Leukemia (2002) 16, 911–919 2002 Nature Publishing Group All rights reserved 0887-6924/02 $25.00 www.nature.com/leu B-CLL cells are capable of synthesis and secretion of both pro- and anti-angiogenic molecules NE Kay1, ND Bone1, RC Tschumper2, KH Howell2, SM Geyer3, GW Dewald4, CA Hanson4 and DF Jelinek2 1 Department of Medicine, Division of Hematology, Mayo Graduate and Medical Schools, Mayo Clinic, Rochester, MN, USA; 2Department of Immunology, Mayo Graduate and Medical Schools, Mayo Clinic, Rochester, MN, USA; 3Department of Biostatistics, Mayo Graduate and Medical Schools, Mayo Clinic, Rochester, MN, USA; and 4Department of Laboratory Medicine and Pathology, Mayo Graduate and Medical Schools, Mayo Clinic, Rochester, MN, USA Initial work has shown that clonal B cells from B-chronic lymphocytic leukemia (B-CLL) are able to synthesize pro-angiogenic molecules. In this study, our goal was to study the spectrum of angiogenic factors and receptors expressed in the CLL B cell. We used ELISA assays to determine the levels of basic fibroblast growth factors (bFGF), vascular endothelial growth factor (VEGF), endostatin, interferon-␣ (IFN-␣) and thrombospondin-1 (TSP-1) secreted into culture medium by purified CLL B cells. These data demonstrated that CLL B cells spontaneously secrete a variety of pro- and anti-angiogenic factors, including bFGF (23.9 pg/ml ± 7.9; mean ± s.e.m.), VEGF (12.5 pg/ml ± 2.3) and TSP-1 (1.9 ng/ml ± 0.3). Out of these three factors, CLL B cells consistently secreted bFGF and TSP-1, while VEGF was expressed in approximately two-thirds of CLL patients. Of interest, hypoxic conditions dramatically upregulated VEGF expression at both the mRNA and protein levels. We also employed ribonuclease protection assays to assay CLL B cell expression of a variety of other angiogenesis-related molecules. These analyses revealed that CLL B cells consistently express mRNA for VEGF receptor 1 (VEGFR1), thrombin receptor, endoglin, and angiopoietin. Further analysis of VEGFR expression by RT-PCR revealed that CLL B cells expressed both VEGFR1 mRNA and VEGFR2 mRNA. In summary, these data collectively indicate that CLL B cells express both proand anti-angiogenic molecules and several vascular factor receptors. Because of the co-expression of angiogenic molecules and receptors for some of these molecules, these data suggest that the biology of the leukemic cells may also be directly impacted by angiogenic factors as a result of autocrine pathways of stimulation. Leukemia (2002) 16, 911–919. DOI: 10.1038/sj/leu/2402467 Keywords: B-CLL; vascular factors; TSP-1; VEGF Introduction The process of angiogenesis in human tumors is now known to play a critical role in many facets of tumor development. The vascularization of solid tumors is linked to its subsequent ability to disseminate with resultant more aggressive disease.1–3 The vascular phase of most tumors is linked to the ability of a tumor cell to initiate and then become committed to the production of pro-angiogenic factors.4,5 This latter component is dominant to the simultaneous synthesis of negative regulators or anti-angiogenic factors by the same tumor. The shift in balance between production of anti-angiogenic factors and pro-angiogenic factors is a process that has been referred to as an angiogenic switch. It has been suggested that the angiogenic switch marks the onset of neovascularization which is probably a key step during tumor progression.4,5 The vast majority of the initial work in tumor-related angiogenesis has been conducted using solid tumor lines or models. More recently, abnormal bone marrow angiogenesis has also Correspondence: NE Kay, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA; Fax: 507-266-9277 Received 27 June 2001; accepted 16 January 2002 been observed in a number of diseases, including a variety of leukemias. Thus, patients with acute lymphocytic leukemia, acute myeloid leukemia, non-Hodgkin’s lymphoma, multiple myeloma, myelofibrosis and acute promyelocytic leukemia were shown to have increases in bone marrow microvessel density.6–13 Recently, we and others14–18 have defined a variety of abnormalities in the angiogenic status of patients with B-chronic lymphocytic leukemia (B-CLL). These abnormalities have included increased microvessel density in marrow14 and lymph nodes15 and increased levels of certain pro-angiogenic vascular growth factors including basic fibroblast growth factor (bFGF) and vascular endothelial cell growth factor (VEGF) in blood and urine of B-CLL patients.14,17,18 There is evidence that the tumor cells themselves may be an important source of these pro-angiogenic factors. Indeed, these vascular growth factors have been demonstrated in the cytoplasm of the CLL B cell16,17 and are present in the culture medium of CLL B cells (Kay, unpublished observations). Despite evidence that B-CLL cells express and release vascular proteins, the precise role that these factors play in the disease biology of B-CLL remains unclear. Thus, it remains possible that bFGF and VEGF may effect increased angiogenesis as well as have direct effects on the leukemic cells. Indeed, a recent publication has suggested that VEGF may play a role in an autocrine pathway for B-CLL cells.15 The goal of this study was to obtain a more complete characterization of the angiogenic status of the CLL B cell. In particular, we were interested in determining if the CLL B cell is capable of undergoing an angiogenic switch resulting in an increased production rate of pro-angiogenic factors. Recent insight into human angiogenesis mechanisms now permits a detailed analysis of both the pro- and anti-angiogenic factors as well as the array of vascular factor receptors expressed by a tumor cell. This report presents evidence that the CLL B cell clones from early stage and/or stable disease patients are capable of expressing and modulating their production of a wide variety of both pro- and anti-angiogenic molecules. This information adds to the possibility that CLL B cell clones can undergo an angiogenic switch. In addition, we analyzed the vascular factor data in relation to the presence of germline (GL) vs somatic mutation-type B cell clones (SM). The latter was done since recent evidence including our own has suggested that GL-type patients have a worse clinical outcome compared to SM-type patients.19–21 Methods B-CLL patients The cohorts of