head and neck squamous cell growth suppression using ...€¦ · the recombinant wild-type p53...

8
Vol. 3. 185-191, February 1997 Clinical Cancer Research 185 Head and Neck Squamous Cell Growth Suppression Using Adenovirus-p53-FLAG: A Potential Marker for Gene Therapy Trials’ S. Mark Overholt,2 Ta-Jen Liu,2 Dorothy L. Taylor, Mary Wang, Adel K. El-Naggar, Ed Shillitoe, Karen Adler-Storthz, Lisa St. John, Wei-Wei Zhang, Jack A. Roth, and Gary L. Clayman3 Departments of Head and Neck Surgery [S. M. 0.. T-J. L., D. L. 1., M. W.. G. L. Cl, Pathology [A. K. E-N.l, Section of Thoracic Molecular Oncology. and Department of Thoracic and Cardiovascular Surgery IW-W. Z.. J. A. Ri. The University of Texas M. D. Anderson Cancer Center, Houston, Texas 77030; Department of Microbiology and Immunology. State University of New York. Syracuse, New York 13210 IE. 5.1: Department of Basic Sciences. The University of Texas Health Science Center Dental Branch, Houston, Texas 77030 1K. A-SI: and University of Texas. Houston, Graduate School of Biomedial Sciences, Houston. Texas 77030 IL. S. J.I ABSTRACT The recombinant wild-type p53 adenovirus has been proven effective against the growth of human head and neck squamous cell cancer (SCCHN) cell lines in vitro and in a nude mouse model. The addition of a FLAG peptide se- quence was used in this study, along with the p53 adenovirus vector as a marker of the site of the gene therapy activity. It provides clear evidence of the exogenous gene product within the transduced carcinoma cells. No alterations in transcription or translation of the p53 gene product were noted with the addition of the FLAG sequence to the original p53 adenovirus vector. Immunohistochemical analysis dis- played simultaneous expression of the p53 and FLAG pro- teins in the infected cells. The p53 protein remained local- Received 9/16/96: revised I 1/7/96: accepted I 1/8/96. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. I This work was supported in part by American Cancer Society Career Development Award 93-9 (to G. L. C.): M. D. Anderson Cancer Center Core Grant NIH-National Cancer Institute CA- I 6672: National Cancer Institute Grant ROl CA-45l87 (to J. A. R.): Training Grant CA0961 I (to J. A. R.); National Institute Grant RS5DE/OD10846 (to K. A-S.): National Institute Grant ROl DE-l0842 (to E. S.): National Science Foundation Graduate Research Fellowship (to L. S. J.): gifts to the Division of Surgery from Tenneco and Exxon for the Core Lab Facility: and a generous gift from the Mathers Foundation. 2 Contributed equally to the concept. investigation. and writing of this work. 3 To whom requests for reprints should be addressed. at Department of Head and Neck Surgery. The University of Texas M. D. Anderson Cancer Center, I 5 15 Holcombe Boulevard. Box 69, Houston, TX 77030. Fax: (713) 794-4662. ized to the nucleus, whereas the FLAG protein was additionally noted in the cytoplasm. In vitro growth suppres- sion assays and in vivo microscopic residual tumor model experiments in nude mice showed a similar tumoricidal effect with the p53-FLAG adenovirus vector to that with the previously studied p53 adenovirus vector without the addi- tion of the FLAG sequence. We conclude that the addition of the FLAG octapeptide sequence allows identification of those cells that have been affected by the molecular therapy independent of the endogenous gene expression of the cells. This novel molecular tracer may prove useful in character- izing infection efficiency and in gene therapy trials. INTRODUCTION Locoregional control of disease, despite improvement over the past several decades, continues to impact significantly on patients with SCCHN.4 Distant metastases fortuitously occur late in the course of these tumors and. overall, occur in only approximately II% ( I ). Although long-term goals remain fo- cused on the complicated sequence of molecular events leading to carcinoma, the immediate goals that challenge researchers are 2-fold: (a) detect changes in epithelium before overt malignancy occurs; and (b) develop strategies to eradicate local and regional disease with minimal host morbidity. Brennan et ai. (2) have recently described the use of p53 satellite mapping around the resection margins of SCCHNs. They demonstrated that muta- tions of the p53 gene in the margins of resection increased the risk of local recurrence. Boyle et a!. (3) found that p53 muta- tions precede invasion in SCCHN. Mutations of the p53 gene are present in up to 60% ofSCCHNs (4-8). As new markers for premalignancy develop with greater site and tumor specificity and sensitivity. we may be able to predict early progression toward malignancy. Gene therapy strategies have shown promise in controlling local and regional tumor burden in both in vitro and in animal models (9-15). Specifically. in a head and neck squamous cell carcinoma. growth suppression and cytotoxicity have been doe- umented using the wild-type p53 gene delivered via a replica- lion-defective adenovirus vector (9). A dose-dependent tumori- cidal effect independent of the host endogenous p53 status has additionally been shown. The mechanism of action appears to be apoptosis (16. 17). Furthermore. a recent microscopic residual disease model that mimics postsurgical resection situations in humans has been described ( 10). Such a model lays the ground- work for carryover application in a human trial. 4 The abbreviations used are: SCCHN, head and neck squamous cell carcinoma: Ad, adenovirus: CMV. cytomegalovirus: pfu. plaque-form- ing unit(s): TNF. tumor necrosis factor. on July 2, 2021. © 1997 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

