Abstract
Vascular endothelial growth factor (VEGF) performs multifaceted functions in the tumor microenvironment promoting angiogenesis, suppressing anti-tumor immune response, and possibly exerting autocrine functions on tumor cells. However, appropriate syngeneic animal models for in vivo studies are lacking. Using retroviral transfection and fluorescence-activated cell sorting, we generated a C57BL6 murine ovarian carcinoma cell line that stably overexpresses the murine VEGF164 isoform and the enhanced green fluorescent protein. VEGF164 overexpression dramatically accelerated tumor growth and ascites formation, significantly enhanced tumor angiogenesis, and substantially promoted the survival of tumor cells in vivo. In vitro, VEGF164 overexpression significantly enhanced cell survival after growth factor withdrawal and conferred resistance to apoptosis induced by cis-platin through an autocrine mechanism. VEGF/green fluorescent protein-expressing tumors were not recognized by the adaptive immune system. After vaccination, a specific anti-tumor T-cell response was detected, but tumor growth was not inhibited. This engineered murine carcinoma model should prove useful in the investigation of the role of VEGF in modulating the tumor microenvironment and affecting the complex interactions among angiogenesis mechanisms, anti-tumor immune mechanisms, and tumor cell behavior at the natural state or during therapy in ovarian carcinoma.
Vascular endothelial growth factor (VEGF) plays a critical role in angiogenesis during embryonic development, growth, wound healing, and the cyclic regeneration of female reproductive tissues. 1,2 Solid tumor growth also depends on the development of new blood supply, which is supported by VEGF. 1,3 Human VEGF exists as five isoforms, VEGF121, VEGF145, VEGF165, VEGF189, and VEGF206, produced by alternative splicing of a primary transcript from a single VEGF gene. 4 Similarly, murine VEGF exists as at least four isoforms, VEGF120, VEGF144, VEGF164, and VEGF188, respectively. 5,6 The larger molecular weight isoforms, VEGF206 and VEGF189 are cell-associated, whereas the smaller isoforms VEGF165 and VEGF121 are secreted. 7 The different isoforms may be differentially regulated and may exert different functions during development. 8,9 VEGF164 has been shown to be primarily responsible for tumor angiogenesis in mouse models. 10,11 Two shared tyrosine-kinase receptors, VEGF receptor-1 (flt-1) and VEGF receptor-2 (KDR/flk-1) 12,13 and two co-receptors, neuropilin-1 and neuropilin-2 14,15 mediate the effects of VEGF.
Accumulating evidence indicates that VEGF exerts multifaceted functions in tumors and its overexpression of VEGF by tumors has been correlated with poor outcome. 16-21 VEGF receptors have been detected in a variety of tumor cells 22-29 and VEGF promotes the growth, proliferation, survival and/or migration of tumor cells. 24,26,27,30-32 In addition, VEGF exerts a local intratumoral as well as systemic immune suppression by inhibiting the differentiation and maturation of dendritic cells (DCs), 33,34 a process that is necessary for tumor antigen presentation and stimulation of anti-tumor T cells. Although the angiogenic effects of VEGF have been thoroughly documented in animal models, the role of VEGF in modulating the tumor microenvironment and affecting the complex interactions among angiogenesis mechanisms, anti-tumor immune mechanisms, and tumor cell behavior at the natural state or during tumor therapy remains elusive. Such studies necessitate dependable animal models fulfilling specific requirements. First, the growth of experimental tumors needs to be angiogenesis-dependent. Second, a syngeneic model is necessary to study molecular and cellular interactions in the immunocompetent host. Furthermore, experimental tumors need to mimic human tumors in their immunological behavior, namely they should harbor potential antigens but be able to evade immune recognition and/or attack. Finally, to study the direct effects of VEGF, tumor cells should be susceptible to the autocrine effects of VEGF. Ideally, an animal model should recapitulate a human tumor in which VEGF may exert important biological effects.
Epithelial ovarian cancer is the most frequent cause of gynecological cancer-related mortality and accounts for ∼15,000 deaths in the United States yearly. 35 Increased levels of tumor VEGF have been reported in invasive ovarian carcinoma compared to benign tumors or tumors of low malignant potential. 36-38 Besides tumor growth, VEGF has been implicated in the pathogenesis of ovarian cysts and ascites, 39,40 where markedly elevated levels of VEGF are seen. 38 Serum levels of VEGF are 10-fold higher in patients with advanced ovarian cancer compared to healthy controls. 38 Importantly, increased serum and/or tumor levels of VEGF have been associated with poor clinical outcome. 16,41,42 Finally, ovarian carcinoma cells express the VEGF receptor-2 KDR/flk-1. 22 Ovarian carcinoma therefore offers important opportunities to investigate the multifaceted functions of VEGF.
In the present study, we report the generation of a syngeneic model of ovarian carcinoma in the C57BL6 mouse overexpressing murine VEGF164. This engineered murine model offers a valuable tool to investigate the effects of VEGF in the biology of ovarian carcinoma and its response to therapy in the immunocompetent host. This model is also valuable for the investigation of tumor-host interactions and anti-tumor immune mechanisms as well as cellular and molecular mechanisms underlying tumor spread and metastasis in ovarian cancer.
Materials and Methods
Cell Culture and Reagents
ID8, a cell line derived from spontaneous in vitro malignant transformation of C57BL6 mouse ovarian surface epithelial cells, was a generous gift from Dr. Paul F. Terranova, University of Kansas. 43 ID8 cells were maintained in Dulbecco’s modified Eagle’s medium (Invitrogen, Carlsbad, CA) supplemented with 4% fetal bovine serum, 100 U/ml penicillin, 100 μg/ml streptomycin, 5 μg/ml insulin, 5 μg/ml transferrin, and 5 ng/ml sodium selenite (Roche, Indianapolis, IN) in a 5% CO2 atmosphere at 37°C. In some experiments, ID8 cells were cultured in serum-free and insulin-free media overnight, or in serum-free conditions in the presence or absence of cis-platin for 24 hours. Dose-response experiments were performed to define the sensitivity of ID8 cells to the drug, and final experiments were performed at a 50 μmol/L concentration. Select cells treated with cis-platin were exposed to recombinant murine VEGF (100 ng/ml; R&D Systems, Minneapolis, MN). All experiments were repeated three times. All reagents were analytical grade and purchased from Sigma (St. Louis, MO) unless otherwise specified.
Construction of VEGF/Green Fluorescent Protein (GFP) Retroviral Vector and Cell Transfection
The cDNA of murine VEGF164 was a generous gift from Dr. Patricia D’Amore, Harvard University. 5 A murine stem cell retroviral vector MigR1 containing a coding sequence for enhanced GFP and an internal ribosome entry site, as well as BOSC23, a murine kidney 293T-derived packaging cell line, were generously provided by Dr. Warren Pear, University of Pennsylvania. 44 The VEGF/GFP retroviral vector was constructed as follows: plasmid containing VEGF164 cDNA was digested with XbaI and XhoI to release VEGF164 cDNA; MigR1 vector was digested with EcoRI and XhoI. VEGF164 cDNA fragment was inserted into MigR1 upstream of internal ribosome entry site and GFP using T4 DNA ligase. The sequence of the above constructs was confirmed by multiple restriction digestion analysis and direct sequencing (not shown). Retroviral vector containing GFP alone or VEGF164 plus GFP was transfected into BOSC23 cells using the calcium phosphate method. ID8 cells were infected with retrovirus in the presence of 8 μg/ml of polybrene. After 72 hours, infected cells were examined under a fluorescent microscope.
Flow Cytometry and Cell Sorting
Cells were analyzed on a FACScan (Becton Dickinson, San Jose, CA) using CellQuest flow cytometry analysis software (Becton Dickinson). GFP was detected using a 530/30-nm bandpass filter. Ascites leukocytes were detected with allophycocyanin (APC)-labeled rat anti-mouse CD45 monoclonal antibody (BD Pharmingen, San Diego, CA). Monoclonal antibodies against major histocompatibility complex class I molecules (MHC-I) (H-2kb/H-2Db, biotinylated), MHC-II (KH74, biotinylated), isotype control (IgG2ak, biotinylated), CD11c (HL3, APC-conjugated), CD86 (GL-1, fluorescein isothiocyanate-conjugated), and CD3 (17A2, fluorescein isothiocyanate-conjugated) were purchased from BD Pharmingen. Data were recorded and analyzed with CellQuest software (Becton Dickinson). Sorting was performed on a MoFlo cell sorter (Cytomation, Fort Collins, CO) equipped with an argon laser beam. All flow cytometry data were analyzed by uploading data files into FlowJo (TreeStar, Inc.). Cells were sorted at a flow rate of 1000 to 3000 cells/second as GFP-negative, GFP-low-positive, and GFP-high-positive polyclonal cell populations.
Animals
Six- to 8-week-old female C57BL6 mice (Charles River Laboratories, Wilmington, MA) were used in protocols approved by the Institutional Review Board of the Wistar Institute and the University of Pennsylvania.
In Vivo Tumor Generation
Subconfluent ID8 cells were trypsinized, washed twice, and harvested by centrifugation at 1000 × g for 5 minutes. A single-cell suspension was prepared in phosphate-buffered saline (PBS), or PBS mixed with an equal volume of cold Matrigel (BD Biosciences, Bedford, MA) at 10 mg/ml. For flank injections, a total volume of 0.5 ml containing 5 × 106 VEGF/GFP-transfected cells was injected subcutaneously into the flank of 8-week-old C57BL6 mice, whereas in some experiments the other flank was injected with the same number of control GFP-transfected cells (n = 7) or wild-type cells (n = 7). Tumors were detectable 2 weeks later and tumor size was measured weekly thereafter using a Vernier caliper. Tumor volumes were calculated by the formula V = 1/2 (L × W)2, where L is length (longest dimension) and W is width (shortest dimension). 45 Mice were sacrificed 5 weeks after flank injection. For intraperitoneal injections, a total volume of 0.7 ml of PBS containing 7 × 106 VEGF/GFP-transfected cells (n = 10), GFP-transfected cells (n = 10), or wild-type cells (n = 10) cells was inoculated into the mouse peritoneal cavity. Animals were followed for survival or were sacrificed 8 weeks after inoculation to evaluate tumor growth. Moribund animals were euthanized according to the protocols of the Wistar Institute and the University of Pennsylvania. To measure serum VEGF levels, whole blood samples were obtained by retro-orbital bleed and allowed to clot for 1 hour at room temperature. Intraperitoneal VEGF levels were determined in ascites supernatants collected 6 weeks after inoculation of intraperitoneal tumor or from healthy control animals.