B-CLL patients studied were diagnosed using the NCI Criteria.22 B-CLL patients included in this study were either previously untreated patients (n = 30) and were in Rai B-CLL cells express pro- and anti-angiogenic molecules NE Kay et al 912 stage 0–1, or were treated patients who were in Rai stages 2– 4. Patients in the latter group were only studied in this protocol if distant from therapy by at least 3 weeks. Venous blood and urine samples were only obtained from patients providing informed consent. erated in a NuAire incubator (NuAire, Plymouth, MN, USA) with 2.0% O2, 5% CO2 and N2. Isolation of CLL B cells We used a ribonuclease protection assay (RPA) to measure mRNA levels of relevant angiogenic factors and their receptors. Total RNA was isolated from cells by the Trizol method (Life Technologies, Grand Island, NY, USA). RNase protection assays were done using the RiboQuant Multi-Probe RNase Protection Assay System from BD PharMingen (San Diego, CA, USA). The Human Angiogenesis template set was used, in conjunction with the In Vitro Transcription Kit (BD Pharmingen kit component), for the synthesis of the radiolabeled anti-sense RNA probes. The probes used in this RPA were able to detect mRNA for the vascular factor receptors Flt1 (VEGFR1), Flt4, Tie 1, Tie 2, and the thrombin receptor. In addition, this RPA detected mRNA coding for CD31 (PECAM1), endoglin, angiopoietin, VEGF, VEGF C, and the housekeeping gene L32. The RPA was performed according to the manufacturers’ recommended protocol. Peripheral blood mononuclear cells (PBMC) were separated from heparinized venous blood by density gradient centrifugation. To remove adherent cells, PBMC were suspended in RPMI 1640 supplemented with 10% fetal calf serum, and cells were incubated in plastic dishes at 37°C for 1 h prior to collection of non-adherent cells. In order to obtain at least 95% purity of CLL B cells, non-adherent cells were depleted of T cells by incubation with sheep erythrocytes. Following density gradient centrifugation, the rosette-negative cells were isolated from the gradient interface, and these cells typically consisted of ⬎97% CD19+/CD5+ cells. In some cases we used an alternative method for purification of CLL B cells. In brief, highly purified CD19+ B cells (⬎95%) were obtained from PBMCs by standard negative selection using a cocktail of subset-specific antibodies conjugated with magnetic beads (Miltenyi Biotech, Auburn, CA, USA). These purified CLL B cells were then used immediately for the laboratory studies described below or were cryopreserved in RPMI 1640, 20% fetal calf serum, and 10% DMSO and stored in liquid nitrogen until use. These cells were then analyzed for mRNA and/or protein expression of cytoplasmic or secreted vascular factors as described below. Detection of pro- and anti-angiogenic factors and vascular factor receptors in CLL patients Detection of pro- and anti-angiogenic factors in urine: Urine samples were collected using sterile techniques and stored at −70°C before analysis. Control urine samples were also obtained from age-matched individuals. We then measured urine levels of the pro-angiogenic factors, bFGF and VEGF as well as the anti-angiogenic factors TSP-1, endostatin, and IFN-␣. Basic FGF, VEGF, TSP-1, endostatin, and IFN-␣ levels were quantified using Quantikine solid phase enzymelinked immunoabsorbent assays (ELISA; R & D Systems, Minneapolis, MN, USA). The lower limits of detection by ELISA for these vascular factors were ⬍1–2 ng/ml (for TSP-1, interferon alpha, and endostatin) and 0.5–5 pg/ml (for bFGF and VEGF). The data are presented as mean ± standard error of the mean where replicates were done. Assessment of CLL B cell culture supernatants for the presence of pro- and anti-angiogenic factors: Purified CLL B cells were cultured in RPMI containing 10% FCS at 1 × 106 cells per ml for 24 h prior to collection of cell-free supernatants. We assessed the levels of pro-angiogenic factors (bFGF and VEGF) and also anti-angiogenic factors (TSP-1, endostatin, IFN-␣) in the medium obtained from purified, cultured CLL B cells after a 24 h in vitro incubation. We used PBMC from healthy donors as controls for the secreted CLL B cell culture medium experiments. In some experiments we cultured CLL B cells at 1 × 106 cells per ml for 24 h in a hypoxic (1% O2) or normoxic environment before collecting culture medium for assessment of VEGF levels. Hypoxic conditions were genLeukemia Quantification of mRNA coding for angiogenic factors/receptors VEGFR-1, VEGFR-2 and TSP-1 RT-PCR cDNA was synthesized in a 15 l reaction using the FirstStrand cDNA Synthesis Kit (Pharmacia, Piscataway, NJ, USA) 2.0 g of total RNA was dissolved in 8 l RNase-free water and incubated at 65°C for 10 min. While on ice, 5 l of the bulk first-strand mix, 1 l DTT, and 1 l pd(N)6 primer were added to the RNA and then incubated for 60 min at 37°C. VEGFR1 (FLT-1), VEGFR-2 (KDR), and TSP-1 RT-PCR was carried out in a 50 l reaction containing the following reagents: 25 l of the HotStarTaq Master Mix Kit (Qiagen); 18 l sterile water; 2.5 l each of forward and reverse primers (10 M); 2 l of cDNA preparation. The cycling conditions for the KDR and FLT-1 primers were as follows: stage 1, 95°C for 15 min; stage 2, 40 cycles of 94°C for 30 s, 55°C (KDR) or 58°C (FLT1) for 60 s, 72°C for 60 s; stage 3, 72°C for 10 min. The cycling conditions for the TSP-1 and -actin primers were as follows: stage 1, 95°C for 15 min; stage 2, (20, 23, 25, 27, 30, 35 or 40 cycles) of 94°C for 30 s, 57°C for 60 s; 72°C for 60 s; stage 3, 72°C for 10 min. The primers used were as follows: KDR forward 5⬘-GACTGCACAAACCAGCTTCTG-3⬘; reverse 5⬘-GACATCAATTGCTGAAAGC-3⬘, band size = 588 bp; FLT1 forward 5⬘-TGTCATCGTTTCCAGACCC-3⬘; reverse 5⬘-TGTT TCTTCCCACAGTCCC-3⬘, band size = 339 bp; TSP-1 forward 5⬘-GCCTGATGACAAGTTCCAAGA-3⬘; reverse 5⬘-CTTTGCG ATGCGGAGTCT-3⬘, band size = 394 bp; -actin forward 5⬘GGATCCGACTTCGAGCAAGAGATGGCCAC-3⬘; reverse 5⬘CAATGCCAGGGTACATGGTGGTG-3⬘, band size = 299 bp. HeLa cells and A549 cells were used as positive controls for KDR and Flt-1 mRNA. RT-PCR products were separated by agarose gel electrophoresis on a 2% gel and products were visualized under ultraviolet illumination of ethidium bromide stained bands. Nucleotide sequencing of the immunoglobulin (Ig) variable heavy (VH) chain region of clonal CLL B cells This was done to permit definition of CLL