(FHIT) Gene Overexpression Tumor Growth by ...

5 downloads 0 Views 1MB Size Report
Kisselev, L. L., Justesen, J., Wolfson, A. D., and Frolova, L. Y. Diadenosine ... Evan, G., and Littlewood, T. A matter of life and cell death. Science (Washington.
Induction of Apoptosis and Inhibition of Tumorigenicity and Tumor Growth by Adenovirus Vector-mediated Fragile Histidine Triad (FHIT) Gene Overexpression Lin Ji, Bingliang Fang, Nancy Yen, et al. Cancer Res 1999;59:3333-3339.

Updated version

Access the most recent version of this article at: http://cancerres.aacrjournals.org/content/59/14/3333

Cited Articles

This article cites by 34 articles, 20 of which you can access for free at: http://cancerres.aacrjournals.org/content/59/14/3333.full.html#ref-list-1

Citing articles

This article has been cited by 54 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/59/14/3333.full.html#related-urls

E-mail alerts Reprints and Subscriptions Permissions

Sign up to receive free email-alerts related to this article or journal. To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at [email protected]. To request permission to re-use all or part of this article, contact the AACR Publications Department at [email protected].

Downloaded from cancerres.aacrjournals.org on June 1, 2013. © 1999 American Association for Cancer Research.

[CANCER RESEARCH 59, 3333–3339, July 15, 1999]

Advances in Brief

Induction of Apoptosis and Inhibition of Tumorigenicity and Tumor Growth by Adenovirus Vector-mediated Fragile Histidine Triad (FHIT) Gene Overexpression1 Lin Ji, Bingliang Fang, Nancy Yen, Kwun Fong, John D. Minna, and Jack A. Roth2 Section of Thoracic Molecular Oncology, Department of Thoracic and Cardiovascular Surgery, The University of Texas, M. D. Anderson Cancer Center, Houston, Texas 77030 [L. J., B. F., N. Y., J. A. R.], and Hamon Center for Therapeutic Oncology Research and Departments of Internal Medicine and Pharmacology, The University of Texas Southwestern Medical Center, Dallas, Texas 75235-8593 [K. F., J. D. M.]

Abstract We studied the effects of fragile histidine triad (FHIT) gene overexpression mediated by an adenoviral vector, Ad-FHIT, on cell proliferation, apoptosis, and cell cycle kinetics in human cancer cells and on tumorigenicity and tumor growth in nude mice. Overexpression of the FHIT gene significantly inhibited cell growth in various Ad-FHIT-transduced human lung cancer cells and head and neck carcinoma cells with FHIT gene abnormalities, but not in normal human bronchial epithelial cells. Fewer than 20% of cells in all Ad-FHIT-transduced cells survived at 7 days after transduction. Overexpression of the FHIT gene induced cell apoptosis and altered cell cycle processes. The apoptotic cell population markedly increased, and cells accumulated in S phase after Ad-FHIT transduction. The tumorigenicity of human H1299 lung cancer cells transduced by Ad-FHIT, in comparison with that of the control transductants and untreated cells, was eliminated in vivo. Subcutaneous tumor growth in nude mice who received intratumoral injections of Ad-FHIT, at a total dose of 3 3 1010 plaque-forming units/tumor for H1299 tumors and 4 3 1010/tumor for A549 tumors, were suppressed by more than 85% and 90%, respectively, compared with that in nude mice who received injections of empty vector at the same dose or with PBS alone. Together, our results suggest that the FHIT gene, when delivered at high efficiency by a recombinant adenoviral vector, functions as a tumor suppressor gene both in vitro and in vivo.

Introduction The FHIT3 gene was isolated by exon trapping and positional cloning (1). Since the discovery of the FHIT gene, several lines of evidence have suggested that it is a tumor suppressor gene. The genomic structure of FHIT overlaps with the FRA3B fragile site and coincides with a genomic region that is known to be frequently involved in allelic loss, genetic rearrangement, and cytogenetic abnormality in a broad range of sporadic human cancers and solid tumors. Although point mutations within the FHIT gene have rarely been reported, genomic alterations such as homozygous deletions of

exons or insertions of intronic sequences and aberrant transcripts of the FHIT gene, as well as the lack of detectable Fhit protein, have all been frequently observed in a variety of primary tumors (including lung, stomach, breast, colon, cervix, prostate, and head and neck cancers) and in their derived cell lines (2–10). However, a variety of observations have also raised questions about whether the FHIT gene is, indeed, a classic tumor suppressor gene (6, 11–13). Experimental evidence is needed to clarify the function of FHIT as a tumor suppressor gene. If loss of FHIT function leads to human cancers, one might predict that replacement of the abnormal FHIT gene with the wild-type gene would result in tumor suppression similar to that shown for the Rb or p53 tumor suppressor genes, including inhibition of cell growth in vitro and suppression of tumorigenicity and tumor growth in vivo. Siprashvili et al. (14) recently showed that replacement of the wild-type FHIT gene in human cancer cell lines that lacked endogenous Fhit expression significantly reduced their tumorigenicity in nude mice. In contrast, Otterson et al. (15) recently reported that introduction of wild-type FHIT into HeLa cells, a cervical carcinoma cell line, did not alter its tumorigenicity in animals. To date, studies of the function of FHIT as a tumor suppressor gene have been performed in tumor cell lines with transient or stable transfection by wild-type FHIT-expressing plasmids. Thus, we decided to use an adenoviral vector as an alternative means for studying the tumor suppressor function of FHIT. We studied the effects of FHIT gene overexpression mediated by recombinant Ad vector (Ad-FHIT) on cell proliferation, apoptosis, and cell cycle kinetics in human lung and head and neck cancer cells with varied status of FHIT gene or gene products and on tumorigenicity and tumor growth in nude mice. Materials and Methods