Animal Vaccinations
For the preparation of whole tumor cell vaccine, ID8 cells transfected with VEGF/GFP-positive retrovirus were rinsed with PBS twice, cultured in serum-free media for 48 hours to minimize fetal bovine serum xenoantigens, and subsequently subjected to UVB rays at increasing energy to identify the dose of UVB that induces apoptosis in 100% of cells. Apoptosis was quantified through annexin-V and propidium iodide staining by flow cytometry, whereas killing efficacy was confirmed by cell proliferation assay using CellTiter96 kits (Promega, Madison, WI) according to the manufacturer. Immediately after exposure to lethal UVB, cells were rinsed twice in PBS and 1 × 105 apoptotic tumor cells suspended in 0.3 ml of PBS were injected subcutaneously to 6-week-old healthy female mice. Control mice received PBS injection. Mice were immunized twice, 1 week apart, and challenged with subcutaneous inoculations of 5 × 106 live VEGF/GFP-transfected ID8 cells 1 week after the second vaccination. Animals were sacrificed 8 weeks later and tumors were resected and measured as above.
Live Fluorescent Stereo Microscopy
The gross morphology of tumors was observed using a fluorescence stereo microscope (SMZ800; Nikon, Tokyo, Japan) equipped with a 100-W mercury lamp. Emitted fluorescence was acquired through a long pass filter Ex 480/20 on a CoolSNAP Pro color digital camera (Media Cybernetics, Silver Spring, MD).
RNA Isolation and Reverse Transcriptase-Polymerase Chain Reaction (RT-PCR)
Total RNA was isolated from 1 × 106 cultured cells or 100 to 500 mg of fresh tissue with TRIzol reagent (Invitrogen). After treatment with RNase-free DNase (Invitrogen) for 15 minutes at room temperature, RNA was further purified with the RNeasy RNA isolation kit (Qiagen, Valencia, CA). Total RNA (2 μg) was reverse-transcribed in a 20-μl reaction system using the Superscript first-strand synthesis kit for RT-PCR (Invitrogen) under conditions described by the supplier. Reverse-transcribed cDNA (2 μl) was amplified in 25 μl of PCR reaction system containing 200 μmol/L each dNTP, 20 pmol of each primer, the standard buffer supplemented with 1.5 U Taq polymerase (Roche), and 1.5 mmol/L MgCl2. After initial denaturation at 94°C for 4 minutes, 30 cycles of PCR were performed with denaturation at 94°C for 30 seconds, annealing at 60°C for 30 seconds, and extension at 72°C for 45 seconds. The last extension was at 72°C for 7 minutes. Specific oligonucleotide primers (Table 1) ▶ were synthesized based on published sequences. To avoid false-positive results because of amplification of contaminated genomic DNA in the cDNA preparation, all primers were designed to span two exons separated by an intron.
Table 1.
Primer Sequence
| Primer name | Sequence |
|---|---|
| VEGF outer F | GAA GTC CCA TGA AGT GAT CAA G |
| VEGF outer R | TCA CCG CCT TGG CTT GTC A |
| VEGF common F | GCC AGC ACA TAG AGA GAA TGA GC |
| VEGF120 R | CGG CTT GTC ACA TTTT TCT GG |
| VEGF164 R | CAA GGC TCA CAG TGA TTT TCT GG |
| VEGF188 R | AAC AAG GCT CAC AGT GAA CGC T |
| VEGF probe | ACA GCA GAT GTG AAT GCA GAC CAA AGA AAG |
| GAPDH F | CCT GCA CCA CCA ACT GCT TA |
| GAPDH R | TCA TGA GCC CTT CCA CAA |
| GAPDH probe | CCT GGC CAA GGT CAT CCA C |
| GFP F | AAG AAC GGC ATC AAG GTG AAC T |
| GFP R | ACT GGG TGC TCA GGT AGT GGT T |
| GFP probe | CGT GCA GCT GGC CGA CCA CTA C |
| flt-1 F | ACC TGT CCA ACT ACC TCA AGA GC |
| flt-1 R | CTG GTT CCA GGC TCT CTT TCT T |
| KDR/flk-1 F | CGA CAT AGC CTC CAC TGT TTA TG |
| KDR/flk-1 R | TTT GTT CTT GTT CTC GGT GAT GT |
| Neuropilin-1 F | ATT TGA AGT TTA TGG CTG CAA GA |
| Neuropilin-1 R | ATT GGA TGC TGT AAT CTG GGA GT |
DNA Isolation
Total DNA was isolated from 1 × 106 cultured cells or 100 to 500 mg of fresh tissue lysed in 10 mmol/L of Tris/HCl (pH 7.4) containing 0.2% Triton X-100 and 15 mmol/L of ethylenediaminetetraacetic acid. Lysates were treated with 50 μg of proteinase K overnight at 50°C and 10 μg of RNase for 1 hour at 37°C. DNA was gently extracted with phenol/chloroform.
Real-Time TaqMan PCR
The VEGF isoform and GFP was quantified by real-time PCR on the ABI Prism 7700 Sequence Detection System (Applied Biosystems, Foster City, CA). PCR was performed using TaqMan PCR Core Reagents (Applied Biosystems) according to the manufacturer’s instructions. PCR cycles consisted of initial denaturation at 95°C for 10 minutes, followed by 40 cycles of at 95°C for 15 seconds and at 60°C for 60 seconds. PCR amplification of the housekeeping gene, mouse glyceraldehyde-3-phosphate dehydrogenase (GAPDH), was performed for each sample as control for sample loading and to allow normalization among samples. A standard curve was constructed with PCR-II TOPO cloning vector (Invitrogen) containing the same inserted fragment and amplified by the TaqMan system. The relative expression units in each sample were calculated with respect to the standard calibration curve. Each sample was run twice and each PCR experiment included two nontemplate control wells. PCR products were confirmed as single bands using gel electrophoresis.
Western Blotting
Cultured cells (8 × 106) were lysed in 1 ml of lysis buffer containing 50 mmol/L Tris-HCl (pH 7.4), 150 mmol/L NaCl, 1% Triton X-100, 10 mmol/L 4-(2-aminoethly)benzensulfonyl fluoride hydrochloride (AEBSF), 8 μmol/L aprotinin, 0.22 mmol/L leupeptin, 0.4 mmol/L bestatin, 0.15 mmol/L pepstatin A, and 0.14 mmol/L E-64. Protein was separated by 12% sodium dodecyl sulfate-polyacrylamide gel electrophoresis under denaturing conditions and transferred to nitrocellulose membrane. Nonspecific binding was blocked by overnight incubation at 4°C in 0.1 mol/L of PBS (pH 7.4) containing 3% bovine serum albumin and 0.1% Tween-20. Membranes were then incubated with an anti-VEGF goat polyclonal antibody (C-20; 2 hours, 37°C, 1:200 dilution; Santa Cruz Biotechnologies, Santa Cruz, CA), followed by incubation in horse anti-goat secondary antibody conjugated with horseradish peroxidase (1 hour, room temperature, 1:5000). Immunoreactive proteins were visualized using the enhanced chemiluminescence detection system (Amersham Biosciences, Piscataway, NJ).
Enzyme-Linked Immunosorbent Assay
Capture enzyme-linked immunosorbent assay was performed using anti-mouse VEGF antibody (BAF493, R&D Systems) as capture antibody and anti-VEGF164 biotinylated antibody (AF-493-NA, R&D Systems) as detection antibody in the concentrations described by the manufacturer. The reaction plate was revealed by the 2,2′-aziro-bis-(3-ethylbenzothiazoline-g-sulfonic acid) diammonium salt (ABTS) detection system (Roche) after streptavidin-horseradish peroxidase (Pharmingen) incubation. Optical densities were read at 405 nm and VEGF concentrations were determined by comparison with standard curves generated with recombinant mouse VEGF164 (R&D Systems). Media from exponentially growing cultures were collected every 24 hours. The rate of VEGF secretion was calculated as previously described. 46
Immunostaining
Immunohistochemical staining was performed using the avidin-biotin-peroxidase method. Sections were in cold acetone for 10 minutes, pretreated with 3% H2O2 for 20 minutes to block endogenous peroxidase activity and incubated in matched normal sera (Vector Laboratories, Burlingame, CA). Rat anti-mouse CD31 (Pharmingen) was diluted at 1:200. The Vectastain ABC kit was applied as described by the manufacturer (Vector Laboratories). Sections were counterstained with Gill’s hematoxylin (Vector Laboratories). Images were acquired through Cool SNAP Pro color digital camera (Media Cybernetics). CD31-staining density was analyzed using Image-Pro Plus 4.1 software (Media Cybernetics). For immunofluorescent staining, sections were sequentially incubated in 5% normal serum, anti-mouse CD31 antibody (1:100), biotin-labeled anti-rat immunoglobulin (Ig)G, and avidin-Texas Red (Vector Laboratories). Sections were counterstained with propidium iodide before being inspected under the fluorescent microscope.
Apoptosis Assays
DNA Ladder Assay
DNA was extracted as above, separated by 1.2% agarose gel electrophoresis, and visualized with ethidium bromide staining.
Annexin-V Assay
Annexin-V staining was detected by flow cytometry using an apoptosis detection kit (R&D Systems). Both floating and adherent cells were collected and processed as recommended by the manufacturer. After 15 minutes of incubation with annexin-V-biotin at room temperature, cells were resuspended and incubated in binding buffer containing 4 μg/ml of streptavidin Red 670 (Invitrogen) for 15 minutes. Cells were analyzed using a FACScan flow cytometer (Becton Dickinson). For annexin-V cytochemistry, cells cultured on glass coverslips were incubated in annexin-V-biotin for 15 minutes at room temperature, incubated in binding buffer containing streptavidin-Texas Red (Vector Laboratories) for 15 minutes, washed with PBS, and immediately analyzed under the fluorescent microscope.
