a population of patients with mastocytosis. Int Arch Allergy Immunol 1997; 113: 184186.

  • 23

    Longley BJ, Reguera MJ, Ma Y. Classes of c-KIT activating mutations: proposed mechanisms of action and implications for disease classification and therapy. Leuk Res 2001; 25: 571576.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 24

    London CA, Galli SJ, Yuuki T, et al. Spontaneous canine mast cell tumors express tandem duplications in the proto-oncogene c-kit. Exp Hematol 1999; 27: 689697.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 25

    Ma Y, Longley BJ, Wang X, et al. Clustering of activating mutations in c-KIT's juxtamembrane coding region of canine mast cell neoplasms. J Invest Dermatol 1999; 112: 165170.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 26

    Zemke D, Yamini B, Yuzbasiyan-Gurkan V. Mutations in the juxtamembrane domain of c-KIT are associated with higher grade mast cell tumors in dogs. Vet Pathol 2002; 39: 529535.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 27

    Downing S, Chien MB, Kass PH, et al. Prevalence and signficance of kit internal tandem duplications in canine mast cell tumors. Am J Vet Res 2002; 63: 17181723.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 28

    Iwai T, Yokota S, Nakao M, et al. Internal tandem duplication of the FLT3 gene and clinical evaluation in childhood acute myeloid leukemia. The Children's Cancer and Leukemia Study Group, Japan. Leukemia 1999; 13: 3843.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 29

    Nakoa M, Yokota S, Iwai T, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia 1996; 10: 19111918.

    • Search Google Scholar
    • Export Citation
  • 30

    Stanulla M, Welte K, Hadam MR, et al. Coexpression of stem cell factor and its receptor c-kit in human malignant glioma cell lines. Acta Neuropathol 1995; 89: 158165.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 31

    Hines SJ, Organ C, Kornstein MJ, et al. Coexpression of the c-kit and stem cell factor genes in breast carcinomas. Cell Growth Differ 1995; 6: 769779.

    • Search Google Scholar
    • Export Citation
  • 32

    Hibi K, Takahashi T, Sekido Y, et al. Co-expression of the stem cell factor and the c-kit genes in small cell lung cancer. Oncogene 1991; 6: 22912296.

    • Search Google Scholar
    • Export Citation
  • 33

    Carow CE, Levenstein M, Kaufmann SH, et al. Expression of the hematopoietic growth factor receptor FLT3 (STK-1/Flk2) in human leukemias. Blood 1996; 87: 10891096.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 34

    Drexler HG. Expression of FLT3 receptor and response to FLT3 ligand by leukemic cells. Leukemia 1996; 10: 588599.

  • 35

    Jiang W, Hiscox S, Matsumoto K, et al. Hepatocyte growth factor/scatter factor, its molecular, cellular and clinical implications in cancer. Crit Rev Oncol Hematol 1999; 29: 209248.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 36

    Maulik G, Shrikhande A, Kijima T, et al. Role of the hepatocyte growth factor receptor, c-Met, in oncogenesis and potential for therapeutic inhibition. Cytokine Growth Factor Rev 2002; 13: 4159.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 37

    Kan M, Zhang GH, Zarnegar R, et al. Hepatocyte growth factor/hepatopoietin A stimulates the growth of rat kidney proximal tubule epithelial cells (RPTE), rat nonparenchymal liver cells, human melanoma cells, mouse keratinocytes and stimulates anchor-age-independent growth of SV-40 transformed RPTE. Biochem Biophys Res Commun 1991; 174: 331337.

    • Search Google Scholar
    • Export Citation
  • 38

    Montesano R, Matsumoto K, Nakamura T, et al. Identification of a fibroblast-derived epithelial morphogen as hepatocyte growth factor. Cell 1991; 67: 901908.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 39

    Weidner KM, Behrens J, Vandekerckhove J, et al. Scatter factor: molecular characteristics and effect on the invasiveness of epithelial cells. J Cell Biol 1990; 111: 20972108.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 40

    Grant DS, Kleinman HK, Goldberg ID, et al. Scatter factor induces blood vessel formation in vivo. Proc Natl Acad Sci U S A 1993; 90: 19371941.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 41

    Birchmeier C, Gherardi E. Developmental roles of HGF/SF and its receptor, the c-Met tyrosine kinase. Trends Cell Biol 1998; 8: 404410.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 42

    Birchmeier C, Birchmeier W, Gherardi E, et al. Met, metastasis, motility and more. Nat Rev Mol Cell Biol 2003; 4: 915925.

  • 43

    Tsao MS, Liu N, Chen JR, et al. Differential expression of Met/hepatocyte growth factor receptor in subtypes of non-small cell lung cancers. Lung Cancer 1998; 20: 116.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 44

    Ruco LP, Ranalli T, Marzullo A, et al. Expression of Met protein in thyroid tumours. J Pathol 1996; 180: 266270.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 45

    Ruco LP, Stoppacciaro A, Ballarini F, et al. Met protein and hepatocyte growth factor (HGF) in papillary carcinoma of the thyroid: evidence for a pathogenetic role in tumourigenesis. J Pathol 2001; 194: 48.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 46

    Schmidt L, Duh FM, Chen F, et al. Germline and somatic mutations in the tyrosine kinase domain of the MET proto-oncogene in papillary renal carcinomas. Nat Genet 1997; 16: 6873.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 47

    Di Renzo MF, Olivero M, Martone T, et al. Somatic mutations of the MET oncogene are selected during metastatic spread of human HNSC carcinomas. Oncogene 2000; 19: 15471555.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 48

    Graveel CR, London CA, Vande Woude GF. A mouse model of activating Met mutations. Cell Cycle 2005; 4: 518520.