Upload: others

Post on 11-Feb-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

  • Vol. 3. 185-191, February 1997 Clinical Cancer Research 185

    Head and Neck Squamous Cell Growth Suppression Using

    Adenovirus-p53-FLAG: A Potential Marker for Gene

    Therapy Trials’

    S. Mark Overholt,2 Ta-Jen Liu,2

    Dorothy L. Taylor, Mary Wang,

    Adel K. El-Naggar, Ed Shillitoe,

    Karen Adler-Storthz, Lisa St. John,

    Wei-Wei Zhang, Jack A. Roth, and

    Gary L. Clayman3

    Departments of Head and Neck Surgery [S. M. 0.. T-J. L., D. L. 1.,

    M. W.. G. L. Cl, Pathology [A. K. E-N.l, Section of Thoracic

    Molecular Oncology. and Department of Thoracic and Cardiovascular

    Surgery IW-W. Z.. J. A. Ri. The University of Texas M. D. Anderson

    Cancer Center, Houston, Texas 77030; Department of Microbiology

    and Immunology. State University of New York. Syracuse, New

    York 13210 IE. 5.1: Department of Basic Sciences. The University of

    Texas Health Science Center Dental Branch, Houston, Texas 77030

    1K. A-SI: and University of Texas. Houston, Graduate School of

    Biomedial Sciences, Houston. Texas 77030 IL. S. J.I

    ABSTRACT

    The recombinant wild-type p53 adenovirus has been

    proven effective against the growth of human head and neck

    squamous cell cancer (SCCHN) cell lines in vitro and in a

    nude mouse model. The addition of a FLAG peptide se-

    quence was used in this study, along with the p53 adenovirus

    vector as a marker of the site of the gene therapy activity. It

    provides clear evidence of the exogenous gene product

    within the transduced carcinoma cells. No alterations in

    transcription or translation of the p53 gene product were

    noted with the addition of the FLAG sequence to the original

    p53 adenovirus vector. Immunohistochemical analysis dis-

    played simultaneous expression of the p53 and FLAG pro-

    teins in the infected cells. The p53 protein remained local-

    Received 9/16/96: revised I 1/7/96: accepted I 1/8/96.

    The costs of publication of this article were defrayed in part by the

    payment of page charges. This article must therefore be hereby marked

    advertisement in accordance with 18 U.S.C. Section 1734 solely toindicate this fact.