Cell Culture. The 293 cell line and the transformed 293/GV16 cell line expressing yeast GAL4/VP16 trans-activating proteins (16, 17) were used in the construction, amplification, and titration of adenoviral vectors. Both cell Received 4/7/99; accepted 5/28/99. lines were maintained in DMEM containing 4.5 g/liter of glucose with 10% The costs of publication of this article were defrayed in part by the payment of page FBS. Human lung cancer cells, human head and neck cancer cells, and normal charges. This article must therefore be hereby marked advertisement in accordance with HBECs were used to test the effects of Ad vector-mediated FHIT overexpres18 U.S.C. Section 1734 solely to indicate this fact. 1 This study was partially supported by grants from the National Cancer Institute and sion in vitro. Human lung cancer H1299 and H460 cells were maintained in the NIH, by Specialized Program of Research Excellence (SPORE) in Lung Cancer Grant RPMI 1640 supplemented with 5% FBS, and A549 cells were maintained in P50-CA70907, by The University of Texas M. D. Anderson Cancer Center Support Core Ham’s F-12 nutrient mixture with 10% FBS. Human head and neck cell lines Grant CA16672, by gifts to the Division of Surgery and Anesthesiology from Tenneco and 1483 and UMSCC22B [22B; kindly provided by Dr. Li Mao (The University Exxon for the Core Laboratory Facility, and by an American Society of Clinical Oncology of Texas M. D. Anderson Cancer Center, Houston, TX)] were maintained in (ASCO) Young Investigator Award (to K. F.). 2 To whom requests for reprints should be addressed, at Section of Thoracic Molecular DMEM/F-12 medium with 10% FBS. Normal HBECs were maintained in the Oncology, Department of Thoracic and Cardiovascular Surgery, Box 109, The University medium supplied by the manufacturer (Clonetics Inc., Walkersville, MD) and of Texas M. D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030. according to the instructions. Phone: (713) 792-6935; Fax: (713) 794-4901; E-mail: [email protected]. 3 The abbreviations used are: FHIT, fragile histidine triad; Ad, adenovirus; HBEC, Construction of Recombinant Ads. The recombinant Ad-CMV-FHIThuman bronchial epihelial cell; FBS, fetal bovine serum; CMV, cytomegalovirus; GFP, GAL4 (Ad-FHIT) was constructed using a cosmid-Ad vector system by green fluorescent protein; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium homologous recombination in recA1 bacteria, Escherichia coli. The FHIT bromide; MOI, multiplicity(ies) of infection; TUNEL, terminal deoxynucleotidyl transcDNA (568 bp) was placed in an expression cassette containing a CMV ferase-mediated nick end labeling; PI, propidium iodide; FACS, fluorescence-activated promoter and bovine growth hormone (BGH) poly(A) sequence. The CMVcell sorting; pfu, plaque-forming unit. 3333

Downloaded from cancerres.aacrjournals.org on June 1, 2013. © 1999 American Association for Cancer Research.

FHIT GENE EXPRESSION AND FUNCTION IN VITRO AND IN VIVO

FHIT-BGH poly(A) expression cassette was inserted into the E1-deleted region in an Ad vector containing an inactivated E4 region. A vector expressing GFP, Ad-CMV-GFP (Ad-GFP), was used to monitor transduction efficiency by the viral vectors and as a nonspecific transgene expression control. Ad-E12-GAL4 (Ad-EV), an empty E12 vector containing the same E4 modification as in the Ad-FHIT vector, was used as a negative control. These viral vectors were amplified in either 293 cells or 293/VP16 cells and purified with the conventional CsCl gradient centrifugation procedure, as described previously (16, 17). Viral titers were determined by both absorbance measurement and plaque assay for pfu. Potential contamination of the viral preparation by the wild-type virus was monitored by PCR analysis. Determination of Cell Growth Rate. Inhibition of cell growth by Ad transduction was assayed by MTT staining of viable cells (Sigma Chemical Co., St. Louis, MO). In brief, cells were transduced with adenoviral vectors at varied MOI by directly applying the diluted vectors into the growth medium. The experimentally treated cells were harvested at a designated time, and 200 ml of cell suspension was added to each well in 96-well plates. A one-tenth volume of MTT solution (5 mg MTT/ml PBS) was added to each well, and each plate was incubated for 2– 4 h at 37°C until a purple precipitate was visible. The medium was then carefully removed, and precipitates were dissolved in 100 ml of DMSO. Growth rate was plotted as the percentage of viable cells in PBS-treated controls (a value arbitrarily set at 100%). Each experiment was repeated at least three times with each treatment given in duplicate or triplicate. Data were presented as an average of the results from individual experiments. Western Blot Analysis. Expression of FHIT in Ad-FHIT-transduced cells or untransduced (PBS-treated) controls was analyzed by Western blotting. Cells grown in 6-well plates (1–2 3 106/well) were treated with adenoviral vectors or with PBS alone as a control. Cells were transduced with Ad-FHIT in vitro. At designated time points, cells were harvested by centrifugation for 5 min at 2500 rpm at 4°C. Cells were washed two times with cold PBS and then resuspended in 100 ml of cell lysis solution [20 mM HEPES (pH 7.9), 150 mM NaCl, 0.5 mM DTT, 10 mM KCl, 0.2 mM EDTA, 25% glycerol, and complete proteinase inhibitors (Boehringer Mannheim, Indianapolis, IN)]. Cells were sonicated with two pulses of 30 s each at a setting of 6.0 (MICROSON; Heat Systems Inc., Farmingdale, NY). The sonicated cells were then spun for 10 min at 14,000 rpm at 4°C. Cell lysates were collected and stored at 280°C. Protein concentrations of the cell lysates were determined using Bio-Rad protein assay reagent (Bio-Rad, Fremont, CA). Proteins were then separated by SDS-PAGE. For electrophoresis, each lane was loaded with about 60 mg of crude proteins and run at 100 V for 1–2 h. Proteins were then transferred from gels to Hybond-ECL membranes (Amersham International, Little Chalfont, England). Membranes were blocked in a blocking solution (3% dry milk and 0.1% Tween 20 in PBS) for 1 h at room temperature. Membranes were then incubated with a 1:1000 dilution of primary rabbit antihuman Fhit fusion protein polyclonal antiserum (Zymed Inc., South San Francisco, CA) and a 1:1000 dilution of mouse anti-b-actin monoclonal antibodies (Amersham International). Immunocomplexes were detected with the secondary horseradish peroxidase-labeled rabbit antimouse IgG or goat antirabbit IgG antibodies using an enhanced chemiluminescence kit (Amersham International), according to the manufacturer’s instructions. Apoptosis and Cell Cycle Analysis. Apoptosis was analyzed by FACS using the TUNEL reaction with FITC-labeled dUTP (Boehringer Mannheim Biochemicals, Mannheim, Germany). In brief, cells (1 3 106/well) were seeded on 6-well plates and infected with Ad constructs. PBS containing no viral constructs was used as a control. Cells (including floating cells in the medium) were harvested at designated times and washed twice with PBS. Cells were fixed in 1% formaldehyde for 15 min at 4°C, permeabilized in 500 ml of 70% ethanol, and stored at 4°C. Cells were then washed twice with PBS, resuspended in 50 ml of TUNEL reaction mixture [0.2 M potassium cacodylate, 25 mM Tris-HCl (pH 6.6), 2.5 mM cobalt chloride, 0.25 mg/ml BSA, 100 units/ml terminal deoxynucleotidyltransferase, and 10 mM FITC-labeled dUTP], and incubated for 60 min at 37°C in a humidified atmosphere in the dark. After incubation, cells were washed twice in PBS and resuspended in 500 ml of PI solution (5 mg/ml PI and 10 mg/ml Rnase A in PBS). Cells were analyzed for green FITC, fluorescence through a 520-nm BP filter, and for red (PI), fluorescence through a 620-nm LP filter. Green fluorescence was taken as a marker of DNA fragmentation and apoptosis; red fluorescence, as a marker of DNA content and cell-cycle status.