B cell clones that were either germline or somatic mutation type clones. B-CLL cells express pro- and anti-angiogenic molecules NE Kay et al Preparation of RNA and cDNA: Total RNA was isolated from using the Trizol reagent (GIBCO BRL, Life Technologies, Grand Island, NY, USA) according to the manufacturer’s instructions. Two g of total RNA was converted into cDNA using the Pharmacia First-Strand cDNA Synthesis Kit. Analysis of Ig VH gene mutation status: Following dilution of the cDNA reaction with distilled water, up to 50 l, 2 l of cDNA were amplified in eight separate reactions. Thus, for each B-CLL sample, seven PCR reactions were set up using sense VH family-specific framework region primers that are specific for each of the seven human Ig VH gene families in conjunction with an antisense C primer. The primer sequences were identical to those previously used by Fais et al.23 The eighth PCR reaction was set up using primers specific for -actin as previously reported.24 The PCR reactions were carried out using a Qiagen Hotstart PCR Kit (Qiagen, Valencia, CA, USA) in a total volume of 50 l and included 20 pmol of each primer, 200 M of each dNTP, and 2.5 U Hotstart Taq. A Perkin Elmer 9600 thermocycler was used as follows: initial denaturation at 94°C for 15 min followed by 34 cycles of 30 s at 94°C, 1 min at 60°C, and 1 min at 72°C. A final cycle of 10 min at 72°C was used before holding PCR reactions at 4°C. PCR products were electrophoresed on 1.5% agarose gels, bands visualized by ethidium bromide staining, and PCR products were sequenced after purification with Wizard PCR Preps (Promega, Madison, WI, USA) using an automated sequenator (Applied Biosystems, Foster City, CA, USA). In some instances, PCR products were first subcloned into the TA cloning vector (Invitrogen, San Diego, CA, USA), processed using Wizard minipreps (Promega), and sequenced using M13 forward and reverse primers. experiments. These data demonstrate that CLL B cells spontaneously secrete a variety of pro- and anti-angiogenic factors. We consistently observed CLL cell secretion of bFGF, albeit a wide range of secretion was observed (Figure 1a). In addition, a subset of CLL patients secreted endostatin and IFN␣. Only leukemic cells from one out of 35 patients secreted endostatin at approximately 0.4 ng per ml, while 32 of 35 patients’ cells secreted IFN-␣ into the culture medium. Surprisingly, we also found that CLL B cells from all patients tested secreted what appeared to be consistently detectable levels of the anti-angiogenic protein TSP-1 into the culture medium (Table 1 and Figure 1b). While all CLL B cell clones secreted bFGF and TSP-1 into the media, we only detected VEGF in the culture medium of the cultured CLL B cells from 17 of 24 patients. We also assessed the presence of TSP-1 in the cytoplasm of B-CLL cells (n = 6) using flow cytometry as previously described by us.25 All B cell clones were positive for TSP-1 in their cytoplasm with a mean percent of 57.9 ± 14.8 (range of 2.1–89.6%). The mean fluorescence intensity ranged from 59.3 to 386.8 for the six B-CLL clones tested. The mean levels of bFGF, VEGF and TSP-1 were compared for B-CLL patients (n = 29) when subdivided into low risk Rai stage (stage 0, n = 18) vs high risk Rai stage (stage 3–4, n = 11) and also for B-CLL patients subdivided based on GL vs SM type clones (Figure 2a and b). The mean levels (± standard error of the mean) bFGF (pg/ml), VEGF (pg/ml), and TSP-1 (ng/ml) in low vs high risk B-CLL patients were 72.1 ± 25.6 vs 337.9 ± 254.5, 2.4 ± 0.7 vs 6.6 ± 3.2 and 222.3 ± 114.1 vs 1.3 ± 0.4, respectively. We also compared bFGF, VEGF and TSP-1 for GL (n = 10) vs SM type clones (n = 19) in this 913 Analysis of Ig gene sequences: Nucleotide sequences were aligned with those in the V BASE sequence directory using DNAPLOT software. In a manner consistent with other reports, germline Ig sequences were defined as those with ⬍2% sequence deviation from the most similar germline Ig VH gene and those that had acquired somatic mutations were defined as those with ⭓2% sequence differences. Results CLL cells secrete both pro- and anti-angiogenic factors We initially determined the secretion of pro- and anti-angiogenic vascular factors from clonal B cells isolated from CLL patients (n = 35). The patients utilized for this were from all Rai stages but not currently on therapy. We assessed the levels of angiogenic factors in the medium of CLL B cells that had been freshly isolated and incubated without in vitro stimulation for 24 h. Table 1 displays the data obtained from these Table 1 Summary of mean values of pro- and anti-angiogenic factors secreted by CLL B cells Mean ± s.e. Range bFGFa VEGFa TSP-1a Interferon-␣a 23.9 ± 7.9 0.6–64 12.5 ± 2.3 0–33 1.9 ± 0.3 0.23–8.1 2.7 ± 0.3 1.2–6.48 a Values presented are in picograms/ml for bFGF and VEGF while TSP-1 and IFN-␣ are in nanograms/ml. These data represent the mean values obtained for 35 B-CLL patients. Figure 1 Both pro- and anti-angiogenic vascular factors are secreted from CLL B cells. (a) bFGF (pg/ml) is secreted from CLL cells albeit at variable levels. (b) CLL B cells from all patients tested secreted high levels of the anti-angiogenic protein TSP-1 (ng/ml) into the culture medium. Leukemia B-CLL cells express pro- and anti-angiogenic molecules NE Kay et al 914 Figure 2 Box plots of ELISA levels of BFGF, VEGF and TSP-1 for (a) low (L) risk vs high (H) risk and (b) GL- vs SM-type clones in a cohort of 29 B-CLL patients. same cohort of 29 patients. These values were 107.2 ± 54.1 vs 180.8 ± 48.3, 5.9 ± 1.9 vs 3.3 ± 1.8 and 202.7 ± 65.9 vs 413.8 vs 135.7 for bFGF, VEGF and TSP-1, respectively. We also evaluated and compared the level of vascular factors detected in the urine from a cohort of 31 B-CLL patients. The mean levels of bFGF, VEGF, and TSP-1 in urine were 1.7 pg/ml ± 0.7 (range 0.51–3.4), 166.6 pg/ml ± 102 (range 49.5– 454.1) and 0.15 ng/ml ± 0.21 (range 0.006–0.9), respectively. CLL B cells have augmented VEGF mRNA and protein production with exposure to hypoxia We have previously demonstrated that VEGF is found in the urine of CLL patients.14 However, since we did not detect VEGF in the culture medium of all CLL B cells exposed to ambient air, we then tested whether hypoxia, a known inducer of VEGF production,26 could induce CLL cells to upregulate the production and/or secretion of VEGF. CLL cells from