    I This work was supported in part by American Cancer Society Career

    Development Award 93-9 (to G. L. C.): M. D. Anderson Cancer Center

    Core Grant NIH-National Cancer Institute CA- I6672: National CancerInstitute Grant ROl CA-45l87 (to J. A. R.): Training Grant CA0961 I

    (to J. A. R.); National Institute Grant RS5DE/OD10846 (to K. A-S.):National Institute Grant ROl DE-l0842 (to E. S.): National Science

    Foundation Graduate Research Fellowship (to L. S. J.): gifts to the

    Division of Surgery from Tenneco and Exxon for the Core Lab Facility:

    and a generous gift from the Mathers Foundation.2 Contributed equally to the concept. investigation. and writing of this

    work.

    3To whom requests for reprints should be addressed. at Department ofHead and Neck Surgery. The University of Texas M. D. Anderson

    Cancer Center, I 5 15 Holcombe Boulevard. Box 69, Houston, TX

    77030. Fax: (713) 794-4662.

    ized to the nucleus, whereas the FLAG protein was

    additionally noted in the cytoplasm. In vitro growth suppres-

    sion assays and in vivo microscopic residual tumor model

    experiments in nude mice showed a similar tumoricidal

    effect with the p53-FLAG adenovirus vector to that with the

    previously studied p53 adenovirus vector without the addi-

    tion of the FLAG sequence. We conclude that the addition of

    the FLAG octapeptide sequence allows identification of

    those cells that have been affected by the molecular therapy

    independent of the endogenous gene expression of the cells.

    This novel molecular tracer may prove useful in character-

    izing infection efficiency and in gene therapy trials.

    INTRODUCTION

    Locoregional control of disease, despite improvement over

    the past several decades, continues to impact significantly on

    patients with SCCHN.4 Distant metastases fortuitously occur

    late in the course of these tumors and. overall, occur in only

    approximately I I % ( I ). Although long-term goals remain fo-

    cused on the complicated sequence of molecular events leading

    to carcinoma, the immediate goals that challenge researchers are

    2-fold: (a) detect changes in epithelium before overt malignancy

    occurs; and (b) develop strategies to eradicate local and regional

    disease with minimal host morbidity. Brennan et ai. (2) have

    recently described the use of p53 satellite mapping around the

    resection margins of SCCHNs. They demonstrated that muta-

    tions of the p53 gene in the margins of resection increased the

    risk of local recurrence. Boyle et a!. (3) found that p53 muta-

    tions precede invasion in SCCHN. Mutations of the p53 gene

    are present in up to 60% ofSCCHNs (4-8). As new markers for

    premalignancy develop with greater site and tumor specificity

    and sensitivity. we may be able to predict early progression

    toward malignancy.

    Gene therapy strategies have shown promise in controlling

    local and regional tumor burden in both in vitro and in animal

    models (9-15). Specifically. in a head and neck squamous cell

    carcinoma. growth suppression and cytotoxicity have been doe-

    umented using the wild-type p53 gene delivered via a replica-

    lion-defective adenovirus vector (9). A dose-dependent tumori-

    cidal effect independent of the host endogenous p53 status has

    additionally been shown. The mechanism of action appears to be

    apoptosis (16. 17). Furthermore. a recent microscopic residual

    disease model that mimics postsurgical resection situations in

    humans has been described ( 10). Such a model lays the ground-

    work for carryover application in a human trial.

    4 The abbreviations used are: SCCHN, head and neck squamous cell

    carcinoma: Ad, adenovirus: CMV. cytomegalovirus: pfu. plaque-form-

    ing unit(s): TNF. tumor necrosis factor.

    on July 2, 2021. © 1997 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 186 p53-FLAG: Potential Marker for Gene Therapy Trials

    As more promising therapies develop and we entertain the

    application of gene therapy in the head and neck, the ability to

    trace the sites affected by the therapy and identify which cells

    have been affected by treatment becomes more important. De-

    livery of a novel tracer would, therefore, provide definitive

    evidence for infection efficiency and extent of effect, both

    locally and distantly.