In Situ Terminal dUTP Nick-End Labeling (TUNEL) Assay
The ApopTag peroxidase in situ detection kit (Intergen, Purchase, NY) was used to visualize apoptotic cells in vivo and in vitro. The procedure was performed according to the manufacture’s instructions. Briefly, cells cultured on glass coverslips or tumor tissue sections were fixed with 1% paraformaldehyde in PBS, followed by cold ethanol and acetic acid after fixation. After incubation with residues of digoxigenin nucleotide and terminal deoxynucleotide transferase for 1 hour at 37°C, cells were incubated with peroxidase-labeled anti-digoxigenin antibody. DNA fragments were visualized with diaminobenzidine and H2O2.
ELISPOT Assay
Generation of Tumor-Pulsed DCs
DC precursor cells were procured through bone marrow flushing of hind legs from 6-week-old healthy female C57BL6 mice, rinsed once, and plated in RPMI media under standard conditions in the presence of recombinant murine granulocyte-macrophage colony-stimulating factor (GM-CSF) (20 ng/ml; Peprotech, Rocky Hill, NJ) for 8 days. 47 Differentiation into immature DCs was assessed by flow cytometry detection of specific DC marker expression including Cd11c, MHC-II, and CD86. 47 VEGF/GFP-positive ID8 cells were rinsed twice in PBS to eliminate fetal bovine serum xenoantigens, cultured in serum-free media overnight, and then exposed to UVB rays (1500 μW/cm2) to induce apoptosis as described earlier and 12 hours later were co-incubated with immature DCs at a 1:1 ratio (tumor cells:DCs) for 48 hours. Tumor necrosis factor-α (50 U/ml, Preprotech) was added for 3 days. DCs were harvested, rinsed, and counted by trypan blue exclusion. Control DCs included unpulsed DCs matured with tumor necrosis factor-α as above; DCs pulsed as above with autologous murine splenocytes cultured on serum-free media overnight and exposed to UVB to induce apoptosis.
Isolation of Splenic T Cells
To determine the frequency of peripheral tumor-reactive T cells, T cells were isolated from splenocytes procured from tumor-naïve nonvaccinated mice as well as tumor-vaccinated or mock-vaccinated mice bearing flank tumors. Animals were vaccinated with apoptotic tumor cells or mock-vaccinated with PBS (control) as described above and subsequently challenged with flank injections of live tumor cells. Eight weeks after injection of live tumor, mice were euthanized and spleens were resected and minced in a sterile manner to yield a single cell suspension. Splenocytes were also obtained from control age-matched healthy female mice. Erythrocytes were eliminated by hypotonic shock. Splenocytes were plated in culture dishes in RPMI media under standard conditions for 30 minutes and a 95% pure population of T cells (as assessed by flow cytometry) was isolated by collecting the nonadherent fraction.
Interferon (IFN)-γ ELISPOT Assays
For ELISPOT, 107 autologous nonadherent T cells were cultured with tumor-pulsed DCs prepared as above at a 10:1 ratio in RPMI medium supplemented with 3% mouse serum. Control DCs and live tumor cells were also used as controls. Plates (MultiScreen-IP, Millipore, Bedford, MA) were coated overnight at 4°C with 50 μl/well of monoclonal anti-mouse IFN-γ (Pharmingen) at 1 μg/ml in sodium carbonate buffer (2.93 mg/ml sodium bicarbonate, 1.59 mg/ml sodium carbonate, 0.2 mg/ml sodium azide in distilled water). Plates were washed three times in sterile PBS and blocked with RPMI 3% mouse serum for 1 hour at room temperature. T cells generated as above were washed three times in RPMI, resuspended in RPMI 3% mouse serum at 4 × 105 T cells/ml with DCs at a ratio of 10:1 (T cell:DC) and plated in triplicate at 100 μl/well. After 20 hours of co-incubation in standard culture conditions, cells were removed by washing with PBST (PBS, 0.1% Tween-20). Anti-mouse IFN-γ biotinylated monoclonal antibody (2 μg/ml, Pharmingen) was added to each well for 2 hours in PBS containing 0.5% mouse serum and 0.1% Tween-20. After additional washing, streptavidin-alkaline phosphatase at 1:10,000 dilution in PBS was added and incubated for 1 hour at room temperature. After washing, diaminobenzidine was added for 20 to 30 minutes at room temperature. The reaction was stopped by immersion in distilled water. Spots were scanned and counted by computer-assisted ELISPOT image analysis (Hitech Instruments, Edgemont, PA). Digitized images were analyzed for the presence of areas in which color density, spot size, and circularity exceeded background by a factor set on the basis of the comparison of control wells.
Statistical Analysis
Data statistical analysis was performed using SPSS statistics software package (SPSS, Chicago, IL). All of the results are expressed as mean ± SD, and P < 0.05 was used for significance.
Results
Stable VEGF164 Overexpression in ID8 cells
The murine VEGF164 cDNA was successfully inserted in the murine stem cell retrovirus backbone upstream of enhanced GFP, from which it was separated by an internal ribosome entry site, ensuring the transcription of two separate products. After 24 hours of incubation with MigR1 vector carrying VEGF plus GFP or GFP alone, BOSC23 supernatants containing retrovirus were harvested and immediately used to infect ID8 cell monolayers. More than 15% GFP-positive cells were detected by flow cytometry analysis after two passages (Figure 1, A and B) ▶ . Cell populations with high GFP expression were sorted by fluorescence-activated cell sorting from cultures transfected with VEGF/GFP-positive or control GFP-positive retrovirus. The purity of each population was examined immediately by flow cytometry and was revealed to be more than 99.7% (Figure 1A) ▶ .
Figure 1.

Stable overexpression of VEGF164 in ID8 cells after retroviral transfection and sorting. ID8 cells were transfected with retrovirus containing GFP or GFP plus VEGF164. A: Top: flow cytometry analysis of GFP expression of cells transfected with VEGF164/GFP-positive retrovirus during sorting. Approximately 7.5% of the cells express low-medium levels of GFP (gate R2, red), whereas ∼7% express high levels of GFP (gate R3, blue). Bottom: Flow cytometry assessment of the purity of cells sorted based on high GFP expression. The purity of the population is >99.7% B: Top: GFP-positive ID8 cells are observed 72 hours after transfection under a fluorescent microscope. Bottom: After sorting, most cells express high levels of GFP. C: Total intracellular VEGF protein level analyzed by Western blotting after 20 passages in vitro. Intracellular VEGF protein level in cells transfected with VEGF/GFP-positive retrovirus is threefold higher compared to control cells. D: Assessment of the stability of GFP expression by flow cytometry. More than 90% of transfected cells stably express GFP after 20 passages (red, control nontransfected cells; blue, VEGF/GFP transfected cells). E: Quantification of VEGF164 expression by real-time quantitative RT-PCR. Cells transfected with GFP/VEGF-positive retrovirus and sorted based on high expression of GFP (GFP/VEGF) express 18-fold higher levels of VEGF164 mRNA (left) and eightfold higher levels of total VEGF mRNA (right) compared to wild-type control ID8 cells (WT) or ID8 cells transfected with GFP-positive retrovirus (GFP).
Total intracellular VEGF protein was assessed by Western blotting. A specific band was detected in all cell populations examined. When antibody was preincubated with recombinant murine VEGF, no band was detected (not shown). Total intracellular VEGF protein level was threefold higher in VEGF/GFP-transfected cells compared to wild-type or GFP-transfected ID8 cells by Western blot (Figure 1C) ▶ , whereas secretion of VEGF protein in culture media was 12-fold higher by enzyme-linked immunosorbent assay (Table 2) ▶ . Flow cytometry analysis proved that GFP was stably expressed in more than 90% of cells transfected with GFP or VEGF/GFP retrovirus after 20 passages (Figure 1D) ▶ . VEGF164 and total VEGF mRNA levels were more than 11-fold and 4.5-fold higher, respectively, by real-time quantitative RT-PCR in VEGF/GFP-transfected cells compared to wild-type cells or control cells transfected with GFP only (Figure 1E) ▶ .
Table 2.
Summary of VEGF Protein Expression Levels by Enzyme-Linked Immunosorbent Assay
| GFP | VEGF/GFP | |
|---|---|---|
| Conditioned medium | 34 ± 16 pg/(106 cells × hour) | 408 ± 45 pg/(106 cells × hour) |
| Serum | 52 ± 6 (pg/ml) | 135 ± 26 (pg/ml) |
| Ascites | 245 ± 58 (pg/ml) | 3172 ± 230 (pg/ml) |
| Solid tumor | 4 ± 3 (pg/mg) | 112 ± 36 (pg/mg) |
VEGF164 Overexpression Dramatically Accelerates Ascites Formation and Tumor Growth in Vivo
Animals inoculated intraperitoneally with VEGF/GFP-positive ID8 cells, displayed diffuse peritoneal carcinomatosis consisting of multiple tumor nodules of 1 to 10 mm, which were dispersed on the parietal and visceral surfaces of the peritoneal cavity at 8 weeks. Resembling human ovarian carcinoma, tumor nodules were particularly prevalent in the diaphragmatic peritoneum, the porta hepatis, and the pelvis (not shown). Control animals injected intraperitoneally with GFP-transfected or wild-type ID8 cells displayed occasional nodules <2 mm on the diaphragmatic peritoneum and porta hepatis at 8 weeks. Resembling human ovarian carcinoma, animals inoculated intraperitoneally with ID8 cells formed cellular ascites, which in late stages of disease became hemorrhagic. Ascites accumulation was markedly higher in mice bearing VEGF/GFP-transfected intraperitoneal tumors (10 to 12 ml) compared to mice bearing GFP-transfected tumors (1 to 3 ml) 8 weeks after intraperitoneal inoculation (Figure 2A) ▶ . Furthermore, resembling human malignant ascites associated with ovarian carcinoma, ∼33% cells isolated from ascites were CD45+ leukocytes (data not shown). Animals bearing VEGF/GFP intraperitoneal tumors exhibited 12.9-fold higher ascites levels and 2.6-fold higher serum levels of VEGF compared to animals bearing control GFP tumors 2 weeks after inoculation of cells (Table 2) ▶ . After intraperitoneal inoculation of 1 × 107 cells, animals injected with VEGF/GFP-positive cells displayed a median survival of 8 weeks, whereas control animals injected intraperitoneally with GFP-transfected or wild-type cells displayed a median survival of 16 weeks (P < 0.05) (Figure 2B) ▶ .
Figure 2.