Animal Studies. All animals were maintained, and all animal experiments were conducted under institutional guidelines established for the Animal Core Facility at M. D. Anderson Cancer Center. Nu/Nu mice were obtained from Charles River (Cambridge, MA). Five mice were used for each treatment group. For the tumorigenicity assay, H1299 cells were transduced in vitro with Ad-FHIT at a MOI of 10, with PBS alone as a mock control and Ad-EV as a negative control. The transduced cells were harvested at 24 h and 48 h after transduction, respectively. Viability of cells was determined by trypan blue exclusion staining. Viable cells (1 3 107) were then injected s.c. into the right flank of female nude mice, 6 – 8 weeks of age. Tumor formation in mice was observed twice or three times weekly up to 3 months. Tumor dimensions were measured every 2 or 3 days by using a linear caliper. The tumor volume was calculated using the equation V(mm3) 5 a 3 b2/2, where a is the largest dimension and b is the perpendicular diameter. To study the effect of FHIT on tumor growth, H1299 cells were used to establish s.c. tumors in nude mice. Briefly, 1 3 107 cells were injected into the right flank of female nude mice, 6 – 8 weeks of age. When the tumors reached 5–10 mm in diameter (at about 2 weeks postinjection), animals received intratumoral injections of Ad-FHIT or control vectors, respectively, for four times at days 1, 4, 8, and 11, at a total dose of 3 3 1010 pfu/tumor. Tumor size was measured and calculated as described above. At the end of the experiment, animals were killed and tumors were excised and processed for pathological and immunohistochemical analysis. Statistics. Both the in vitro and the in vivo results were expressed as mean 6 SD or mean 6 SE. Student’s two-sided t test was used to compare the values of the test and control samples. A value of P , 0.05 was taken as significant.

Results Inhibition of Tumor Cell Growth by Overexpression of FHIT. To test the hypothesis that the FHIT gene functions as a tumor suppressor in vitro, we performed a series of experiments to study the effects of overexpression of the FHIT gene on cell proliferation in various Ad-FHIT-transduced human lung cancer cells, head and neck carcinoma cells, and a normal HBEC line varying in FHIT gene and gene product expression status (Fig. 1). Cells from each line were transduced in vitro by Ad-FHIT vectors administered at various MOI; cells treated with PBS, empty vector Ad-EV, and Ad-GFP were used as controls. The transduction efficiency was determined by examining the GFP-expressing cells in the Ad-GFP-transduced cell population under a fluorescence microscope. The transduction efficiency of the adenoviral vectors was .80% at the highest MOI applied for each cell line. Cell proliferation was analyzed by determining the viability of cells at 1, 2, 5, and 5 days after transduction, respectively. Viability was significantly reduced in Ad-FHIT- transduced H1299 cells that exhibit deletion of exon 5 and a 120-bp insertion in the FHIT gene (Ref. 9; Fig. 1A), A549 cells that exhibit deletion of exon 4 and fusion of exon 3 and exon 5 (Ref. 7; Fig. 1B), H460 cells that exhibit deletions of exons 5, 6, and 7 (Ref. 9; Fig. 1C), and 1483 cells that exhibit deletion of exon 4 (Ref. 18; Fig. 1D). In all cases, the viability of transduced cells was compared with that of untransduced (PBStreated) control cells (the viability of which was set at 100%). Fewer than 20% of cells transduced by Ad-FHIT at the highest MOI survived 5 and 7 days after transduction when compared with the PBS mock– treated controls, whereas the growth of cells transduced by the Ad-EV and Ad-GFP control vectors at the same MOI was only slightly affected. In contrast, adenoviral vector-mediated FHIT expression minimally affected cell growth in 22B cells (Fig. 1E), a head and neck carcinoma cell line containing and expressing the normal FHIT gene. Nearly 80% of cells transduced by Ad-FHIT, at a MOI of 20, were still viable 7 days after transduction. Furthermore, we tested the specificity of the observed inhibitory effects of Fhit overexpression on tumor cell proliferation and the potential cytotoxicity of the overexpressed Fhit by analyzing the proliferation of normal HBECs transduced with Ad-FHIT (Fig. 1F). A

3334

Downloaded from cancerres.aacrjournals.org on June 1, 2013. © 1999 American Association for Cancer Research.