    The FLAG biosystem (18), originally described in 1988,

    allows one to tag and examine the eft’eet of an exogenously applied

    treatment. Originally, it was used in the purification of large pro-

    teins, but its design lends itself to other applications. This strategy

    generates a fusion protein of the FLAG peptide and the gene of

    interest. A monoclonal antibody against the polypeptide enables us

    to detect the location of a fusion protein that would correspond to

    clonal expression of the delivered gene. The polypeptide is eight

    amino acids in length, and its small size should not disrupt the

    expression of the delivered gene therapy protein. If the biological

    activity remains unchanged, the ability to co-deliver such a marker

    holds great implication for future human trials. The goal of this

    investigation was to compare the biological effect of the previously

    described wild-type pS3 adenovirus to that of the vector modified

    with the FLAG octapeptide sequence. We demonstrated that the

    biological effect was not changed by the addition of the FLAG

    sequence to the adenovirus p53 vector (AdCMV-p53), and that by

    using immunohistochemical techniques, we have the ability to trace

    the location of the effect following the adenovirus administration.

    MATERIALS AND METHODS

    Cell Lines and Culture Conditions. Human SCCHN

    cell lines Tu-l 38 and 686-LN were both established by the De-

    partment of Head and Neck Surgery. The University of Texas

    M. D. Anderson Cancer Center, and have been characterized pre-

    viously (19, 20). These cells were grown in DMEM/Fl2 medium

    supplemented with 10% heat-inactivated fetal bovine serum with

    streptomycin/penicillin at 37#{176}Cand 5% carbon dioxide.

    Recombinant Adenovirus Preparation and Infection.

    The recombinant p53 adenovirus (AdCMV-p53) contains the

    CMV promoter, wild-type p53 eDNA, and SV4O polyadenyl-

    ation signal in a minigene cassette inserted into the El-deleted

    region of modified AdS (21). Viral stocks were propagated in

    293 cells. Cells were harvested 36-40 h after infection, pel-

    leted, resuspended in PBS, and lysed. Cell debris was then

    removed by subjecting the cells to CsCl gradient purification.

    Concentrated virus was dialyzed, aliquoted, and stored at

    -80#{176}C.Infection was carried out by the addition of virus to theDMEMIFI2 medium with 10% fetal bovine serum to the cell

    monolayers. The cells were incubated at 37#{176}Cfor 60 mm with

    agitation. Then complete medium (DMEMIF12 with 10% fetal

    bovine serum) was added to the cells in the appropriate volume

    for the given Petri dish and incubated for the desired time.

    Generation of the p53-FLAG Adenovirus. The p53

    eDNA sequence was excised from the pCS3-SN [kindly pro-

    vided by Dr. G. Lozano (The University of Texas M. D. Ander-

    son Cancer Center, Houston, IX)1 by digestion with BwnHI and

    cloned into the BainHI site of pGEM7Z. A recombinant plasmid

    with the proper insert orientation was then digested with AccI

    and Kpnl to remove 21 amino acids from the 3’ end of p53

    eDNA. A linker with AccI-KjmI compatible ends containing the

    sequence of the FLAG peptide including a stop codon was then

    ligated into the digested plasmid to create the p53-FLAG fusion

    gene. The resulting p53-FLAG fusion gene was then cloned into

    an expression vector with the human CMV promoter and SV

    polyadenylation signal. The final construct was subsequently

    inserted into a shuttle vector pXCJL. I (2 1 ) to generate a recom-

    binant p53-FLAG adenovirus.