VEGF164 overexpression in tumor dramatically accelerates ascites formation and tumor growth in vivo. A: Ascites accumulation is markedly higher in C57BL6 mice inoculated intraperitoneally with 1 × 107 ID8 cells transfected with VEGF/GFP-positive retrovirus (right) compared to mice inoculated with 1 × 107 ID8 cells transfected with GFP-positive retrovirus (left) 8 weeks after tumor inoculation. B: Animals inoculated with ID8 cells transfected with VEGF/GFP-positive cells display a median survival of 8 weeks, whereas control animals injected intraperitoneally with GFP-transfected cells display a median survival of 14.8 weeks (P < 0.05, n = 10/group). C: VEGF/GFP-transfected ID8 cells give rise to markedly larger flank tumors (left flank) compared to control GFP-transfected ID8 cells injected to the contralateral (right) flank. D: Comparison of contralateral tumors resected from three mice. Left: Tumors from GFP-transfected cells growing in the right flanks. Right: Tumors from VEGF/GFP-transfected cells growing in the left flanks. E: Growth of flank tumors after injection of VEGF/GFP-transfected cells (VEGF) and contralateral control GFP-transfected cells (control). Cells were suspended in Matrigel (n = 7/group; *, P < 0.05; **, P < 0.01). F: Stable overexpression of VEGF164 in vivo is demonstrated by RT-PCR. Results from three mice bearing flank tumors are shown. In each mouse, the left RNA sample is from the tumor generated with GFP-transfected, whereas the right sample is from the contralateral tumor generated with VEGF/GFP-transfected tumor. The rightmost lane represents RNA from cultured VEGF/GFP-transfected ID8 cells (positive control). VEGF isoforms 188, 164, and 120 were amplified with isoform-specific primers. Only VEGF164 is overexpressed in vivo. β-actin RNA documents equal amount of RNA used for all samples. G: Real-time quantitative RT-PCR confirms stable overexpression of total VEGF mRNA in vivo. Tumors formed by VEGF/GFP-transfected cells (VEGF) display fourfold higher total VEGF mRNA levels compared to contralateral control tumors formed by GFP-transfected cells (control). Data were normalized with the housekeeping gene GAPDH.
In the flank model, VEGF/GFP-transfected ID8 cells were injected subcutaneously into one flank, whereas the same number of control GFP-transfected ID8 cells (n = 7/group) or wild-type ID8 cells (n = 7) were injected to the other flank in the presence of Matrigel. The tumor volume of VEGF/GFP-transfected cells was significantly larger (0.587 ± 0.083 cm3) compared to control contralateral GFP-transfected cells (0.033 ± 0.01 cm3, P < 0.01) 5 weeks after inoculation (Figure 2 ▶ ; C to E). Wild-type ID8 cells yielded similar tumors to GFP-transfected cells (not shown). Cell injection without Matrigel led to initially slower flank tumor growth, but similarly significant differences were noted between tumors formed by VEGF/GFP-transfected cells and contralateral control GFP-transfected cells (not shown). To confirm the stable in vivo expression of VEGF164, we examined the mRNA level of total VEGF in the tumor tissue by both RT-PCR and real-time RT-PCR (Figure 2, F and G) ▶ . Tumors formed by VEGF/GFP-transfected cells displayed approximately fivefold higher mRNA levels (relative expression units 194.7 ± 34.0) compared to contralateral control tumors formed by GFP-transfected cells (37.2 ± 11.4, P < 0.05).
To eliminate possible interactions between tumors with different VEGF expression growing in opposite flanks of the same animal, animals were inoculated with only one type of tumor cells in one flank (n = 7/group). Identical results were obtained as above: VEGF/GFP tumors grew at a dramatically faster rate compared to control GFP tumors. The volume of VEGF/GFP-positive tumors was significantly larger (0.862 ± 0.252 cm3) compared to control GFP-positive tumors (0.046 ± 0.016 cm3, P < 0.01) 5 weeks after inoculation.
GFP Expression Is Stable in Vivo and Provides a Sensitive Tool to Monitor Tumor Growth and Metastasis
In both the flank and intraperitoneal model, GFP or VEGF/GFP-transfected tumors were clearly visible under a fluorescent stereo microscope (Figure 3) ▶ . The borders between the tumor and normal tissue could be easily observed owing to the distribution of GFP fluoresce. Furthermore, the nonluminous tumor-associated blood vessels were clearly observed against the fluorescent background of GFP-expressing tumors under the fluorescent stereomicroscope. Prominent vessels were readily seen in VEGF-overexpressing tumors (Figure 3, B and D) ▶ . Notably, in the intraperitoneal model, early metastatic tumors, which were very difficult to identify grossly or under normal light stereomicroscope because of their considerably small size and random distribution, could be readily detected under epifluorescence owing to GFP expression (Figure 3, G and H) ▶ . To test the use of GFP in detecting extraperitoneal metastasis, mice were sacrificed 10 weeks after inoculation of flank tumors and the presence of metastatic tumor in the lungs was examined by fluorescent stereo microscopy. Tumor metastasis to lungs was observed in one of seven mice in the VEGF164/GFP group, but in no mice in the GFP group. To detect microscopically invisible metastasis, Genomic GFP was quantified by real-time PCR in DNA extracted from four normal tissues (ie, lung, liver, kidney, and heart). GFP gene was detected in lung (five of seven), liver (two of seven7), and kidney (one of seven) in animals inoculated with VEGF164/GFP ID8 cells, whereas it was only detected in lung (three of seven) of animals inoculated with control GFP tumors (Figure 3I) ▶ .
Figure 3.
GFP expression is stable in vivo and provides a sensitive tool to monitor tumor growth and metastasis. Stable expression of enhanced GFP in vivo allows for rapid identification of tumors in both flank and intraperitoneal models. A-F: Flank tumors resected from both sides of the same mouse. The borders between the tumor and normal tissue can be easily observed owing to the distribution of GFP fluorescence. Furthermore, the nonluminous tumor-associated blood vessels are clearly observed against the fluorescence of the GFP-expressing tumors under the fluorescent stereomicroscope. In control tumors from GFP-transfected ID8 cells (A and B), few blood vessels grow into the tumor; whereas in tumors from VEGF/GFP-transfected ID8 cells (C and D), prominent blood vessels growing into the tumor from nearby normal tissue are observed. Tumor from GFP-transfected ID8 cells under light microscope (E) and fluorescence stereomicroscope (F). A large central necrosis area is observed. Note the absence of prominent vessels in comparison with (D). Areas of necrosis were absent in tumors from VEGF/GFP-transfected ID8 cells. G and H: In the intraperitoneal tumor model, microscopic tumor nodules are detected on the spleen by stereomicroscopy. GFP expression allows for accurate detection of tumors. I: Real-time PCR revealed the presence of GFP gene, the genomic tumor marker, in multiple normal tissues of mice bearing VEGF164/GFP flank tumors, whereas in control mice, GFP was only detected in the lung.
Overexpression of VEGF164 Is Associated with Enhanced Angiogenesis and Increased Survival of Tumor Cells in Vivo
We analyzed angiogenesis in vivo by microvascular density (MVD). Tumors grown from VEGF/GFP-transfected cells were associated with more prominent microvasculature (Figure 3, A to D ▶ , and Figure 4 ▶ ) compared to GFP-transfected tumors. Quantitative analysis of CD31-staining density revealed that in tumors generated by VEGF/GFP-transfected cells, MVD was significantly higher (5.63 ± 0.76) compared to control tumors (1.35 ± 0.45) (P < 0.05) (Figure 4; A, B, and D) ▶ . Large necrotic areas were observed in GFP-positive tumors (n = 5/7, 71.5%) by stereomicroscopy (Figure 3, E and F) ▶ and histology (Figure 5, A and B) ▶ , whereas no necrosis was observed in VEGF/GFP-transfected tumors (n = 0/7, 0%). The prevalence of tumor cell apoptosis was compared in tumors generated with GFP-transfected cells and contralateral tumors generated with VEGF/GFP-transfected ID8 cells by in situ TUNEL assay (Figure 5) ▶ . An inverse association was seen between MVD and areas of necrosis in control tumors (Figure 5, A and B) ▶ . Only scattered apoptotic cells were detected in tumors overexpressing VEGF164 (Figure 5H) ▶ , whereas in control GFP-transfected tumors, a significantly higher number of apoptotic cells was detected both proximal (Figure 5F) ▶ as well as distant to necrotic areas (Figure 5G) ▶ .
Figure 4.

Overexpression of VEGF164 is associated with enhanced angiogenesis in vivo. A and B: Immunohistochemical detection of CD31 (brown) in flank tumors, followed by hematoxylin counterstaining. MVD calculated by the expression of CD31 was dramatically lower in control tumors generated with GFP-transfected cells (A, control) compared to tumors generated with VEGF/GFP-transfected cells (B, VEGF). C: Immunofluorescent staining of CD31 (red) shows prominent vascularization in association with VEGF/GFP-positive tumor cells (green). Nuclei are counterstained by 4,6-diamidiino-2-phenylindole (DAPI) (blue). Most of CD31+ microvasculature is confined within the tumor (GFP+/DAPI+) and does not extend into the GFP−/DAPI+ surrounding stroma. D: Quantitative analysis of CD31 staining density by image analysis. Tumors generated by VEGF/GFP-transfected cells (VEGF) show significantly higher density of CD31+ cells than control tumors (control, P < 0.05).
Figure 5.

VEGF164 overexpression promotes the survival of tumor cells in vivo. A pair of flank tumors from the same mouse is analyzed for detection of apoptosis in vivo with the TUNEL assay. MVD is detected by CD31 immunofluorescent staining. A and B: Tumor generated with GFP-transfected cells analyzed by TUNEL and hematoxylin counterstaining (A) and CD31 (B). A large necrotic area can be seen A, in which decreased MVD is appreciated with CD31 staining (B). Insets F and G are magnified below. C-E: Tumor generated with VEGF/GFP-transfected cells analyzed by CD31 (C) shows dramatically more pronounced MVD in the area occupied by tumor cells, as assessed by detection of GFP (D) and DAPI (E). F and G: Magnification of insets F and G from A. TUNEL assay reveals marked apoptosis in tumor cells in proximity to the necrotic area (F) as well as distal to the necrotic site (G) in control tumor generated with GFP-transfected cells. Tissue is counterstained with hematoxylin. H: Fewer TUNEL-positive apoptotic cells are detected in contralateral tumor generated with GFP/VEGF-transfected cells.