three patients were cultured either under normoxic or hypoxic conditions. The levels of VEGF in the culture medium under normoxic conditions for each patient (31.8, 23.9 and 0 pg/ml, respectively) were found to increase (147.1, 36.8 and 23 pg/ml, respectively) when separate aliquots of the same CLL B cells were cultured under hypoxic conditions. Although RT-PCR analysis in a separate cohort of B-CLL patients (n = Leukemia 8) indicated that freshly isolated CLL B cells expressed variable levels of mRNA for VEGF (data not shown), we next wanted to determine whether hypoxic conditions could induce a quantitative change in the level of VEGF mRNA expressed in the CLL B cells. To accomplish this, we used a ribonuclease protection assay (RPA) to assess VEGF mRNA levels from the same three B cell clones. Figure 3 illustrates the dramatic upregulation of VEGF mRNA for CLL cells exposed to hypoxia using the RPA method. The ratio increase in VEGF mRNA for each CLL patient under hypoxic compared to normoxic conditions was 21.8, 8.4 and 5.0, respectively. CLL B cells express mRNA for several vascular factors and vascular factor receptors Since the CLL B cell clones were capable of secreting a wide variety of both pro- and anti-angiogenic factors, we then decided to use the RPA method to screen for mRNA expression of other known vascular factors and vascular receptors in CLL B cells. Figures 3 and 4 illustrate representative RPA results from isolated blood B cells from CLL patients. The eight CLL patients illustrated in Figure 3a and b were chosen based on whether the B cell clones were somatic mutation-type clones (n = 4) or germline-type clones (n = 4). This latter feature was determined by nucleotide sequencing B-CLL cells express pro- and anti-angiogenic molecules NE Kay et al 915 Figure 3 Upregulation of VEGF mRNA for CLL cells (n = 3) exposed to hypoxic vs normoxic conditions using the RPA method. The fold induction ratios for the three patients were 21.8, 8.4 and 5.0, respectively. across the immunoglobulin (Ig) heavy chain variable region. In all cases, the CLL B cells expressed mRNA for VEGFR1 (Flt1), albeit at different levels. In addition, there was consistent expression of CD31 (PECAM-1) mRNA in all the CLL B cell clones. Thrombin receptor (TR) mRNA expression was also found in three of the four germline-type clones and all four of the somatic mutation-type clones. mRNA for endoglin was noted in all eight patients shown in Figure 3. We also detected mRNA for the Tie receptor (Figure 4) in one of four somatic mutation-type clones and two of four GL-type clones. Angiopoietin mRNA was also present in all eight CLL patients. An analysis for these same mRNA species by RPA in five additional CLL B cell clones was also done. In these clones, we detected VEGFR-1 and CD31 mRNA in all five CLL B cell clones and thrombin receptor mRNA in three of the five CLL clones (data not shown). The probes available in this commercial RPA did not allow assessment of VEGFR-2 (KDR), the other major receptor known to bind and become activated by VEGF.27 Thus, we designed primers specific for that receptor and performed RT-PCR for both VEGFR-1 and VEGFR-2. Figure 5 illustrates the results of those experiments. We did detect mRNA for VEGFR-2 (seven of seven clones) and VEGFR1 (four of five CLL clones) in the CLL clones regardless of somatic or germline origin. The five patients studied for VEGFR1 were either GL (n = 2) or SM (n = 3) while the seven patients studied for VEGFR2 were either GL (n = 3) or SM (n = 5). Finally, we also designed probes for TSP-1 and conducted RT-PCR assays which did show that mRNA for this anti-angiogenic molecule exists in clonal CLL B cells (Figure 6). We studied two CLL B cell clones and normal peripheral blood B cells. Of interest, normal peripheral blood B cells also expressed TSP-1 mRNA. In order to see if hypoxia could alter TSP-1 message as previously reported,28 we performed the PCR reactions on RNA obtained from cells cultured under normoxic and hypoxic conditions. In one of two CLL clones, we were able to detect a reduction in TSP-1 mRNA with hypoxic exposure compared to normoxic conditions (Figure 6). This Figure 4 RPA analysis of various angiogenesis-related molecules. Using a Molecular Dynamics STORM Phosphorimager, mRNA levels for each of the proteins indicated were quantitated and expressed relative to levels of the L32 housekeeping gene. Four GL-type CLL (panel a) and four SM-type B-CLL (panel b) were assessed for expression levels of Flt-1, Tie, Tie-2, thrombin receptor (TR), CD31, endoglin (END) and angiopoietin (ANG). Figure 5 RT-PCR analysis of VEGFR-1 and -2 expression. Five BCLL patients were examined for VEGFR-1 in (a) (SM = 3, GL = 2) and five separate B-CLL patients (SM = 3 and GL = 2) were examined for VEGFR-2 in (b). A549 cells served as a positive control (C) for VEGFR1 and HeLa cells served as a positive control for VEGFR-2. latter finding prompted us to do simultaneous assays for VEGF and TSP-1 in the culture medium of CLL B cells from three patients that have been exposed to normoxic or hypoxic conditions. These latter patients were not the same as the ones tested for TSP-1 mRNA in Figure 6. In three experiments summarized in Table 2, all three B-CLL clones had an upregulation of VEGF with hypoxia and a diminution in their TSP-1 levels when cultured in hypoxia compared to normoxia. In preliminary work not presented here, we find that normal peripheral blood B cells also secrete TSP-1 protein as detected by ELISA. Leukemia B-CLL cells express pro- and anti-angiogenic molecules NE Kay et al 916 Figure 6 RT-PCR analysis of TSP-1 mRNA levels. Two B-CLL patients and purified blood B cells from a healthy control were assessed for TSP-1 levels by employing different cycling conditions as noted on the bottom of each gel. Note the presence of mRNA for TSP-1 and actin in all lanes for both blood B cells and the two CLL B cell clones. Also note that patient CLL1 has a downward modulation of TSP-1 mRNA after hypoxic exposure. This is in contrast to the consistent levels of mRNA for both actin and TSP-1 in both the blood B cells and CLL B cells from patient 2. N, normoxia; H, hypoxia. Table 2 Levels of VEGF and TSP-1in the culture medium of CLL B cells cultured under normoxic or hypoxic condtionsa VEGF – normoxic VEGF – hypoxic TSP-1 normoxic TSP-1 hypoxic CLL 1 CLL 2 CLL 3 25.3 63.6 