    Northern Blot Analysis. Total RNA was isolated by the

    acid-guanidinium thiocyanate method of Chomczynski and Sacehi

    (22). Northern analyses were performed on 20 �.tg oftotal RNA run

    on a 1% agarose gel containing 2.2 M formaldehyde. The mem-

    brane was hybridized overnight at 65#{176}Cwith a p53 eDNA probe

    labeled by the random primer method in 5 X SSC, 5 X Denhardt’ssolution, 0.5% SDS denatured salmon sperm DNA (20 j.tg/ml).

    Western Blot Analysis. Total cell lysates were prepared

    by sonicating the cells 24 h after infection in RIPA buffer ( 150

    mM NaC1, 1.0% NP4O, 0.5% sodium deoxycholate. 0.1% SDS,

    and 50 filM Iris, pH 8.0). Fifty �.tg of protein from samples were

    subjected to 10% PAGE and transferred to a Hybond-ECL

    membrane (Amersham Corp.). The membrane was blocked with

    Blotto/Tween (5% nonfat dry milk and 0.2% Tween 20 in PBS)

    and probed with the primary antibodies. anti-FLAG M2 mono-

    clonal antibody (IB13002), mouse anti-human p53 monoclonal

    antibody (PAb I 80 1 ), and mouse anti-human B-actin mono-

    clonal antibody (Amersham). The secondary antibody, horse-

    radish peroxidase-conjugated goat anti-mouse IgG (Boehringer

    Mannheim, Indianapolis, IN), was then exposed to the mem-

    brane. The membrane was then processed and developed as the

    manufacturer specified.

    Cell Growth Assay. Cells were plated at a concentration

    of 2 X l0� cells/mI in six-well plates in triplicate, and cells were

    infected with AdCMV-p53-FLAG, AdCMV-p53. or the repli-

    cation-defective adenovirus control (DL3I2), at an multiplicity

    of infection of I 00. A mock infection was also used as a control,

    against which to compare the effect of the replication-deficient

    adenovirus and AdCMV-p53. Cells were then trypsinized and

    counted at specified time points. Viability was checked using

    trypan blue exclusion.

    Immunohistochemical Staining of in Vitro Cell Layers.

    Infected cell monolayers were fixed with 3.8% buffered forma-

    lin and treated with H,O� in methanol for 5 mm. Immunohis-

    tochemical staining was performed using the Vectastain Elite kit

    (Vector Laboratories, Burlingame. CA). The primary antibody

    was either the mouse anti-FLAG M2 monoclonal antibody

    (IB13002) or mouse anti-human p53 monoclonal antibody

    (PAbl8Ol), and the secondary antibody was an avidin-labeled

    anti-mouse IgG (Vector). The biotinylated horseradish peroxi-

    dase avidin-biotin complex reagent was used to detect the anti-

    gen-antibody complex. Preadsorption controls were used in each

    experiment. The cells were then counterstained with Harris

    hematoxylin (Sigma Chemical Co., St. Louis, MO).

    Immunohistochemical Staining of Tissue from in Vivo

    Studies. Formalin-fixed, paraffin-embedded in vito animal

    experimental tissues were cut at 4-5 p.si. dried at 60#{176}C.depar-

    af’finized, and hydrated with distilled water. Sections were then

    treated with 0.5% saponin in distilled water and rinsed in several

    changes of distilled water; endogenous peroxidase activity was

    blocked with 3% hydrogen peroxide in methanol, followed by

    rinsing in several changes of distilled water. Sections were

    on July 2, 2021. © 1997 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 1234567 89

    2.8 Kb

    1.9 Kb

    Endogenous

    Exogenous

    Clitiical Cancer Research 187

    Fig. I Expressiots of exogenous p53

    mRNA 24 h after infection with

    AdCMV-p53 and AdCMV-p53-

    FLAG. La,u’ I. RNA molecularweight marker. LAUl(’S’ 2-5, Tv- I 38

    (niutated PS3) cell line. Lane.s 6-9.

    686-LN (wild-type PS3) cell line.