FHIT GENE EXPRESSION AND FUNCTION IN VITRO AND IN VIVO

Fig. 1. Effects of exogenous FHIT on cell growth of human lung cancer, head and neck carcinoma, and HBEC lines. Cells were transduced with adenoviral vectors Ad-FHIT (FHIT) at the MOI indicated (inset), Ad-EV (EV) and Ad-GFP (GFP) at the highest MOI as used for Ad-FHIT in each cell line, and PBS alone as a mock control. The viability of cells was determined by MTT staining. The growth rate was expressed as the percentage of viable adenoviral vector-transduced cells in relation to PBS-treated control cells (100%). Bars, SDs of the mean in three individual experiments. Treatments were given in duplicate for each experiment. The significance of the difference in growth rate between vector-treated cells and PBS-treated controls was analyzed by two-sided Student’s t test. P , 0.05 was taken as significant. The differences between the surviving fractions of the Ad-EV-transduced cells versus PBS-treated controls were not significant (P 5 0.16 to P 5 0.78 from different time points and cell lines). The differences between the cell growth rate of the Ad-FHIT- transduced cells versus the Ad-EV-transduced, Ad-GFP-transduced, or PBS-treated controls at same MOI were significant in H1299, A549, H460, and 1483 cell lines (P 5 0.0001 to P 5 0.002) but, not significant in 22B and HBEC cell lines (P 5 0.10 to P 5 0.95, from different time points and cell lines).

,10% loss of cell viability was observed in Ad-FHIT-transduced cells at various MOI when compared with that in untransduced control cells, and similar losses were also observed in Ad-EV-transduced cells throughout the posttransduction time course. The effects of FHIT on cell proliferation in Ad-FHIT-transduced cells were consistent with the levels of exogenous FHIT gene expression and the status of the endogenous FHIT gene in these cell lines, as demonstrated by Western blot analysis using polyclonal rabbit antihuman Fhit antibodies as probes (Fig. 2). Overexpression of the Fhit protein was detected in all Ad-FHIT-transduced cells, but not in untransduced controls. In addition, the levels of Fhit expression increased with increasing MOI and with days after transduction in all cell lines examined (Fig. 2, Lanes 4 –15). No endogenous expression of Fhit was detected in H1299, A549, H460, and 1483 tumor cells (Fig. 2, A-D, Lanes 1–3), which have been shown to lack the FHIT gene and to form aberrant FHIT transcripts. Endogenous Fhit protein expression was detectable at low levels in both normal FHIT transcript-expressing 22B cells (Fig. 2C) and normal HBEC cells (Fig. 2D). Together, these data indicated that the Ad-FHIT vector efficiently mediated FHIT gene expression in these transduced tumor cells and that the expressed FHIT proteins could inhibit tumor cell proliferation and growth in vitro. Effects of FHIT Overexpression on Apoptosis and Cell Cycle Kinetics. To further investigate the observed inhibitory effects of Fhit overexpression on tumor cell proliferation as it relates to cell death pathways, we examined the ability of exogenous Fhit to induce

apoptosis and its impact on cell cycle kinetics in the Ad-FHITtransduced tumor cells. The effect of Fhit on apoptosis in Ad-FHITtransduced H1299 cells was analyzed by FACS using the TUNEL reaction (Fig. 3A). Induction of apoptosis was detected in Ad-FHITtransduced cells. More than 20% and 35% of cells were apoptotic at days 5 and 7 after transduction with Ad-FHIT, respectively, whereas only about 7% and 10% of cells treated with PBS alone and transduced with Ad-EV vector, respectively, were TUNEL positive at the same time periods. The level of induction of apoptosis in the AdFHIT-transduced cells increased with time after transduction and correlated with the expression of Fhit protein (Fig. 2) and the viability of cells (Fig. 1), suggesting that the inhibition of tumor cell proliferation by Fhit is mediated directly or indirectly through induction of apoptosis. Furthermore, the effect of FHIT gene expression on cell cycle processes was analyzed by FACS using PI staining (Fig. 3B). Interestingly, cell cycle processes in the Ad-FHIT-transduced cells seemed to be significantly affected by overexpression of Fhit at later stages of transduction compared with the untransduced and Ad-EV-transduced controls. Cell cycle profiles indicated that .32% of the H1299 cells transduced with Ad-FHIT were arrested at S phase at 5 days posttransduction (Fig. 3B, I), whereas only 8% of untransduced control cells (Fig. 3B, G) and 9% of Ad-EV-transduced cells (Fig. 3B, H) were arrested at S phase. This phenomenon had not been observed in any previously reported in vitro experiment using FHIT-expressing plasmids.

3335

Downloaded from cancerres.aacrjournals.org on June 1, 2013. © 1999 American Association for Cancer Research.

FHIT GENE EXPRESSION AND FUNCTION IN VITRO AND IN VIVO

recorded from first injection until 20 days after last injection for the H1299 s.c. tumor model (Fig. 5A) and until 24 days after last injection for the A549 tumor model (Fig. 5B), respectively. All of the tumors in the mice treated with Ad-FHIT showed significantly suppressed growth compared with tumors in PBS-treated and Ad-EV-treated mice in both tumor models (P , 0.001; Fig. 5, A and B). At the termination of these experiments, the tumors injected with Ad-FHIT were, on average, only about one-tenth the size of the tumors injected with PBS alone or with Ad-EV vector. Discussion

Fig. 2. Western blot analysis of expression of exogenous Fhit proteins in Ad-transduced cells and of endogenous Fhit proteins in control cells. Cell lines, the recombinant adenoviral vector used to transduce each cell line, and the various MOI used are indicated. Briefly, crude protein extracts were prepared from Ad-FHIT-transduced cells at days 1, 2, 5, and 7 after transduction, as indicated, and from control cells at day 7 after transduction. A total of 40 – 60 mg of crude proteins was loaded onto each lane. Immunocomplexes formed with a mouse antihuman b-actin monoclonal antibody and rabbit anti-Fhit polyclonal antibodies are shown.