VEGF164 Promotes the Survival of Murine Ovarian Cancer Cells Through an Autocrine/Paracrine Mechanism
We examined the expression of VEGF receptors flt-1, KDR/flk-1, and co-receptor neuropilin-1 by RT-PCR. We readily detected neuropilin-1 but not flt-1 or KDR/flk-1 in ID8 cells by this method. To enhance the sensitivity of RT-PCR, we used nested PCR to detect the expression of VEGF receptors. We found that flt-1 was expressed at low levels in ID8 cells, but KDR/flk-1 was still undetectable (Figure 6) ▶ . KDR/flk-1 and flt-1 were detected in whole tumor RNA, which was used as positive control.
Figure 6.
VEGF receptor flt-1 but not flk-1/KDR is expressed in ID8 cells. Expression of VEGF receptors is assessed by nested RT-PCR in wild-type ID8 cells (WT) and ID8 cells transfected with VEGF/GFP-positive retrovirus (GFP/VEGF). In ID8 cells, VEGFR1/flt-1 is detected at low levels, whereas VEGFR2/flk-1/KDR is not detectable. The VEGF164-specific co-receptor neuropilin-1 is detected at high levels in ID8 cells. Transfection of VEGF164 did not change the expression levels of the above receptors. As positive control, flt-1 and KDR/flk-1 are detected in whole tumor RNA.
The abundance of different VEGF isoform transcripts was examined by RT-PCR. After starvation of serum and insulin for 18 hours, mRNA levels of VEGF188, VEGF164, and VEGF120 were significantly reduced in control wild-type and GFP-transfected cells, whereas cells transfected with VEGF/GFP retrovirus exhibited suppressed VEGF188 and VEGF120 and constitutively elevated levels of VEGF164. The addition of recombinant murine VEGF did not alter the expression of endogenous VEGF (not shown). Growth factor withdrawal induced marked increase in apoptosis in control ID8 cells as well as ID8 cells transfected with GFP-positive retrovirus compared to growth factor-supplemented standard culture conditions (<3%, not shown). However, cells overexpressing VEGF164 displayed twofold to threefold lower amount of apoptosis under conditions of growth factor deprivation (10 ± 2%) compared to ID8 cells transfected with GFP-positive retrovirus (29 ± 3%) or control ID8 cells (22 ± 7%, P < 0.05), as assessed by annexin-V staining (Figure 7, A and B) ▶ . To assess whether the observed effect on apoptosis was because of an autocrine/paracrine effect of VEGF or to genetic alterations induced in ID8 cells by retroviral insertional mutagenesis, 48 several VEGF/GFP-transfected subclones were tested under these conditions and were found to display significantly increased resistance to growth factor deprivation-induced apoptosis compared to control cells (not shown). Furthermore, control GFP-transfected cells or wild-type ID8 cells were exposed to serum and insulin deprivation in the presence or absence of recombinant murine VEGF. A threefold reduction in apoptosis was observed in the presence of exogenous VEGF (P < 0.05, not shown). These results indicate that VEGF inhibits apoptosis in ID8 ovarian cancer cells directly through an autocrine/paracrine mechanism. Interestingly, no apoptotic cells were found expressing GFP, in agreement with a recent report that GFP expression is lost in cells undergoing apoptosis. 49
Figure 7.
VEGF overexpression promotes survival of ID8 cells in vitro and confers resistance to cis-platin-induced apoptosis in ID8 cells. A: Detection of apoptosis by flow cytometry analysis of annexin-V staining. Top: Control. Markedly fewer ID8 cells transfected with VEGF/GFP-positive retrovirus (GFP/VEGF) exhibit annexin-V staining (apoptosis) after growth factor withdrawal compared to control wild-type ID8 cells (WT) or ID8 cells transfected with GFP-positive retrovirus (GFP). Bottom: cis-platin. Markedly fewer ID8 cells transfected with VEGF/GFP-positive retrovirus exhibit apoptosis after exposure to 50 μmol/L of cis-platin for 24 hours compared to control wild-type ID8 cells or ID8 cells transfected with GFP-positive retrovirus. B: Summary of flow cytometry data from three different experiments. ID8 cells transfected with VEGF/GFP-positive retrovirus exhibit significantly less apoptosis after growth factor withdrawal (left) or exposure to cis-platin (right) compared to control wild-type ID8 cells (WT) or ID8 cells transfected with GFP-positive retrovirus (GFP; *, P < 0.05). C: In situ TUNEL and annexin-V apoptosis assay. Top: Control. In situ TUNEL assay detects apoptosis in control WT or GFP ID8 cells cultured under growth factor withdrawal conditions. Apoptotic TUNEL-positive cells are also detected in VEGF/GFP-positive cells. Middle: cis-platin. In situ TUNEL assay detects apoptosis in control WT or GFP ID8 cells exposed to 50 μmol/L of cis-platin for 24 hours. Note markedly increased prevalence of apoptosis and decreased number of cells after exposure to cis-platin compared to cells grown under growth factor deprivation (above). Apoptosis and cell number reduction is markedly less prominent in VEGF/GFP-positive cells. Bottom: cis-platin. In situ fluorescent annexin-V assay using biotinylated annexin-V followed by streptavidin-Red 670 staining. Apoptotic annexin-V-positive cells (red) are noted among control WT ID8 cells and GFP+ ID8 cells (green) transfected with GFP-positive or VEGF/GFP-positive retrovirus.
Overexpression of VEGF164 Protects Murine Ovarian Cancer Cells from Apoptosis Induced by cis-Platin
GFP-transfected and VEGF/GFP-transfected ID8 cells were exposed to increasing doses of cis-platin for increasing periods of time under serum and insulin-free conditions. cis-Platin was found to induce apoptosis in ID8 cells in a dose- and time-dependent manner (data not shown). Under insulin and serum starvation, up to 41 ± 5% of GFP-transfected cells (wild-type ID8, 36 ± 4%) underwent apoptosis when exposed to 50 μmol/L of cis-platin for 24 hours, whereas only 14 ± 2% of VEGF/GFP-transfected cells underwent apoptosis with cis-platin treatment (P < 0.05) (Figure 7, A and B) ▶ . Similar results were obtained with in situ TUNEL assay and in situ fluorescent annexin V staining (Figure 7C) ▶ . Furthermore, a marked reduction in DNA laddering was observed in VEGF/GFP-positive cells after exposure to 50 μmol/L of cis-platin for 24 hours compared to wild-type or GFP-transfected cells (Figure 8) ▶ . Several VEGF/GFP-transfected subclones were tested under these conditions and were found to display significantly increased resistance to platinum-induced apoptosis compared to control cells (not shown). Furthermore, control GFP-transfected cells or parental ID8 cells were exposed to cis-platin in the presence or absence of recombinant murine VEGF (Figure 9) ▶ . Exogenous VEGF conferred partial resistance to apoptosis induced by cis-platin in ID8 cells, with an approximate 25% reduction in the prevalence of apoptotic cells compared to cells exposed to cis-platin in the absence of VEGF (P < 0.05) (Figure 9, A and B) ▶ .
Figure 8.
VEGF overexpression reduces cis-platin-induced apoptosis of ID8 cells in vitro. DNA laddering analysis demonstrates that ID8 cells transfected with VEGF/GFP-positive retrovirus exhibit markedly less DNA fragmentation after exposure to cis-platin compared to control wild-type ID8 cells (WT) or ID8 cells transfected with GFP-positive retrovirus (GFP). M1 and M2 are two molecular markers.
Figure 9.
Exogenous VEGF partially reduces cis-platin-induced apoptosis in ID8 cells in vitro. A: Detection of apoptosis by flow cytometry analysis of annexin-V staining. Addition of cis-platin markedly increases apoptosis in wild-type ID8 cells compared to control cells cultured under serum-free, insulin-free conditions. Addition of recombinant murine VEGF partially reduces cis-platin-induced apoptosis. B: Summary of flow cytometry data from three different experiments. Addition of recombinant murine VEGF induces a significant reduction in apoptosis after exposure of cells to cis-platin.
The Syngeneic Mouse Model of Ovarian Carcinoma Overexpressing VEGF Is Suitable for Immunological Studies
We examined the expression of major complex histocompatibility molecules by ID8 cells or cells transfected by GFP/VEGF or GFP-positive retrovirus. Approximately 50% of cells expressed surface MHC class I molecules (Figure 10A) ▶ , whereas no cells expressed MHC class II molecules (not shown). Similar expression of MHC class I and II was observed in cells transfected by GFP/VEGF or GFP-positive retrovirus (not shown). ELISPOT analysis was performed to quantify the frequency of peripheral tumor-reactive T cells. In the absence of tumor vaccination, control animals exhibited no evidence of tumor-reactive T cells compared to healthy tumor-naïve nonvaccinated C57BL6 female mice of matched age. To generate tumor antigen for in vivo vaccination, ID8 cells transfected by GFP/VEGF-positive retrovirus were irradiated with UVB rays at increasing energy. Approximately 99% of cells were apoptotic by annexin-V and propidium iodide staining when exposed to UVB (not shown). Mice were immunized with two subcutaneous injections of apo-ptotic tumor cells or saline and subsequently challenged with subcutaneous inoculations of live tumor cells. Both animal groups developed flank tumors. A minimal, nonsignificant reduction in tumor volume was noted in immunized compared to control animals 8 weeks after inoculation of flank tumors (not shown). Remarkably, a significant increase in the frequency of tumor-reactive T cells secreting IFN-γ was noted after tumor vaccination in these animals compared to control mice (P < 0.05; Figure 10, B and C ▶ ).
Figure 10.
Genetically engineered tumors may be recognized by the adaptive immune system. A: Flow cytometry detection of major histocompatibility complex class-I (MHC-I) molecule expression in VEGF/GFP-positive ID8 cells. Approximately 50% of cells exhibit expression of surface MHC class-I molecules (red). White: isotype-negative control. B and C: ELISPOT analysis of tumor-reactive T cells in spleens of tumor-naïve nonvaccinated mice (ctrl) as well as mock-vaccinated (nonvaccinated, NV) or tumor-vaccinated (V) mice bearing flank tumors. B: Summary of three different experiments. A threefold increase in the frequency of tumor-reactive T cells secreting IFN-γ is noted in vaccinated animals (V) compared to control mice (*, P < 0.05). C: Depiction of one representative well per condition in one representative experiment. In the absence of tumor vaccination, control animals (NV) exhibit no evidence of tumor-reactive T cells compared to healthy tumor-naïve nonvaccinated C57BL6 female mice of matched age (ctrl). Marked increase in the number of spots staining for IFN-γ is noted, representing clones of antigen-specific (tumor-reactive) T cells recognizing tumor antigen presented by autologous DCs.