10.3 7.2 19.5 36.4 10 8.3 24.0 28.9 6.4 4.6 a ELISA assays were done for VEGF or TSP-1 in the culture medium obtained after a 24-h culture of purified CLL B cells (1 × 106 per ml). CLL B cells from three separate patients were cultured under both normoxic and hypoxic conditions. Data are shown in pg/ml for VEGF and ng/ml for TSP-1. Discussion This investigation has detailed the array of angiogenic factors that are expressed by the clonal leukemic B cells in B-CLL patients. The major impetus for this study was our original observation that the bone marrows of B-CLL patients exhibit increased densities of blood vessels.14 This current study has focused on the circulating blood cells of CLL patients and our results indicate that unstimulated, circulating clonal B cells are able to synthesize and secrete a variety of both pro- and anti-angiogenic factors. While bFGF and VEGF proangiogenic factors were detected, there was also evidence of secreted anti-angiogenic factors endostatin, TSP-1 and IFN-␣, albeit at varying levels. The anti-angiogenic factor TSP-129,30 was secreted consistently and at higher levels than endostatin or IFN-␣. Hypoxic conditions were found to dramatically alter the low level of both mRNA and secreted levels of VEGF from the blood CLL B cells. In addition, we detected the presence of mRNA for the VEGF receptors; VEGFR-1 and VEGFR-2; the adhesion molecules CD31 and endoglin (CD105); the proangiogenic factor, angiopoietin; and mRNA for the Tie1 receptor and thrombin receptor. Taken together, these results strongly suggest that the blood B cells in CLL are malignant cells with significant potential to interact with and/or be modulated by angiogenic factors. In addition, the tissues infiltrated by these malignant B cell clones could have altered Leukemia development and growth of resident blood vessels based on the angiogenic molecules expressed by CLL B cells. The role of angiogenesis in the biology of B-CLL is as yet unclear. However, our initial study of bone marrow tissue and a more recent study of nodal sites in B-CLL14,15 adds to the evidence that abnormal angiogenesis exists in this disease. In both studies, either increased numbers of blood vessels in the bone marrow or neovascularization of lymph node sites were documented. Major questions from the B-CLL tissue-based observations include what is the role if any for abnormal angiogenesis in the initiation or progression of this B cell leukemia and do the CLL B cells themselves assist in the induction of this angiogenesis? The latter study did find evidence for VEGF protein in both clonal B cells circulating in the blood and those cells resident in nodal tissue sites.15 Our own data on circulating clonal B cells indicate that some, but not all, B cell clones isolated from blood spontaneously release VEGF protein into culture medium. Of interest, we always detected VEGF in the urine of CLL patients indicating that in vivo there is a consistent production of this pro-angiogenic factor.14 Since VEGF expression can be modulated by several factors including hypoxia,26 activated oncogenes31,32 and various cytokines,33–36 the exact angiogenic status of a CLL clone in vivo in different sites is probably not easy to predict. Our experiments confirm15 that CLL B cells exposed to hypoxia have a brisk increase in the detectable levels of VEGF protein secretion. The elevation of VEGF in hypoxic conditions was variable from clone to clone but does suggest that under the right microenvironmental conditions CLL B cells are capable of releasing increased levels of VEGF. In addition, the increase in CLL VEGF protein release was paralleled by quantitative increases in mRNA for VEGF. This suggests that the augmented VEGF secretion is related to increased synthesis of new VEGF protein. The ELISA methods we used to evaluate VEGF in CLL cells utilized a monoclonal antibody reactive for VEGF isoform 165. VEGF 165 is the predominant molecular type synthesized by both normal and malignant cells.37 This VEGF isoform and VEGF isoform 121 have also been detected from CLL cells stimulated by hypoxia.15 Interestingly, it is the 165 isoform that is a potent mitogen for vascular endothelial cells. VEGF isoform 165 is able to bind both VEGFR1 and 2 B-CLL cells express pro- and anti-angiogenic molecules NE Kay et al receptors and subsequently generate angiogenesis and vasculogenesis.38,39 Indeed, the study by Chen et al15 did show that CLL culture medium was able to induce angiogenesis in vitro. Of interest, the other major biologic impact of VEGF is to enhance the survival of endothelial cells.40 VEGF has been shown to increase levels of Bcl-2 and A1, anti-apoptotic proteins in human endothelial cells.41 Since resistance to apoptosis is also a critical feature of CLL B cells, this increases the likelihood that VEGF could have important biologic effects on CLL B cells by virtue of an autocrine pathway. Our demonstration that CLL B cells consistently express mRNA for VEGFR1 and VEGFR2 and express VEGFR1 and 2 by flow cytometry (results not shown) adds to the evidence that a VEGF-based autocrine pathway in CLL B cells may exist. Although we do not have direct evidence that such an autocrine pathway exists in this disease, we do have preliminary data that suggest that incubating CLL cells with anti VEGF-receptor antibody can increase CLL B cell apoptotic rates. There are two high-affinity VEGF specific receptor tyrosine kinase (RTK) receptors (Flt-1 or VEGFR-1 and flk-1/KDR or VEGFR-1) described (reviewed in Ref. 37) that bind VEGF and generate cell signals that ultimately result in the modulation of angiogenesis and vasculogenesis. Activation of the two VEGF RTK receptors by VEGF has been reported to induce differential endothelial cell responses implying that signal transduction pathways may be different for the two receptors.42,43 The VEGFR-1 and -2 receptors are known to be expressed predominantly in endothelial cells.38 However, other cell types have also been shown to express angiogenictype receptors including both normal and tumor cells. The VEGFR-1 receptor is found on renal mesangial cells, monocytes and trophoblasts.44–46 VEGFR-2 has been found on marrow stem cells, megakaryocytes and retinal progenitor cells.47,48 Malignant melanoma cells have been found to express VEGFR receptors.49,50 VEGFR-1, and to a lesser extent, VEGFR-2, have been found by RT-PCR in