Inhibition of Tumorigenicity by Ad-FHIT-transduced Cancer Cells. To further determine whether the observed inhibitory effects of FHIT on tumor cell proliferation in vitro could be demonstrated in vivo, we tested the tumorigenic potential of Ad-FHIT-transduced H1299 cells in nude mice (Fig. 4). Viable H1299 cells (1 3 107/ mouse) transduced with Ad-FHIT for 24 h (Fig. 4A) or 48 h (Fig. 4B) were s.c. injected into five mice in each treatment group. Tumors started to form around 10 days after injection and were observed in all mice who received injections of either untreated or Ad-EV-transduced H1299 cells. Once tumors were established, they grew aggressively, ultimately reaching an average volume of about 0.5– 0.9 cm3 in mice who received injections of untreated cells and 0.4 – 0.7 cm3 in mice who received injections of 24-h and 48-h Ad-EV transductants within 5 weeks after injection, respectively. In contrast, however, no tumors formed in any mice injected with Ad-FHIT-transduced H1299 cells within the same periods of time (Fig. 4, A and B), indicating that the tumorigenicity of Ad-FHIT-transduced H1299 cells was eliminated in vivo. Suppression of Tumor Growth by Intratumoral Injection of Ad-FHIT. The high efficiency of transduction and high level of transgene expression mediated by adenoviral vector in vivo allowed us to study the tumor suppressor function of FHIT in vivo by direct intratumoral injection of Ad-FHIT vectors. The growth of tumors was

We have shown: (a) that adenoviral vector-mediated overexpression of the wild-type FHIT gene efficiently inhibited growth of tumor cells of varying FHIT gene and gene product status in vitro; (b) that the tumorigenicity of the Ad-FHIT-transduced tumor cells was eliminated in vivo; and (c) that tumor growth was significantly suppressed by direct injection of the FHIT-expressing adenoviral vector into s.c. tumors in nude mice. These results provided direct evidence for the biological function of FHIT as a tumor suppressor gene both in vitro and in vivo. The lung cancer cell lines H1299, A549, and H460 and the head and neck carcinoma cell line 1483 all exhibit an altered or inactivated FHIT gene, as shown by reverse transcription-PCR and Northern blot analysis (7, 9, 18), lack endogenous Fhit protein expression, as shown by Western blot analysis, and are highly tumorigenic. Alterations in the FHIT locus have been shown to be correlated with loss or reduction of Fhit protein expression in tumors and tumor cell lines (3, 15, 19, 20). Clonal expansion of Fhit-negative cells has been observed in many primary and cultured tumors (14, 19). These data suggest that the lack of Fhit expression might provide a selective advantage for clonal expansion in vivo and implicate FHIT gene in cancer pathogenesis. Moreover, frequent loss of heterozygosity at the 3p14 region and the absence of Fhit protein expression have been observed in precancerous lesions (19, 21); both have also been associated with the G1 morphological grade and early clinical stage in renal carcinoma (20). Together, these data suggest the inactivation of FHIT in the early phases of cell carcinogenesis and pathogenesis. Our observations that exogenous wild-type FHIT gene replacement inhibited growth in these cancer cell lines provide supporting evidence for the theory that functional inactivation of FHIT participates in cancer pathogenesis and are consistent with a role for FHIT as a tumor suppressor gene. On the other hand, overexpression of exogenous Fhit did not affect growth of head and neck carcinoma 22B cells and normal HBECs, which express both normal FHIT transcripts and Fhit protein. Similar results have also been observed by Siprashvili et al. (14) in Ad type 5-transformed human kidney 293 cells expressing endogenous Fhit protein. A high yield of colonies of FHIT transfectants and, in our case, production of Fhit-expressing recombinant Ads could be achieved in this cell line. These results suggest that exogenous Fhit functions specifically in restoring inactivated endogenous FHIT alleles and that it does not cause generalized toxicity. In this study, we have demonstrated for the first time that the growth inhibition of tumor cells is associated with the induced apoptosis and altered cell cycle processes resulting from overexpression of exogenous Fhit. The decreased cell proliferation is consistent with the increased apoptotic activity and the increased level of exogenous Fhit protein expression in Ad-FHIT-transduced tumor cells, suggesting that the inhibition of tumor cell growth by Fhit is mediated by an apoptotic pathway. Increased apoptosis in association with altered cell proliferation and inhibited cell growth has been described in various systems (22–25). However, the effects on cell proliferation, apoptosis, and cell cycle processes demonstrated by Fhit seem different from

3336

Downloaded from cancerres.aacrjournals.org on June 1, 2013. © 1999 American Association for Cancer Research.

FHIT GENE EXPRESSION AND FUNCTION IN VITRO AND IN VIVO

Fig. 3. Effects of expression of Fhit proteins on apoptosis and cell cycle process in Ad-FHIT-transduced H1299 cells. Apoptosis and cell cycle kinetics were analyzed by FACS, using TUNEL reaction with FITC-labeled dUTP and propidium iodide staining, respectively. A, effects on apoptosis. Cells were transduced with adenoviral vectors at a MOI of 10 and harvested at the indicated days after transduction. The rate of apoptosis is expressed as the percentage of FITC-labeled cells in the total cell population. Bars, SDs of the mean in triplicate treatments and TUNEL reactions. B, representative profiles of cell cycle kinetics. The cells used for the cell cycle analysis were the same ones used for the apoptosis analysis.

3337

Downloaded from cancerres.aacrjournals.org on June 1, 2013. © 1999 American Association for Cancer Research.

FHIT GENE EXPRESSION AND FUNCTION IN VITRO AND IN VIVO

raphy showed that binding nucleotide substrates by Fhit, similar to G proteins, changed the molecular surface of the protein and, thus, suggested that a Fhit-substrate complex might be the active form of Fhit involved in the signaling pathway(s) controlling cell growth (28 –31). Ap4A and Ap3A, which are substrates for the Fhit hydrolase, have emerged as putative intracellular and extracellular signaling molecules involved in mediation of adaptive responses by mammalian cells to environmental stress such as heat, oxidation, and DNA damage (1, 30, 32). Furthermore, it has recently been reported that apoptosis in cultured human cells is associated with alterations in Ap3A and Ap4A levels (33, 34). These data suggest that the loss of FHIT gene function could result in alterations in both the levels of substrates and the status of substrate-binding, which, consequently, could inactivate a pathway that leads to apoptosis and initiate the malignant process by stimulating DNA synthesis and cell proliferation. Discovery of the direct target of Fhit-substrate complexes in the cell proliferation, cell cycle progression, and apoptosis cascade would be expected to provide insights into the molecular mechanisms of Fhit function and could undoubtedly shed light on molecular targets or therapeutic agents for the prevention and treatment of various human cancers associated with the functional inactivation of the FHIT gene.