Discussion
VEGF may exert multifaceted functions on tumor cells, angiogenesis, and host immune mechanisms that may not only affect the natural course of ovarian carcinoma but also modify its response to therapy. Although such interactions may be partly studied in xenograft models, syngeneic models are best suited to investigate these events. In this study, we developed a syngeneic model of ovarian carcinoma with stable overexpression of murine VEGF164 in the C57BL6 mouse. The rationale for choosing isoform VEGF164 was based on the secretory nature of this isoform 7 and the evidence that VEGF164 is primarily responsible for the angiogenic effects of VEGF in tumors. 10,11 The model that was generated exhibits marked similarities with human ovarian carcinoma. ID8 cells were originally developed from murine ovarian surface epithelium 43 and therefore represent the epithelial ovarian lineage, a true murine surrogate of human epithelial ovarian carcinoma. Intraperitoneal inoculation of genetically modified ID8 cells yielded peritoneal carcinomatosis that closely resembled stage III human ovarian carcinoma (the most frequent form of disease) with widespread nodules on the parietal and visceral peritoneum. In addition, genetically modified tumors were associated with malignant ascites that contained leukocytes and tumor cells.
VEGF expression in tumor cells may be up-regulated by hypoxic conditions or glucose deprivation via hypoxia-inducible factor. 6,50 On the other hand, genetic alterations such as loss of p53, p73 alterations, or overexpression of src may induce constitutive overexpression of VEGF in tumors. 51-53 Expression of VEGF may vary among ovarian carcinomas, and in fact, several human ovarian carcinoma cell lines constitutively exhibit elevated VEGF expression even under standard oxygen and glucose conditions in vitro (unpublished observations from our laboratory). Our model used genetically modified tumor cells with constitutively elevated expression of VEGF and control tumor cells. In the former, overexpression of VEGF was stable in vivo and resulted in markedly elevated levels of VEGF protein in ascites and moderately elevated serum levels compared to animals bearing control tumors. In the latter, VEGF mRNA levels were similar to those detected in normal tissues with pronounced vascularity such as kidney, liver, and the heart. 6 The serum or ascites content of VEGF detected with the two tumor types falls within the range of VEGF protein levels reported in serum (or ascites from patients with ovarian carcinoma. 38,41,54
Increased serum and/or tumor levels of VEGF have been associated with poor clinical outcome. 16,41,42 The animal model presented in this study provides a suitable tool to dissect the molecular mechanisms underlying the effects of VEGF. As expected, tumors overexpressing VEGF164 exhibited significantly higher MVD compared to controls and significantly decreased tumor cell apoptosis in vivo, in agreement with others. 55,56 Overexpression of VEGF164 significantly accelerated tumor growth and ascites formation, resulting in significantly shorter median survival. These findings prove the validity of the present model as one of angiogenesis-dependent ovarian carcinoma in which to investigate the biological effects of VEGF. Our findings are in contrast with the results recently reported with a xenograft model using human ovarian cancer cells overexpressing human VEGF165 in immunodeficient mice 57 and underscore the significance of experimentation in a syngeneic setting.
Tumor cells are faced with significant proapoptotic insults in vivo including hypoxia, acidosis, substrate starvation, growth factor deprivation, and immune-mediated attack, which may be partly mediated through apoptosis. 58 In addition, a variety of chemotherapy agents rely on apoptosis for their pharmacological cytotoxic effect. 59 Overexpression of VEGF164 not only increased angiogenesis, but also directly supported tumor cell survival through an autocrine/paracrine mechanism and conferred resistance to apoptosis induced by growth factor deprivation or chemotherapy. The present data taken together suggest that VEGF represents an important adaptation of tumor cells to adverse conditions within the tumor microenvironment, not only promoting nutrient supply through angiogenesis but also protecting tumor cells from the proapoptotic tumor environment. Our results are in agreement with observations that VEGF protects cells against apoptosis including that induced by ionizing radiation or chemotherapeutic drugs. 55,60 In leukemia cells, VEGF acts via KDR/flk-1 and downstream PI3-K, Akt kinase, and nitric oxide to prevent apoptosis. 29 Select human ovarian cancer cells may express KDR/flk-1, 22 but we were unable to detect KDR/flk-1 in murine ID8 cells. In endothelial cells, KDR/flk-1 seems to be primarily responsible for the mitogenic effects of VEGF, whereas flt-1 may function as a decoy receptor. 61 However, flt-1 is able to interact with a variety of signal transduction proteins, including the p85 subunit of PI3-K and mitogen-activated protein kinase, generating mitogenic signals. In fact, flt-1 was recently shown to participate in intracellular autocrine regulatory loops mediating the survival of hematopoietic stem cells. 62 Furthermore, Flt-1 exhibits a significantly higher affinity for VEGF165 compared to KDR/flk-1, 61 suggesting that even if expressed at low levels, Flt-1 may still be functional. Although there is yet no evidence for direct signaling mediated by neuropilins, a recent report indicates that in breast carcinoma cells lacking KDR/flk-1 but expressing neuropilin-1, VEGF induced activation of the PI3-K pathway. 63 Based on the above information, we hypothesize that VEGF164 regulates, via flt-1 and/or possibly neuropilins, signaling pathways such as PI3-K to support ID8 cell survival in an autocrine/paracrine manner.
An increasing bulk of evidence suggests that VEGF exerts important immunological functions. VEGF has been shown to inhibit the differentiation and maturation of DCs in vitro and in vivo, 33,34,64,65 inhibiting the development of anti-tumor T cell responses. Furthermore, VEGF may also suppress cytokine-induced leukocyte-endothelial interactions in vivo 66 or decrease leukocyte transendothelial migration. 67 The immune biology of ovarian carcinoma has not been adequately investigated partly because of the lack of suitable syngeneic animal models. The present model fills this gap, because it is suitable for immunological studies related to ovarian cancer biology and therapy, and lends itself to investigation of the immunological effects of VEGF in cancer. Similarly to human ovarian carcinoma, genetically engineered ID8 cells were found to exhibit heterogeneous expression of surface MHC-I molecules. Our findings indicate that insertion of the murine VEGF164 isoform and enhanced GFP via a retrovirus did not significantly alter the immunogenicity of ID8 cells. In fact, in the absence of vaccination, no tumor-specific T cells were detected in mice using the highly sensitive ELISPOT method. These findings are in agreement with a recent report showing that enhanced GFP is not immunogenic in the C57BL6 mouse. 68 After repeated vaccination with apoptotic tumor cells, a significant tumor-specific T cell response was documented that however did not result in significant inhibition of tumor growth. Taken together, these findings suggest that ID8 tumors express antigens that may be recognized by the adaptive immune system if presented at a distant site from the tumor, but in nonimmunized animals the tumors entirely evade immune recognition. Furthermore, tumors evade immune attack by tumor-specific T cells after vaccination. These findings closely resemble the immunological behavior of human ovarian carcinoma in which tumor-reactive T cells are documented among peripheral lymphocytes in patients with advanced disease. 69
An additional advantage offered by the present model relates to the expression of GFP. This facilitates rapid detection of tumor cells by fluorescent microscopy in histological specimens or by flow cytometry in analysis of cell suspensions. Furthermore, it allows for the sensitive detection of tumor cells in vivo using live fluorescent stereo microscopy. The molecular mechanisms underlying ovarian cancer extraovarian spread and intraperitoneal or retroperitoneal lymph node metastasis have been poorly elucidated, partly because of the lack of a suitable animal model. Successful orthotopic injection of tumor cells has been reported in mouse ovary. 70,71 Our model combined with orthotopic injection of tumor cells offers opportunities for the investigation of early mechanisms of ovarian cancer intraperitoneal spread in the immunocompetent host and evaluation of the role of VEGF in this process. Furthermore, besides VEGF, basic fibroblast growth factor, interleukin-8, and transforming growth factor-β have been implicated in tumor angiogenesis and have been detected at high levels in ovarian cancer. 72,73 Genetic manipulation of ID8 cells inserting additional or alternate proangiogenic factors has the potential to shed light on their individual function and possible synergistic interactions in promoting angiogenesis and progression of ovarian carcinoma in the immunocompetent host.
In summary, we present the development of a syngeneic mouse model of ovarian carcinoma with stable overexpression of murine VEGF164. The growth of these tumors was proven to be angiogenesis-dependent. This model provides a useful tool for the study of the multifaceted functions of VEGF on tumor cells, angiogenesis, and anti-tumor immune mechanisms. Furthermore, it offers a suitable tool for the investigation of the impact of VEGF on the efficacy of therapeutic strategies in ovarian carcinoma. Because of the expression of GFP, this model also offers new opportunities for the investigation of the molecular mechanisms underlying ovarian cancer spread in the immunocompetent host.
Acknowledgments
We thank Dr. Paul F. Terranova (University of Kansas) for donating the murine ID8 cells, Dr. Warren Pear (University of Pennsylvania) for donating the MigR1 vector and BOSC23 packaging cell line, and Dr. Patricia D’Amore (Harvard University) for donating the VEGF164 cDNA.
Footnotes
Address reprint requests to George Coukos, M.D., Ph.D., Center for Research on Reproduction and Women’s Health, University of Pennsylvania, 1355 BRB II/III, 421 Curie Blvd., Philadelphia, PA 19104. E-mail: gcoukos@mail.obgyn.upenn.edu.
Supported by grants from the Gynecologic Cancer Foundation, the Berlex Foundation, the University of Pennsylvania Abramson Family Cancer Research Institute, the National Cancer Institute Specialized Program of Research Excellence Grant CA 83638, and National Institutes of Health Grant D43 TW00671 funded by the Fogarty International Center, and the National Institute of Child Health and Human Development (F.B.).