acute leukemic blasts.51–53 Flow cytometry and RT-PCR54 methods allowed detection of Tie receptor expression in approximately 20% of acute leukemia cases. Finally, a recent report has detected the expression, albeit variable, of both Flt-1 and Tie1 in CLL B cells using a Western blot and solid phase radioimmunoassay.55 Thus, there is ample evidence that human leukemic cells and in particular, CLL B cells, can express these RTK receptors. The potential interaction of VEGF and VEGFR1/VEGFR-2 in CLL cells is probably not the only way in which angiogenesis factors might influence CLL B cells. The ELISA data and RPA data indicate that the CLL B cell can generate a surprisingly diverse repertoire of angiogenic molecules. Thus, there are both pro- (bFGF, VEGF) and anti-angiogenic (endostatin, TSP-1, IFN-␣) molecules secreted by the CLL B cell clones. We have previously shown that CLL patients have high levels of bFGF in the urine.14 Our current study indicates that CLL B cells are spontaneously secreting this pro-angiogenic molecule and thus are probably the source of the high levels of bFGF found in the urine. These findings have relevance to the potential for an angiogenic switch in CLL much like that seen in solid tumors.4 The angiogenic switch in tumor cells is characterized at a molecular level by an imbalance in the production of pro- vs anti-angiogenic factors.4,5 In the case of CLL, this could be defined by a variety of changes in the secretion profile of a CLL clone including increased VEGF production and/or decrease in TSP-1, endostatin, and/or IFN-␣. In order to determine if these vascular factors are associated with more aggressive disease in B-CLL, we compared the vascular factor levels secreted by CLL clones to the Rai risk and whether a patient had a GL- or SM-type clone. We found that mean VEGF levels were higher while TSP-1 levels were lower for both high vs low risk Rai and GL- vs SM-type B cell clones. Thus, the levels of these factors are indeed consistent with a relative conversion from more anti-angiogenic factors (ie TSP1) to a pro-angiogenic (ie VEGF) state. Why this might occur is not yet evident in B-CLL but critical features to be evaluated include hypoxic microenvironments and/or genetic mutations that might be occurring in the B cell clone. Hypoxia in marrow or nodal environments could be a factor since we were able to find that hypoxic conditions generated both an increase in mRNA for VEGF and VEGF protein while TSP mRNA was decreased in CLL B cells. Two candidate genetic loci known to regulate angiogenesis include Ras and the p53 tumor suppressor gene.33,56 While Ras mutations have not been commonly found in CLL cells, mutations in p53 are documented in progressive or transformed CLL.57 Since mutations in this tumor suppressor gene are known to downregulate TSP-1,58 it is tempting to speculate that the high levels of TSP-1 spontaneously secreted from CLL cells could be decreased with the onset of p53 mutations in CLL clones. This alteration may be sufficient for the angiogenic switch to occur in CLL patients. Of interest, the normal p53 gene can also increase the level of the pro-angiogenic factor VEGF.59 Future work will need to evaluate the association of hypoxia and/or alterations found in the CLL B cell clone with the production of both pro- and anti-angiogenic molecules. The relatively extensive profile of angiogenic molecules found in CLL cells also included mRNA for CD31, endoglin, Tie1 and angiopoietin. These expression patterns suggest that multiple alternative pathways could exist for angiogenesisrelated functions in CLL B cells. Thus, CD31 or platelet endothelial cell adhesion molecule-1 (PECAM), is an endothelial adhesion molecule known to be present on CLL cells.60 CD31 has been implicated as a pro-angiogenic molecule in human endothelial cells.61 The presence of CD31 on the membrane of CLL B cells may provide important signals for synthesis of vascular growth factors. Endoglin is a receptor for TGF- a molecule with pro-angiogenic activity.62 CLL cells are also known to secrete TGF-,63 thus providing another potential autocrine circuit for modulation of angiogenic factors in the CLL B cell. Of interest, TSP-1 has been implicated in the activation of TGF-64 suggesting an additional role for TSP-1 in clonal B cell biology. Finally, we also detected low levels of mRNA for angiopoietin, another protein that can increase angiogenesis. There are two types of angiopoietin and both can bind to Tie-2.65 However, since we saw no mRNA for the Tie-2 receptor, this autocrine pathway is less likely to be operative in CLL. Taken together, we believe our data show that the leukemic cells in B-CLL have a surprisingly diverse capacity to synthesize many pro- and anti-angiogenic factors. In addition, we now know that the CLL B cells express mRNA and have membrane expression for several critical vascular receptors (VEGFR-1, VEGFR-2 and TIE) along with CD31, an important angiogenic adhesion molecule. The exact role of each one of these vascular factors in the pathophysiology of the disease remains to be determined. However, we believe these data provide the foundation for the pursuit of novel autocrine pathways that could impact at least the apoptosis resistance known to be a prominent feature of the clonal B cells. 917 Leukemia B-CLL cells express pro- and anti-angiogenic molecules NE Kay et al 918 References 1 Folkman J (ed.). Tumor Angiogenesis. BC Becker: Canada, 2000. 2 Folkman J. Tumor angiogenesis: therapeutic implications. N Engl J Med 1971; 285: 1182–1186. 3 Folkman J. Clinical applications of research on angiogenesis. N Engl J Med 1995; 333: 1757–1763. 4 Hanahan D, Folkman J. Patterns and emerging mechanisms of the angiogenic switch. Cell 1996; 86: 353–364. 5 Kerbel R. Tumor angiogenesis: past, present and the near future. Carcinogenesis 2000; 21: 505–515. 6 Perez-Atayde A, Sallan S, Tedrow U, Connors S, Allred E, Folkman J. Spectrum of tumor angiogenesis in the bone marrow of children with acute lymphoblastic leukemia. Am J Pathol 1997; 15: 815– 821. 7 Rajkumar S, Fonseca R, Witzig T, Gertz M, Greipp P. Bone marrow angiogenesis in patients achieving complete response after stem cell transplantation for multiple myeloma. Leukemia 1999; 13: 469–472. 8 Vacca A, Ribatti D, Ruco L, Giacchetta F, Nico B, Quondamatteo F, Ria R, Iurlaro M, Dammacco F. Angiogenesis extent and macrophage density increase simultaneously with pathological progression in B-cell non-Hodgkin’s lymphomas. Br J Cancer 1999; 79: 965–970. 