  • 49

    Graveel C, Su Y, Koeman J, et al. Activating Met mutations produce unique tumor profiles in mice with selective duplication of the mutant allele. Proc Natl Acad Sci U S A 2004; 101: 1719817203.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 50

    Liao AT, McMahon M, London CA. Characterization, expression, and function of c-Met in canine spontaneous cancers. Vet Comp Pathol 2005; in press.

    • Search Google Scholar
    • Export Citation
  • 51

    Liao AT, Chien MB, Shenoy N, et al. Inhibition of constitutively active forms of mutant Kit by multi-targeted indolinone kinase inhibitors. Blood 2002; 100: 585593.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 52

    Jones CL, Grahn RA, Chien MB, et al. Detection of c-kit mutations in canine mast cell tumors using fluorescent polyacrylamide gel electrophoresis. J Vet Diagn Invest 2004; 16: 95100.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 53

    London CA, Hannah AL, Zadovoskaya R, et al. Phase I dose-escalating study of SU11654, a small molecule receptor tyrosine kinase inhibitor, in dogs with spontaneous malignancies. Clin Cancer Res 2003; 9: 27552768.

    • Search Google Scholar
    • Export Citation
  • 54

    O'Farrell A-M, Abrams T, Yuen H, et al. SUGEN kinase inhibitors SU5416 and SU11248 inhibit Flt3 signaling: therapeutic application in AML. Proc Am Soc Hem 2001; 42: 497.

    • Search Google Scholar
    • Export Citation
  • 55

    O'Farrell AM, Abrams TJ, Yuen HA, et al. SU11248 is a novel FLT3 tyrosine kinase inhibitor with potent activity in vitro and in vivo. Blood 2003; 101: 35973605.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 56

    London CA. Kinase inhibitors in cancer therapy. Vet Comp Onc 2004; 2: 177193.

  • 57

    Lynch TJ, Bell DW, Sordella R, et al. Activating mutations in the epidermal growth factor receptor underlying responsiveness of non-small-cell lung cancer to gefitinib. N Engl J Med 2004; 350: 21292139.

    • Crossref
    • Search Google Scholar
    • Export Citation
  • 58

    Naoe T, Kiyoi H. Normal and oncogenic FLT3. Cell Mol Life Sci 2004; 61: 29322938.

  • 59

    Akin C, Metcalfe DD. The biology of Kit in disease and the application of pharmacogenetics. J Allergy Clin Immunol 2004; 114: 1319.

Appendix

Appendix

Primers for Flt3 cloning.

ForwardExon
CCA TGC GGG CGT TGG CGC GCC GCG GCG GCC GGC T1
CTG CCT GTG ATC AAG TGT GTT TTA ATC2
CCA GGC CCC TTC CCT TTC ATC CAA G12, 13
CAA ATT CCA GCA TGC CTG GTT CA17, 18
CTC TGG GTC TTT AAG CAC AGC TCC CTG3
TCT TGG ACC TGG AAG AAG TGT TC11
GCC TGA GAG CAC AGA GGC AGT GTA TGG3
GCT CCG GCG GCC ATG CGG GCG TTG GCG1
GCA ATC CTG CTG GGC TTT TGA CTC AAG G23
CCC AAC TGC ACG GAG GAA ATC AC11, 12
GCA ATT TAG GTA TGA AAG CCA G13, 14
GCC TGG TTC AAG AGA AGT TCA GAT AC18
GCA ATG CCC GTC TGC CTG TAA AGT GG20, 21
Reverse
GGA AGT CCT TCA GAC TAC ATT GTT TCT CTA GG24
CAG GGA GCT GTG CTT AAA GAC CCA GAG3
GAA CAC TTC TTC CAG GTC CAA GA11
TGA ACC AGG CAT GCT GGA ATT TG17, 18
TCT GCC AGC TGA CAT CCT AAA AA23
CTT GGA TGA AAG GGA AGG GGC CTG G12, 13
CTC AAA GTA GCT GCC CTC CTC CAG GG7, 8
CGG GGT CCA AGG CCT CCT CGC TAA CTT CTT TC23
CAG GCT TCA GGG CAC ACC CGA GGT CTT CTG3
CTG CTC GAT GTC CAC TGT CCA AAT AC12
GCA CCT GAT CCT AGT ACC TTC CC14, 15
CAG CAC ATT TAG GTC CTC CTC TTC CTC C19
GTT CCT CTA CGA ATC TTC GAC CTG AGC C24
All Time Past Year Past 30 Days
Abstract Views 73 0 0
Full Text Views 12379 12178 11538
PDF Downloads 229 100 7
Advertisement