References
- 1.Carmeliet P, Jain RK: Angiogenesis in cancer and other diseases. Nature 2000, 407:249-257 [DOI] [PubMed] [Google Scholar]
- 2.Yancopoulos GD, Davis S, Gale NW, Rudge JS, Wiegand SJ, Holash J: Vascular-specific growth factors and blood vessel formation. Nature 2000, 407:242-248 [DOI] [PubMed] [Google Scholar]
- 3.Hanahan D, Weinberg RA: The hallmarks of cancer. Cell 2000, 100:57-70 [DOI] [PubMed] [Google Scholar]
- 4.Robinson CJ, Stringer SE: The splice variants of vascular endothelial growth factor (VEGF) and their receptors. J Cell Sci 2001, 114:853-865 [DOI] [PubMed] [Google Scholar]
- 5.Shima DT, Kuroki M, Deutsch U, Ng YS, Adamis AP, D’Amore PA: The mouse gene for vascular endothelial growth factor. Genomic structure, definition of the transcriptional unit, and characterization of transcriptional and post-transcriptional regulatory sequences. J Biol Chem 1996, 271:3877-3883 [DOI] [PubMed] [Google Scholar]
- 6.Zhang L, Conejo-Garcia JR, Yang N, Huang W, Mohamed-Hadley A, Yao W, Benencia F, Coukos G: Different effects of glucose starvation on expression and stability of VEGF mRNA isoforms in murine ovarian cancer cells. Biochem Biophys Res Commun 2002, 292:860-868 [DOI] [PubMed] [Google Scholar]
- 7.Park JE, Keller GA, Ferrara N: The vascular endothelial growth factor (VEGF) isoforms: differential deposition into the subepithelial extracellular matrix and bioactivity of extracellular matrix-bound VEGF. Mol Biol Cell 1993, 4:1317-1326 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Stalmans I, Ng YS, Rohan R, Fruttiger M, Bouche A, Yuce A, Fujisawa H, Hermans B, Shani M, Jansen S, Hicklin D, Anderson DJ, Gardiner T, Hammes HP, Moons L, Dewerchin M, Collen D, Carmeliet P, D’Amore PA: Arteriolar and venular patterning in retinas of mice selectively expressing VEGF isoforms. J Clin Invest 2002, 109:327-336 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ng YS, Rohan R, Sunday ME, Demello DE, D’Amore PA: Differential expression of VEGF isoforms in mouse during development and in the adult. Dev Dyn 2001, 220:112-121 [DOI] [PubMed] [Google Scholar]
- 10.Grunstein J, Masbad JJ, Hickey R, Giordano F, Johnson RS: Isoforms of vascular endothelial growth factor act in a coordinate fashion to recruit and expand tumor vasculature. Mol Cell Biol 2000, 20:7282-7291 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yu JL, Rak JW, Klement G, Kerbel RS: Vascular endothelial growth factor isoform expression as a determinant of blood vessel patterning in human melanoma xenografts. Cancer Res 2002, 62:1838-1846 [PubMed] [Google Scholar]
- 12.de Vries C, Escobedo JA, Ueno H, Houck K, Ferrara N, Williams LT: The fms-like tyrosine kinase, a receptor for vascular endothelial growth factor. Science 1992, 255:989-991 [DOI] [PubMed] [Google Scholar]
- 13.Terman BI, Dougher-Vermazen M, Carrion ME, Dimitrov D, Armellino DC, Gospodarowicz D, Bohlen P: Identification of the KDR tyrosine kinase as a receptor for vascular endothelial cell growth factor. Biochem Biophys Res Commun 1992, 187:1579-1586 [DOI] [PubMed] [Google Scholar]
- 14.Gluzman-Poltorak Z, Cohen T, Herzog Y, Neufeld G: Neuropilin-2 is a receptor for the vascular endothelial growth factor (VEGF) forms VEGF-145 and VEGF-165. J Biol Chem 2000, 275:18040-18045 [DOI] [PubMed] [Google Scholar]
- 15.Soker S, Takashima S, Miao HQ, Neufeld G, Klagsbrun M: Neuropilin-1 is expressed by endothelial and tumor cells as an isoform-specific receptor for vascular endothelial growth factor. Cell 1998, 92:735-745 [DOI] [PubMed] [Google Scholar]
- 16.Shen GH, Ghazizadeh M, Kawanami O, Shimizu H, Jin E, Araki T, Sugisaki Y: Prognostic significance of vascular endothelial growth factor expression in human ovarian carcinoma. Br J Cancer 2000, 83:196-203 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Abulafia O, Triest WE, Sherer DM: Angiogenesis in malignancies of the female genital tract. Gynecol Oncol 1999, 72:220-231 [DOI] [PubMed] [Google Scholar]
- 18.Junker K: Prognostic factors in stage I/II non-small cell lung cancer. Lung Cancer 2001, 33(Suppl 1):S17-S24 [DOI] [PubMed] [Google Scholar]
- 19.Gasparini G: Prognostic value of vascular endothelial growth factor in breast cancer. Oncologist 2000, 5(Suppl 1):S37-S44 [DOI] [PubMed] [Google Scholar]
- 20.Chung YS, Maeda K, Sowa M: Prognostic value of angiogenesis in gastrointestinal tumours. Eur J Cancer 1996, 32A:2501-2505 [DOI] [PubMed] [Google Scholar]
- 21.Tomisawa M, Tokunaga T, Oshika Y, Tsuchida T, Fukushima Y, Sato H, Kijima H, Yamazaki H, Ueyama Y, Tamaoki N, Nakamura M: Expression pattern of vascular endothelial growth factor isoform is closely correlated with tumour stage and vascularisation in renal cell carcinoma. Eur J Cancer 1999, 35:133-137 [DOI] [PubMed] [Google Scholar]
- 22.Boocock CA, Charnock-Jones DS, Sharkey AM, McLaren J, Barker PJ, Wright KA, Twentyman PR, Smith SK: Expression of vascular endothelial growth factor and its receptors flt and KDR in ovarian carcinoma. J Natl Cancer Inst 1995, 87:506-516 [DOI] [PubMed] [Google Scholar]
- 23.Speirs V, Atkin SL: Production of VEGF and expression of the VEGF receptors Flt-1 and KDR in primary cultures of epithelial and stromal cells derived from breast tumours. Br J Cancer 1999, 80:898-903 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Tian X, Song S, Wu J, Meng L, Dong Z, Shou C: Vascular endothelial growth factor: acting as an autocrine growth factor for human gastric adenocarcinoma cell MGC803. Biochem Biophys Res Commun 2001, 286:505-512 [DOI] [PubMed] [Google Scholar]
- 25.Itakura J, Ishiwata T, Shen B, Kornmann M, Korc M: Concomitant over-expression of vascular endothelial growth factor and its receptors in pancreatic cancer. Int J Cancer 2000, 85:27-34 [DOI] [PubMed] [Google Scholar]
- 26.Liu B, Earl HM, Baban D, Shoaibi M, Fabra A, Kerr DJ, Seymour LW: Melanoma cell lines express VEGF receptor KDR and respond to exogenously added VEGF. Biochem Biophys Res Commun 1995, 217:721-727 [DOI] [PubMed] [Google Scholar]
- 27.Langer I, Vertongen P, Perret J, Fontaine J, Atassi G, Robberecht P: Expression of vascular endothelial growth factor (VEGF) and VEGF receptors in human neuroblastomas. Med Pediatr Oncol 2000, 34:386-393 [DOI] [PubMed] [Google Scholar]
- 28.Dias S, Hattori K, Heissig B, Zhu Z, Wu Y, Witte L, Hicklin DJ, Tateno M, Bohlen P, Moore MA, Rafii S: Inhibition of both paracrine and autocrine VEGF/VEGFR-2 signaling pathways is essential to induce long-term remission of xenotransplanted human leukemias. Proc Natl Acad Sci USA 2001, 98:10857-10862 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Koistinen P, Siitonen T, Mantymaa P, Saily M, Kinnula V, Savolainen ER, Soini Y: Regulation of the acute myeloid leukemia cell line OCI/AML-2 by endothelial nitric oxide synthase under the control of a vascular endothelial growth factor signaling system. Leukemia 2001, 15:1433-1441 [DOI] [PubMed] [Google Scholar]
- 30.Herold-Mende C, Steiner HH, Andl T, Riede D, Buttler A, Reisser C, Fusenig NE, Mueller MM: Expression and functional significance of vascular endothelial growth factor receptors in human tumor cells. Lab Invest 1999, 79:1573-1582 [PubMed] [Google Scholar]
- 31.Masood R, Cai J, Zheng T, Smith DL, Hinton DR, Gill PS: Vascular endothelial growth factor (VEGF) is an autocrine growth factor for VEGF receptor-positive human tumors. Blood 2001, 98:1904-1913 [DOI] [PubMed] [Google Scholar]
- 32.Dias S, Hattori K, Zhu Z, Heissig B, Choy M, Lane W, Wu Y, Chadburn A, Hyjek E, Gill M, Hicklin DJ, Witte L, Moore MA, Rafii S: Autocrine stimulation of VEGFR-2 activates human leukemic cell growth and migration. J Clin Invest 2000, 106:511-521 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Gabrilovich DI, Chen HL, Girgis KR, Cunningham HT, Meny GM, Nadaf S, Kavanaugh D, Carbone DP: Production of vascular endothelial growth factor by human tumors inhibits the functional maturation of dendritic cells. Nat Med 1996, 2:1096-1103 [DOI] [PubMed] [Google Scholar]
- 34.Gabrilovich D, Ishida T, Oyama T, Ran S, Kravtsov V, Nadaf S, Carbone DP: Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo. Blood 1998, 92:4150-4166 [PubMed] [Google Scholar]
- 35.Greenlee RT, Hill-Harmon MB, Murray T, Thun M: Cancer statistics, 2001. CA Cancer J Clin 2001, 51:15-36 [DOI] [PubMed] [Google Scholar]
- 36.Ogawa S, Kaku T, Kobayashi H, Hirakawa T, Ohishi Y, Kinukawa N, Nakano H: Prognostic significance of microvessel density, vascular cuffing and vascular endothelial growth factor expression in ovarian carcinoma: a special review for clear cell adenocarcinoma. Cancer Lett 2002, 176:111-118 [DOI] [PubMed] [Google Scholar]