9 Aguayo A, Kantarjian H, Manshouri T, Gidel C, Estey E, Thomas D, Koller C, Estrov Z, O’Brien S, Keating M, Freireich E, Albitar M. Angiogenesis in acute and chronic leukemias and myelodysplastic syndromes. Blood 2000; 96: 2240–2245. 10 Kini A, Peterson L, Tallman M, Lingen M. Angiogenesis in acute promyelocytic leukemia: induction by vascular endothelial growth factor and inhibition by all-trans retinoic acid. Blood 2001; 97: 3919–24. 11 Dominici M, Campioni D, Lanza F, Luppi M, Barozzi P, Pauli S, Milani R, Cavazzini F, Punturieri M, Trovato R, Torelli G, Castoldi G. Angiogenesis in multiple myeloma: correlation between in vitro endothelial colonies growth (CFU-En) and clinical-biological features. Leukemia 2001; 15: 171–176. 12 Di Raimondo F, Azzaro M, Palumbo G, Bagnato S, Stagno F, Giustoisi GM, Cacciola E, Sortino G, Guglielmo P, Giustolisi R. Elevated vascular endothelial growth factor (VEGF) serum levels in idopathic myelofibrosis. Leukemia 2001; 15: 976–980. 13 Allouche M. Basic fibroblast growth factor and hematopoiesis. Leukemia 1995; 9: 937–942. 14 Kini A, Kay N, Peterson L. Increased bone marrow angiogenesis in B-cell chronic lymphocytic leukemia. Leukemia 2000; 14: 1414–1418. 15 Chen H, Treweeke A, West D, Till KJ, Cawley JC, Zuzel M, Toh CH. In vitro and in vivo production of vascular endothelial growth factor by chronic lymphocytic leukemia cells. Blood 2000; 96: 3181–3187. 16 Aguayo A, O’Brien S, Keating M, Manshouri T, Gidel C, Barlogie B, Beran M, Koller C, Kantarjian H, Albitar M. Clinical relevance of intracellular vascular endothelial growth factor levels in B-cells chronic lymphocytic leukemia. Blood 2000; 96: 768–770. 17 Menzel T, Rahman Z, Calleja E, White K, Wilson EL, Wieder R, Gabrilove J. Elevated intracellular level of basic fibroblast growth factor correlates with stage of chronic lymphocytic leukemia and is associated with resistance to fludarabine. Blood 1996; 87: 1056–1063. 18 Krejci P, Dvorakova D, Krahulcova E, Pachernik J, Mayer J, Hampi A, Dvorak P. FGF-2 abnormalities in B cell chronic lymphocytic and chronic myeloid leukemias. Leukemia 2001; 15: 228–237. 19 Hamblin T, Davis Z, Gardiner A, Oscier D, Stevenson F. Unmutated Ig V-H genes are associated with a more aggressive form of chronic lymphocytic leukemia. Blood 1999; 94: 1848–1854. 20 Damle R, Wasil T, Fais F, Ghiotto F, Valetto A, Allen SL, Buchbinder A, Budman D, Dittmar K, Kolitz J, Lichtman SM, Schulman P, Vinciguerra VP, Rai KR, Ferrarini M, Chiorazzi N. IgV gene mutation status and CD38 expression as novel prognostic indicators in chronic lymphocytic leukemia. Blood 1999; 94: 1840– 1847. 21 Jelinek D, Tschumper R, Geyer S, Bone ND, Dewald GW, Hanson CH, Stenson MJ, Witzig TE, Tefferi A, Kay NE. Analysis of clonal B cell CD38 and immunoglobulin variable region sequence status in relation to clinical outcome for B-chronic Lymphocytic Leukemia. Br J Haematol 2001; 115: 854–861. Leukemia 22 Cheson B, Bennett J, Grever M, Kay NE, Keating MJ, O’Brien S, Rai KR. National Cancer Institute-sponsored Working Group guidelines for chronic lymphocytic leukemia: revised guidelines for diagnosis and treatment. Blood 1996; 87: 4990–4997. 23 Fais F, Ghiotto F, Hashimoto S, Sellars B, Valetto A, Allen SL, Schulman P, Vinciguerra VP, Rai KR, Rassenti LZ, Kipps TJ, Dighiero G, Schroeder HW Jr, Ferrarini M, Chiorazzi N. Chronic lymphocytic leukemia B cells express restricted sets of mutated and unmutated antigen receptors. J Clin Invest 1998; 102: 1515–1525. 24 Westendorf J, Jelinek D. Growth regulatory pathways in myeloma: evidence for autocrine oncostatin M expression. J Immunol 1996; 157: 3081–3088. 25 Kay N, Han L, Bone N, Williams G. Interleukin-4 content in chronic lymphocytic leukaemia (CLL) B cells and blood CD8 T cells from B-CLL patients: impact on clonal B-cell apoptosis. Br J Haematol 2001; 112: 760–767. 26 Shweiki D, Itin A, Soffer D, Keshet E. Vascular endothelial growth factor induced by hypoxia, a mediator hypoxia-initiated angiogenesis. Nature 1992; 359: 843–845. 27 Neufeld G, Cohen T, Gengrinovitch S, Poltorak Z. Vascular endothelial growth factor (VEGF) and its receptors. FASEB J 1999; 13: 9–22. 28 Laderoute K, Alarcon R, Brody M, Calagan JM, Chen EY, Knapp AM, Yun Z, Denko NC, Giaccia AJ. Opposing effects of hypoxia on expression of the angiogenic inhibitor thrombospondin 1 and the angiogenic inducer vascular endothelial growth factor. Clin Cancer Res 2000; 6: 2941–2950. 29 Simantov R, Febbraio M, Crombie R, Asch A, Nachman R, Silverstein R. Histidine-rich glycoprotein inhibits the antiangiogenic effect of thrombospondin-1. J Clin Invest 2001; 107: 45–52. 30 Jin R, Kwak C, Lee S, Soo CG, Park MS, Lee E, Lee SE. The application of an anti-angiogenic gene (thrombospondin-1) in the treatment of human prostate cancer xenografts. Cancer Gene Ther 2000; 7: 1537–1542. 31 Mukhopadhyay D, Knebelmann B, Cohen H, Ananth S, Sukhatme V. The von Hippel–Lindau tumor suppressor gene product interacts with Sp1 to repress vascular endothelial growth factor promoter activity. Mol Cell Biol 1997; 17: 5629–5639. 32 Van Meir E, Polverini P, Chazin V, Su Huang H, deTribolet N, Cavenee W. Release of an inhibitor of angiogenesis upon induction of wild type p53 expression in glioblastoma cells. Nat Genet 1994; 8: 171–176. 33 Ryuto M, Ono M, Izumi H, Yoshida S, Weich HA, Kohno K, Kuwano M. Induction of vascular endothelial growth factor by tumor necrosis factor alpha in human glioma cells – possible roles of SP-1. J Biol Chem 1996; 271: 28220–28228. 34 Pertovaara L, Kaipainen A, Mustonen T, Orpana A, Ferrara N, Saksela O, Alitalo K. Vascular endothelial growth factor is induced in response to transforming growth factor-beta in fibroblastic and epithelial cells. J Biol Chem 1994; 269: 6271–6274. 35 Cohen T, Nahari D, Cerem L, Neufeld G, Levi B. Interleukin-6 induces the expression of vascular endothelial growth factor. J Biol Chem 1996; 271: 736–741. 36 Li J, Perrella M, Tsai J, Yet SF, Hsieh CM, Yoshizumi M, Patterson C, Endege WO, Zhou F, Lee ME. Induction of vascular endothelial growth factor gene expression by interleukin-1 beta in rat aortic smooth muscle cells. J Biol Chem 1995; 270: 308–312. 37 Gerwins P, Skoldenberg E, Claesson-Welsh L. Function of fibroblast growth factors and vascular endothelial growth factors and their receptors in angiogenesis. Crit Rev Oncol/Hematol 2000; 34: 185–194. 