Fig. 4. Effects of FHIT expression on the tumorigenicity of Ad-FHIT-transduced H1299 cells in nude mice. H1299 cells (1 3 107) transduced with either Ad-FHIT (FHIT) or Ad-EV (EV) at a MOI of 10 for 24 h (A) or 48 h (B) were s.c. injected into the right flank of nude mice. Tumor volumes were calculated using the equation, V (mm3) 5 a 3 b2/2, in which a is the largest dimension and b is the perpendicular diameter. Results were reported as the mean 6 SD in five mice for each treatment group. No tumor was formed at anytime in the course of the experiment in any of the five mice inoculated with Ad-FHIT-transduced cells for either 24 h or 48 h. The differences between the tumor volumes of the Ad-EV-treated mice versus PBS-treated controls were not significant (Student’s t test, P 5 0.24 for 24-h transductants and P 5 0.77 for 48-h transductants, respectively).

those of the tumor suppressor genes p53 and Rb. The cellular responses to the expression of exogenous Fhit protein in terms of both growth inhibition and apoptosis induction are slower than those to the overexpression of exogenous p53, although the magnitude of the final effect is similar (24, 25). In our study, overexpression of Fhit proteins was detected at 24 h after transduction, but the apparent growth inhibition and apoptosis induction were observed first at 3 days and peaked at 4 –5 days after transduction. By comparison, however, the growth inhibition and apoptosis induced by exogenous p53 overexpression became apparent at 24 h and peaked at 72 h after transduction in the same cell lines (17, 25, 26). Moreover, Ad-FHIT-transduced cells accumulate in S phase, whereas p53-overexpressing cells (22, 23) and Rb1-positive cells responding to effectors such as p16 overexpression undergo G0-G1 arrest (27). These differences suggest that an apoptotic pathway mediated by the FHIT gene is different from the pathways mediated by the tumor suppressor genes p53 and Rb. It is likely that FHIT-mediated apoptosis is p53-independent because it can occur in cells homozygously deleted for p53 (H1299). The mechanism of induction of apoptosis and alteration of cell cycle processes by Fhit is not yet well understood, but a few clues are emerging from recent structural and functional studies of Fhit protein and its homologues in the HIT protein superfamily (26). Crystallog-

Fig. 5. Suppression of tumor growth induced by intratumoral injection of Ad-FHIT in nude mice. A, effects on tumor growth in H1299-inoculated mice. Arrows, the schedule of injections. When the tumor reached 5–10 mm in diameter at about 2 weeks after tumor inoculation, the viral vectors were given at doses of 5 3 109 pfu in 50 ml of PBS for the first two injections and 1 3 1010 pfu in 100 ml of PBS for the last two injections, respectively; PBS alone was used as a mock treatment control. Results were reported as the mean 6 SD in five mice for each treatment group. B, effects on tumor growth in A549-inoculated mice. Arrows, the schedule of injections. When the tumor reached 5–10 mm in diameter at about 2 weeks after tumor inoculation, the viral vectors were given at doses of 5 3 109 pfu in 50 ml of PBS for the first two injections and 1 3 1010 pfu in 100 ml of PBS for the last three injections, respectively; PBS alone was used as a mock treatment control. Results were reported as the mean 6 SD in five mice for each treatment group. The differences between the tumor volumes of the Ad-FHIT- treated mice versus the Ad-EV-treated mice or PBS-treated controls were significant in both tumor models (Student’s t test, P 5 0.001 and P 5 0.002 for H1299 tumor model and P 5 0.0003 and P 5 0.001 for the A549 tumor model, respectively).

3338

Downloaded from cancerres.aacrjournals.org on June 1, 2013. © 1999 American Association for Cancer Research.

FHIT GENE EXPRESSION AND FUNCTION IN VITRO AND IN VIVO

In our study, overexpression of FHIT efficiently eliminated the tumorigenicity of the highly tumorigenic cell line H1299 in vivo. Although one might argue that the diminished tumorigenicity of the Ad-FHIT-transduced tumor cells resulted from doomed cell viability, our results, nevertheless, indicate that the inhibitory effects on cell proliferation or cell growth demonstrated by overexpression of FHIT in established cell lines are consistent and cannot be abolished or reversed under normal physiological conditions. Moreover, the gene delivery mediated by the AdFHIT vector is highly efficient in vivo. Expression of Fhit could still be detected in Ad-FHIT-injected tumor cells 3 weeks after the last injection (data not shown). Furthermore, the suppression of tumor growth by exogenous FHIT overexpression is very effective, and the magnitude of tumor-suppressing activity of FHIT is comparable with that observed for p53 (24, 25, 35). The transfer of exogenous wild-type p53 into cancer cells carrying mutated p53 leads to a strong induction of apoptosis and growth arrest, but is less effective in cells expressing endogenous p53. The FHIT gene, however, seems to use an alternative apoptosis pathway to perform its tumor-suppressing functions and shows an equally effective growth inhibition in both p53-deleted H1299 and wild-type p53containing A549 lung cancer cells. Thus, gene therapy combining both p53 and FHIT tumor suppressor genes would presumably be more effective because different apoptotic pathways would be activated. On the other hand, the results of previous experiments using tumor cell lines either transiently or stably transfected by wild-type FHITexpressing plasmids for studying biological function of FHIT in vitro and in vivo were variable, depending on cell lines studied (14, 15). This suggests that some cancer cell lines may be more susceptible to physiological level of FHIT expression. However, high levels of expression in cancer cells seem to consistently induce apoptosis. Although the mechanism regulating the differential sensitivity of the FHIT-transduced cells to FHIT-mediated apoptosis is not known, the accumulation of growth-deregulating mutations may sensitize transduced cells to proapoptotic stimuli (36). Ad-FHIT vector has several advantages over plasmids for FHIT gene delivery and for studying the biological functions of Fhit in vitro and in vivo. First, a high efficiency of transduction (.80%) and a high level of FHIT gene expression could be easily achieved in a wide spectrum of cell types by simply adjusting the MOI of viral particles to target cells. Second, the Ad-FHIT-transduced tumor cells could be directly studied for their effect on tumorigenicity in animals without having to select for stably transduced colonies; thus, problems associated with the colony selection process and with unknown effectors or factors generated in resulting cell colonies could be avoided. Third, the Ad-FHIT vector could be directly used to evaluate the role of FHIT as a tumor suppressor gene in vivo by either i.v. or intratumoral injection of the vector into animals. Finally, the Ad-FHIT vector, by being both highly efficient at tumor suppression in vivo and reduced in toxicity and immunogenicity by virtue of E1-E3 deletion and E4 inactivation in the adenoviral vector genome (16, 17), could be very useful in cancer gene therapy. Acknowledgments We thank Dr. Kay Huebner for review of the manuscript.