- 37.Brustmann H, Naude S: Vascular endothelial growth factor expression in serous ovarian carcinoma: relationship with high mitotic activity and high FIGO stage. Gynecol Oncol 2002, 84:47-52 [DOI] [PubMed] [Google Scholar]
- 38.Santin AD, Hermonat PL, Ravaggi A, Cannon MJ, Pecorelli S, Parham GP: Secretion of vascular endothelial growth factor in ovarian cancer. Eur J Gynaecol Oncol 1999, 20:177-181 [PubMed] [Google Scholar]
- 39.Boss EA, Massuger LF, Thomas CM, Geurts-Moespot A, Boonstra H, Sweep CG: Vascular endothelial growth factor in ovarian cyst fluid. Cancer 2001, 91:371-377 [DOI] [PubMed] [Google Scholar]
- 40.Mesiano S, Ferrara N, Jaffe RB: Role of vascular endothelial growth factor in ovarian cancer: inhibition of ascites formation by immunoneutralization. Am J Pathol 1998, 153:1249-1256 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Chen CA, Cheng WF, Lee CN, Chen TM, Kung CC, Hsieh FJ, Hsieh CY: Serum vascular endothelial growth factor in epithelial ovarian neoplasms: correlation with patient survival. Gynecol Oncol 1999, 74:235-240 [DOI] [PubMed] [Google Scholar]
- 42.Hartenbach EM, Olson TA, Goswitz JJ, Mohanraj D, Twiggs LB, Carson LF, Ramakrishnan S: Vascular endothelial growth factor (VEGF) expression and survival in human epithelial ovarian carcinomas. Cancer Lett 1997, 121:169-175 [DOI] [PubMed] [Google Scholar]
- 43.Roby KF, Taylor CC, Sweetwood JP, Cheng Y, Pace JL, Tawfik O, Persons DL, Smith PG, Terranova PF: Development of a syngeneic mouse model for events related to ovarian cancer. Carcinogenesis 2000, 21:585-591 [DOI] [PubMed] [Google Scholar]
- 44.Pear WS, Nolan GP, Scott ML, Baltimore D: Production of high-titer helper-free retroviruses by transient transfection. Proc Natl Acad Sci USA 1993, 90:8392-8396 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Tomayko MM, Reynolds CP: Determination of subcutaneous tumor size in athymic (nude) mice. Cancer Chemother Pharmacol 1989, 24:148-154 [DOI] [PubMed] [Google Scholar]
- 46.Graff BA, Bjornaes I, Rofstad EK: Microvascular permeability of human melanoma xenografts to macromolecules: relationships to tumor volumetric growth rate, tumor angiogenesis, and VEGF expression. Microvasc Res 2001, 61:187-198 [DOI] [PubMed] [Google Scholar]
- 47.Lutz MB, Kukutsch N, Ogilvie AL, Rossner S, Koch F, Romani N, Schuler G: An advanced culture method for generating large quantities of highly pure dendritic cells from mouse bone marrow. J Immunol Methods 1999, 223:77-92 [DOI] [PubMed] [Google Scholar]
- 48.Powell SK, Kaloss M, Burimski I, Weaver L, Long Z, Lyons R, McGarrity GJ, Otto E: In vitro analysis of transformation potential associated with retroviral vector insertions. Hum Gene Ther 1999, 10:2123-2132 [DOI] [PubMed] [Google Scholar]
- 49.Steff AM, Fortin M, Arguin C, Hugo P: Detection of a decrease in green fluorescent protein fluorescence for the monitoring of cell death: an assay amenable to high-throughput screening technologies. Cytometry 2001, 45:237-243 [DOI] [PubMed] [Google Scholar]
- 50.Maxwell PH, Pugh CW, Ratcliffe PJ: Activation of the HIF pathway in cancer. Curr Opin Genet Dev 2001, 11:293-299 [DOI] [PubMed] [Google Scholar]
- 51.Vikhanskaya F, Bani MR, Borsotti P, Ghilardi C, Ceruti R, Ghisleni G, Marabese M, Giavazzi R, Broggini M, Taraboletti G: p73 overexpression increases VEGF and reduces thrombospondin-1 production: implications for tumor angiogenesis. Oncogene 2001, 20:7293-7300 [DOI] [PubMed] [Google Scholar]
- 52.Ravi R, Mookerjee B, Bhujwalla ZM, Sutter CH, Artemov D, Zeng Q, Dillehay LE, Madan A, Semenza GL, Bedi A: Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1alpha. Genes Dev 2000, 14:34-44 [PMC free article] [PubMed] [Google Scholar]
- 53.Wiener JR, Nakano K, Kruzelock RP, Bucana CD, Bast RC, Jr, Gallick GE: Decreased Src tyrosine kinase activity inhibits malignant human ovarian cancer tumor growth in a nude mouse model. Clin Cancer Res 1999, 5:2164-2170 [PubMed] [Google Scholar]
- 54.Kraft A, Weindel K, Ochs A, Marth C, Zmija J, Schumacher P, Unger C, Marme D, Gastl G: Vascular endothelial growth factor in the sera and effusions of patients with malignant and nonmalignant disease. Cancer 1999, 85:178-187 [PubMed] [Google Scholar]
- 55.Grunstein J, Roberts WG, Mathieu-Costello O, Hanahan D, Johnson RS: Tumor-derived expression of vascular endothelial growth factor is a critical factor in tumor expansion and vascular function. Cancer Res 1999, 59:1592-1598 [PubMed] [Google Scholar]
- 56.Harris SR, Schoeffner DJ, Yoshiji H, Thorgeirsson UP: Tumor growth enhancing effects of vascular endothelial growth factor are associated with increased nitric oxide synthase activity and inhibition of apoptosis in human breast carcinoma xenografts. Cancer Lett 2002, 179:95-101 [DOI] [PubMed] [Google Scholar]
- 57.Duyndam MC, Hilhorst MC, Schluper HM, Verheul HM, van Diest PJ, Kraal G, Pinedo HM, Boven E: Vascular endothelial growth factor-165 overexpression stimulates angiogenesis and induces cyst formation and macrophage infiltration in human ovarian cancer xenografts. Am J Pathol 2002, 160:537-548 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Kagi D, Ledermann B, Burki K, Zinkernagel RM, Hengartner H: Molecular mechanisms of lymphocyte-mediated cytotoxicity and their role in immunological protection and pathogenesis in vivo. Annu Rev Immunol 1996, 14:207-232 [DOI] [PubMed] [Google Scholar]
- 59.Coukos G, Rubin SC: Chemotherapy resistance in ovarian cancer: new molecular perspectives. Obstet Gynecol 1998, 91:783-792 [DOI] [PubMed] [Google Scholar]
- 60.Katoh O, Takahashi T, Oguri T, Kuramoto K, Mihara K, Kobayashi M, Hirata S, Watanabe H: Vascular endothelial growth factor inhibits apoptotic death in hematopoietic cells after exposure to chemotherapeutic drugs by inducing MCL1 acting as an antiapoptotic factor. Cancer Res 1998, 58:5565-5569 [PubMed] [Google Scholar]
- 61.Ferrara N: Role of vascular endothelial growth factor in regulation of physiological angiogenesis. Am J Physiol 2001, 280:C1358-C1366 [DOI] [PubMed] [Google Scholar]
- 62.Gerber HP, Malik AK, Solar GP, Sherman D, Liang XH, Meng G, Hong K, Marsters JC, Ferrara N: VEGF regulates haematopoietic stem cell survival by an internal autocrine loop mechanism. Nature 2002, 417:954-958 [DOI] [PubMed] [Google Scholar]
- 63.Bachelder RE, Crago A, Chung J, Wendt MA, Shaw LM, Robinson G, Mercurio AM: Vascular endothelial growth factor is an autocrine survival factor for neuropilin-expressing breast carcinoma cells. Cancer Res 2001, 61:5736-5740 [PubMed] [Google Scholar]
- 64.Gabrilovich DI, Ishida T, Nadaf S, Ohm JE, Carbone DP: Antibodies to vascular endothelial growth factor enhance the efficacy of cancer immunotherapy by improving endogenous dendritic cell function. Clin Cancer Res 1999, 5:2963-2970 [PubMed] [Google Scholar]
- 65.Almand B, Resser JR, Lindman B, Nadaf S, Clark JI, Kwon ED, Carbone DP, Gabrilovich DI: Clinical significance of defective dendritic cell differentiation in cancer. Clin Cancer Res 2000, 6:1755-1766 [PubMed] [Google Scholar]
- 66.Tromp SC, oude Egbrink MG, Dings RP, van Velzen S, Slaaf DW, Hillen HF, Tangelder GJ, Reneman RS, Griffioen AW: Tumor angiogenesis factors reduce leukocyte adhesion in vivo. Int Immunol 2000, 12:671-676 [DOI] [PubMed] [Google Scholar]
- 67.Detmar M: Molecular regulation of angiogenesis in the skin. J Invest Dermatol 1996, 106:207-208 [DOI] [PubMed] [Google Scholar]
- 68.Skelton D, Satake N, Kohn DB: The enhanced green fluorescent protein (eGFP) is minimally immunogenic in C57BL/6 mice. Gene Ther 2001, 8:1813-1814 [DOI] [PubMed] [Google Scholar]
- 69.Schlienger K, Chu CS, Woo EY, Rivers P, Toll AJ, Hudson B, Maus MV, Riley JL, Choi Y, Coukos G, Kaiser LR, Rubin SC, Levine BL, Carroll RG, June CH: TRANCE matured dendritic cells generate MHC class-I restricted T cells specific for autologous tumor in late-stage ovarian cancer patients. Clin Cancer Res , (in press) [PubMed]
- 70.Hoffman RM: Visualization of GFP-expressing tumors and metastasis in vivo. Biotechniques 2001, 30:1016-1024 [DOI] [PubMed] [Google Scholar]
- 71.Hoffman RM: Orthotopic transplant mouse models with green fluorescent protein-expressing cancer cells to visualize metastasis and angiogenesis. Cancer Metastasis Rev 1998, 17:271-277 [DOI] [PubMed] [Google Scholar]
- 72.Paley PJ: Angiogenesis in ovarian cancer: molecular pathology and therapeutic strategies. Curr Oncol Rep 2002, 4:165-174 [DOI] [PubMed] [Google Scholar]
- 73.Papetti M, Herman IM: Mechanisms of normal and tumor-derived angiogenesis. Am J Physiol Cell Physiol 2002, 282:C947-C970 [DOI] [PubMed] [Google Scholar]