38 Ferrara N. Vascular endothelial growth factor: molecular and biological aspects. Curr Top Microbiol Immunol 1999; 237: 1–30. 39 Ferrara N, Davis-Smyth T. The biology of vascular endothelial growth factor. Endocrine Rev 1997; 18: 4–25. 40 Liu W, Ahmad S, Reinmuth N, Shaheen RM, Jung YD, Fan F, Ellis LM. Endothelial cell survival and apoptosis in the tumor vasculature. Apoptosis 2000; 5: 323–328. 41 Gerber H, Dixit V, Ferrara N. Vascular endothelial growth factor induces expression of the antiapoptotic protein Bcl-2 and A1 in vascular endothelial cells. J Biol Chem 1998; 273: 13313–13316. 42 Seetharam L, Gotoh N, Maru Y, Neufeld G, Yamaguchi S, Shibuya M. A unique signal transduction from FLT tyrosine kinase, a receptor for vascular endothelial growth factor (VEGF). Oncogene 1995; 10: 135–147. B-CLL cells express pro- and anti-angiogenic molecules NE Kay et al 43 Waltenberger J, Claesson-Welsh L, Siegbahn A, Shibuya M, Heldin C. Different signal transduction properties of KDR and Flt1, two receptors for vascular endothelial growth factor. J Biol Chem 1994; 269: 26988–26995. 44 Charnock-Jones D, Sharkey A, Boocock C, Ahmed A, Plevin R, Ferrara N, Smith SK. Vascular endothelial growth factor receptor localization and activation in human trophoblast and choriocarcinoma cells. Biol Reprod 1994; 51: 524–530. 45 Barleon B, Sozzani S, Zhou D, Weich H, Mantovani A, Marme D. Migration of human monocytes in response to vascular endothelial growth factor (VEGF) is mediated via the VEGF receptor flt1. Blood 1996; 87: 3336–3343. 46 Takahashi T, Shirasawa T, Miyake K, Yahagi Y, Maruyama N, Kasahara N, Kawamura T, Matsumura O, Mitarai T, Sakai O. Protein tyrosine kinases expressed in glomeruli and cultured glomerular cells: FLT-1 and VEGF expression in renal mesangial cells. Biochem Biophys Res Commun 1995; 209: 218–226. 47 Katoh O, Tauchi H, Kawaishi K, Kimura A, Satow Y. Expression of the vascular endothelial growth factor (VEGF) receptor gene, KDR, in hematopoietic cells and inhibitory effect of VEGF on apoptotic cell death caused by ionizing radiation. Cancer Res 1995; 55: 55687–5692. 48 Yang X, Cepko C. Flk-1, a receptor for vascular endothelial growth factor (VEGF), is expressed by retinal progenitor cells. J Neurosci 1996; 16: 6089–6099. 49 Gitay-Goren H, Halaban R, Neufeld G. Human melanoma cells but not normal melanocytes express vascular endothelial growth factor receptors. Biochem Biophys Res Commun 1993; 190: 702–709. 50 Cohen T, Gitay-Goren H, Sharon R, Shibuya M, Halaban R, Levi B, Neufeld G. VEGF-121, a vascular endothelial growth factor (VEGF) isoform lacking heparin binding ability, requires cell surface heparan sulfates for efficient binding to the VEGF receptors of human melanoma cells. J Biol Chem 1995; 270: 11322–11326. 51 Fiedler W, Graeven U, Ergun S, Verago S, Kilic N, Stockshlader M, Hossfeld DK. Vascular endothelial growth factor, a possible paracrine growth factor in human acute myeloid leukemia. Blood 1997; 89: 1870–1875. 52 Bellamy W, Richter L, Frutiger Y, Grogan T. Expression of vascular endothelial growth factor and its receptors in hematopoietic malignancies. Cancer Res 1999; 59: 728–733. 53 Dias S, Hattori K, Zhu Z, Heisseg B, Choy M, Lane W, Wu Y, Chadburn A, Hyjek E, Gill M, Hicklin DJ, Witte L, Moore MAS, Rafii S. Autocrine stimulation of VEGFR-2 activates human leukemic growth and migration. J Clin Invest 2000; 106: 511–521. 54 Kukk E, Wartiovaara U, Gunji Y, Kaukonen J, Buhring HP, Rappolo I, Matikainen M-T, Vihko P, Partanen J, Palotie A, Alitalo K, 55 56 57 58 59 60 61 62 63 64 65 Alitalo R. Analysis of TIE receptor tyrosinase in haematopoietic progenitor and leukemia cells. Br J Haematol 1997; 98: 195–203. Aguayo A, Manshouri T, O’Brien S, Keating M, Beran M, Koller C, Kantarjian H, Rogers A, Albitar M. Clinical relevance of Flt1 and Tie 1 angiogenesis receptors expression in B-cell chronic lymphocytic leukemia (CLL). Leukemia Res 2001; 25: 279–285. Pal S, Datta K, Khosravi-Far R, Mukhopadhyay D. Role of protein kinase C zeta in Ras-mediated transcriptional activation of vascular permeability factor/vascular endothelial growth factor expression. J Biol Chem 2001; 276: 2395–2403. Callet-Bauchu E, Salles G, Gazzo S, Poncet C, Morel D, Pages J, Coiffier B, Coeur P, Felman P. Translocations involving the short arm of chromosome 17 in chronic B-lymphoid disorders: frequent occurrence of dicentric rearrangements and possible association with adverse outcome. Leukemia 1999; 13: 460–468. Grant S, Kyshtoobayeva A, Kurosaki T, Jakowatz J, Fruehauf J. Mutant p53 correlates with reduced expression of thrombospondin-1, increased angiogenesis, and metastatic progression in melanoma. Cancer Detect Prevent 1998; 22: 185–194. Zhang L, Yu D, Hu M, Xiong SB, Lang AQ, Ellis LM, Pollock RE. Wild-type p53 suppresses angiogenesis in human leiomyosarcoma and synovial sarcoma by transcriptional suppression of vascular endothelial growth factor expression. Cancer Res 2000; 60: 3655–3661. Sembries S, Pahl H, Stilgenbauer S, Dohner H, Schreiver F. Reduced expression of adhesion molecules and cell signaling receptors by chronic lymphocytic leukemia cells with 11q deletion. Blood 1999; 93: 624–631. Horak E, Leek R, Klenk N, Le Jeune S, Smith K, Stuart N, Greenall M, Stepniewska K, Harris AL. Angiogenesis assessed by platelet endothelial cell adhesion molecule antibodies, as indicators of node metastases and survival in breast cancer. Lancet 1992; 340: 1120–1124. Wong S, Hamel L, Chevalier S. Endoglin expression on human microvascular endothelial cells – association with betaglycan and formation of higher order complexes with TGF-beta signaling receptors. Eur J Biochem 2000; 267: 5550–5560. Schuler M, Tretter T, Schneller F, Huber C, Peschel C. Autocrine transforming growth factor-beta from chronic lymphocytic leukemia-B cells interferes with proliferative T cell signals. Immunobiology 1999; 200: 128–139. Yevdokimova N, Wahab N, Mason R. Thrombospondin-1 is the key activator of TGF-beta 1 in human mesangial cells exposed to high glucose. J Amer Soc Neph 2001; 12: 703–712. Horner A, Bord S, Kelsall A, Coleman N, Compston J. Tie2 ligands angiopoietin-1 and angiopoietin-2 are coexpressed with vascular endothelial cell growth factor in growing human bone. Bone 2001; 28: 65–71. 919 Leukemia
© Copyright 2025 Paperzz