References 1. Ohta, M., Inoue, H., Cotticelli, M. G., Kastury, K., Baffa, R., Palazzo, J., Siprashvili, Z., Mori, M., McCue, P., Druck, T., Croce, C. M., and Huebner, K. The FHIT gene, spanning the chromosome 3p14.2 fragile site and renal carcinoma-associated t(3;8) breakpoint, is abnormal in digestive tract cancers. Cell, 84: 587–597, 1996. 2. Druck, T., Hadaczek, P., Fu, T. B., Ohta, M., Siprashvili, Z., Baffa, R., Negrini, M., Kastury, K., Veronese, M. L., Rosen, D., Rothstein, J., McCue, P., Cotticelli, M. G., Inoue, H., Croce, C. M., and Huebner, K. Structure and expression of the human FHIT gene in normal and tumor cells. Cancer Res., 57: 504 –512, 1997. 3. Greenspan, D. L., Connolly, D. C., Wu, R., Lei, R. Y., Vogelstein, J. T., Kim, Y. T., Mok, J. E., Munoz, N., Bosch, F. X., Shah, K., and Cho, K. R. Loss of FHIT expression in cervical carcinoma cell lines and primary tumors. Cancer Res., 57: 4692– 4698, 1997.

4. Huebner, K., Hadaczek, P., Siprashvili, Z., Druck, T., and Croce, C. M. The FHIT gene, a multiple tumor suppressor gene encompassing the carcinogen sensitive chromosome fragile site, FRA3B (Review). Biochim. Biophys. Acta, 1332: M65– M70, 1997. 5. Lebeau, M. M., Drabkin, H., Glover, T. W., Gemmill, R., Rassool, F. V., Mckeithan, T. W., and Smith, D. I. An FHIT tumor suppressor gene. Genes Chromosomes Cancer, 21: 281–289, 1998. 6. Mao, L. Tumor suppressor genes: does FHIT fit? (Editorial; Comment). J. Natl. Cancer Inst., 90: 412– 414, 1998. 7. Sozzi, G., Veronese, M. L., Negrini, M., Baffa, R., Cotticelli, M. G., Inoue, H., Tornielli, S., Pilotti, S., DeGregorio, L., Pastorino, U., Pierotti, M. A., Ohta, M., Huebner, K., and Croce, C. M. The FHIT gene 3p14.2 is abnormal in lung cancer. Cell, 85: 17–26, 1997. 8. Sozzi, G., Sard, L., DeGregorio, L., Marchetti, A., Musso, K., Buttitta, F., Tornielli, S., Pellegrini, S., Veronese, M. L., Manenti, G., Incarbone, M., Chella, A., Angeletti, C. A., Pastorino, U., Huebner, K., Bevilaqua, G., Pilotti, S., Croce, C. M., and Pierotti, M. A. Association between cigarette smoking and FHIT gene alterations in lung cancer. Cancer Res., 57: 2121–2123, 1997. 9. Fong, K. M., Biesterveld, E. J., Virmani, A., Wistuba, I., Sekido, Y., Bader, S. A., Ahmadian, M., Ong, S. T., Rassool, F. V., Zimmerman, P. V., Giaccone, G., Gazdar, A. F., and Minna, J. D. FHIT and FRA3B 3p14.2 allele loss are common in lung cancer and preneoplastic bronchial lesions and are associated with cancer-related FHIT cDNA splicing aberrations. Cancer Res., 57: 2256 –2267, 1997. 10. Virgilio, L., Shuster, M., Gollin, S. M., Veronese, M. L., Ohta, M., Huebner, K., and Croce, C. M. FHIT gene alterations in head and neck squamous cell carcinomas. Proc. Natl. Acad. Sci. USA, 93: 9770 –9775, 1996. 11. Bugert, P., Wilhelm, M., and Kovacs, G. FHIT gene and the FRA3B region are not involved in the genetics of renal cell carcinomas. Genes Chromosomes Cancer, 20: 9 –15, 1997. 12. van den Berg, A., Draaijers, T. G., Kok, K., Timmer, T., Van Der Veen, A. Y., Veldhuis, P. M., de Leij, L., Gerhartz, C. D., Naylor, S. L., Smith, D. I., and Buys, C. H. Normal FHIT transcripts in renal cell cancer- and lung cancer-derived cell lines, including a cell line with a homozygous deletion in the FRA3B region. Genes Chromosomes Cancer, 19: 220 –227, 1997. 13. Michael, D., Beer, D. G., Wilke, C. W., Miller, D. E., and Glover, T. W. Frequent deletions of FHIT and FRA3B in Barrett’s metaplasia and esophageal adenocarcinomas. Oncogene, 15: 1653–1659, 1997. 14. Siprashvili, Z., Sozzi, G., Barnes, L. D., McCue, P., Robinson, A. K., Eryomin, V., Sard, L., Tagliabue, E., Greco, A., Fusetti, L., Schwartz, G., Pierotti, M. A., Croce, C. M., and Huebner, K. Replacement of Fhit in cancer cells suppresses tumorigenicity. Proc. Natl. Acad. Sci. USA., 94: 13771–13776, 1997. 15. Otterson, G. A., Xiao, G. H., Geradts, J., Jin, F., Chen, W. D., Niklinska, W., Kaye, F. J., and Yeung, R. S. Protein expression and functional analysis of the FHIT gene in human tumor cells. J. Natl. Cancer Inst., 90: 426 – 432, 1998. 16. Fang, B., Koch, P. E., and Roth, J. A. Diminishing adenovirus gene expression and viral replication by promoter replacement. J. Virol., 71: 4798 – 4803, 1997. 17. Ji, L., Bouvet, M., Price, R., Roth, J. A., and Fang, B. Reduced toxicity, attenuated immunogenicity and efficient mediation of human p53 gene expression in vivo by an adenovirus vector with deleted E1–E3 and inactivated E4 by GAL4-TATA promoter replacement. Gene Ther., 6: 393– 402, 1999. 18. Mao, L., Fan, Y. H., Lotan, R., and Hong, W. K. Frequent abnormalities of FHIT, a candidate tumor suppressor gene, in head and neck cancer cell lines. Cancer Res., 56: 5128 –5131, 1996. 19. Sozzi, G., Tornielli, S., Tagliabue, E., Sard, L., Pezzella, F., Pastorino, U., Minoletti, F., Pilotti, S., Ratcliffe, C., Veronese, M. L., Goldstraw, P., Huebner, K., Croce, C. M., and Pierotti, M. A. Absence of Fhit protein in primary lung tumors and cell lines with FHIT gene abnormalities. Cancer Res., 57: 5207–5212, 1997. 20. Hadaczek, P., Siprashvili, Z., Markiewski, M., Domagala, W., Druck, T., McCue, P. A., Pekarsky, Y., Ohta, M., Huebner, K., and Lubinski, J. Absence or reduction of Fhit expression in most clear cell renal carcinomas. Cancer Res., 58: 2946 –2951, 1998. 21. Mao, L., Lee, J. S., Kurie, J. M., Fan, Y. H., Lippman, S. M., Lee, J. J., Ro, J. Y., Broxson, A., Yu, R., Morice, R. C., Kemp, B. L., Khuri, F. R., Walsh, G. L., Hittelman, W. N., and Hong, W. K. Clonal genetic alterations in the lungs of current and former smokers. J. Natl. Cancer Inst., 89: 857– 862, 1997. 22. Levine, A. J. p53, the cellular gatekeeper for growth and division. Cell, 88: 323–331, 1997. 23. Naik, P., Karrim, J., and Hanahan, D. The rise and fall of apoptosis during multistage tumorigenesis: down-modulation contributes to tumor progression from angiogenic progenitors. Genes Dev., 10: 2105–2116, 1996. 24. Roth, J. A., and Cristiano, R. J. Gene therapy for cancer: What have we done and where are we going? (Review). J. Natl. Cancer Inst., 89: 21–39, 1997. 25. Clayman, G. L., Liu, T. J., Overholt, S. M., Mobley, S. R., Wang, M., Janot, F., and Goepfert, H. Gene therapy for head and neck cancer: comparing the tumor suppressor gene p53 and a cell cycle regulator WAF1/CIP1 (P21). Arch. Otolaryngol. Head Neck Surg., 122: 489 – 493, 1996. 26. Brenner, C., Garrison, P., Gilmour, J., Peisach, D., Ringe, D., Petsko, G. A., and Lowenstein, J. M. Crystal structures of HINT demonstrate that histidine triad proteins are GalT-related nucleotide-binding proteins. Nat. Struct. Biol., 4: 231–238, 1997. 27. Sherr, C. J. Cancer cell cycles (Review). Science (Washington DC), 274: 1672–1677, 1996. 28. Pace, H. C., Garrison, P. N., Robinson, A. K., Barnes, L. D., Draganescu, A., Rosler, A., Blackburn, G. M., Siprashvili, Z., Croce, C. M., Huebner, K., and Brenner, C. Genetic, biochemical, and crystallographic characterization of Fhit-substrate complexes as the active signaling form of Fhit. Proc. Natl. Acad. Sci. USA, 95: 5484 – 5489, 1998. 29. Lima, C. D., Klein, M. G., and Hendrickson, W. A. Structure-based analysis of catalysis and substrate definition in the HIT protein family. Science (Washington DC), 278: 286 –290, 1997. 30. Sozzi, G., Huebner, K., and Croce, C. M. FHIT in human cancer. Adv. Cancer Res., 74: 141–166, 1998. 31. Green, D. R., and Reed, J. C. Mitochondria and apoptosis (Review). Science (Washington DC), 281: 1309 –1312, 1998. 32. Kisselev, L. L., Justesen, J., Wolfson, A. D., and Frolova, L. Y. Diadenosine oligophosphates (Ap(n)A), a novel class of signalling molecules? (Review). FEBS Lett., 427: 157–163, 1998. 33. Vartanian, A., Narovlyansky, A., Amchenkova, A., Turpaev, K., and Kisselev, L. Interferons induce accumulation of diadenosine triphosphate (Ap3A) in human cultured cells. FEBS Lett., 381: 32–34, 1996. 34. Vartanian, A., Prudovsky, I., Suzuki, H., Dal Pra, I., and Kisselev, L. Opposite effects of cell differentiation and apoptosis on Ap3A/Ap4A ratio in human cell cultures. FEBS Lett., 415: 160 –162, 1997. 35. Wang, X. W., and Harris, C. C. TP53 tumor suppressor gene: clues to molecular carcinogenesis and cancer therapy (Review). Cancer Surv., 28: 169 –196, 1996. 36. Evan, G., and Littlewood, T. A matter of life and cell death. Science (Washington DC), 281: 1317–1322, 1998.

3339

Downloaded from cancerres.aacrjournals.org on June 1, 2013. © 1999 American Association for Cancer Research.