    Laiies 2 and 6. mock infection. Lanes

    3 and 7. DL3 I 2 infection. Lotus 4 and

    8. AdCMV-p53 infection. Lanes’ 5 and

    9. AdCMV-p53-FLAG infection.

    microwave-irradiated in distilled water for 3 mm using a Sharp

    model R9H8 I microwave oven operating at a frequency of 2450

    MHz at 700 W. After cooling, sections were washed in several

    changes of distilled water and placed in PBS; immunochemical

    studies were performed by using the avidin-biotin-peroxidase

    complex method of Hsu ci ai. (23) in the following manner.

    Sections were blocked with normal horse serum and incubated

    overnight at 4#{176}Cwith anti-FLAG M2 monoclonal antibody at a

    concentration of 60 p.g/ml (International Biotechnologies, New

    Haven, CT) and rabbit antihurnan p53 polyclonal antibody,

    clone OM-1. 1:80 (Signet Laboratories, Denham. MA). An

    antirabbit IgG Elite kit (Vector) was then used to apply bioti-

    nylated antirabbit IgG and avidin-biotin-peroxidase complexes

    that were incubated for 45 mm each. The immunostaining

    reaction was visualized by using 0.5% dimethylaminoazoben-

    zene in PBS containing 0.01% H,02 (pH 7.6), counterstained

    with 0.01% toluidine blue. dehydrated. cleared. and mounted in

    Permount.

    In Vivo Microscopic Residual Disease Experiments. In

    t’ivo experiments were performed in a defined pathogen-free

    environment using athymic nude mice. The microscopic resid-

    ual disease model described previously by our laboratory was

    applied ( 10). Experiments were reviewed and approved by in-

    stitutional review committees for both animal care and utiliza-

    tion and the Biosafety Committee for recombinant DNA re-

    search. Briefly, 4-7-week-old female nude mice were

    anesthetized using i.p. ketamine/acepromazine at a dose of 70

    mg/kg of body weight. The bodies were prepared with alcohol

    wipes and incisions were made on four dorsal flanks. s.c. pock-

    ets were elevated, and the desired number of tumor cells was

    delivered in 100 pi of medium into the pocket. The flap was

    then sealed with an interrupted horizontal mattress 5.0 nylon

    suture, insuring a water-tight seal. At 48 h, the flaps were

    reopened, and 100 ii of an appropriate concentration of virus

    were delivered into the same bed inoculated previously with the

    tumor cells. Two different sets of repeated experiments were

    performed. The first was a dose-response experiment using the

    AdCMV-p53-FLAG virus in three of the flaps at descending

    concentrations ( l0�, l0�, 106 pfu). The fourth flap served as a

    control and was randomized to either PBS or the replication-

    defective adenovirus (DL312). The second study was performed

    using l0� pfu of AdCMV-p53-FLAG. AdCMV-p53, and repli-

    cation-defective adenovirus in three separate flaps. The fourth

    flap was inoculated with the same volume ( 100 p.1) of sterile

    PBS. Forty-eight h after treatment with virus, two animals were

    sacrificed, and the flaps were harvested for immunohistochem-

    ical analysis. The remaining animals were observed for 2 1 days

    and then sacrificed. Tumor volumes were measured for corn-

    parison using calipers.

    RESULTS

    Expression of mRNA after Infection with AdCMV-p53

    and AdCMV-p53-FLAG Virus. Both To- I 38 and 686-LN

    were examined for expression of p53 mRNA. Total RNA was

    isolated after adenovirus infection. Northern blot analysis was

    performed. Similar levels of exogenous AdCMV-p53 rnRNA

    were detected between AdCMV-p53- and AdCMV-p53-FLAG-

    infected cells. Fig. I shows the comparable levels of p53 mRNA

    expression af’ter infection with AdCMV-p53 and AdCMV-p53-

    FLAG in Lanes 4 and S for Iu-138 and in Lanes 8 and 9 for

    MDA 686-LN. Variation of intensity is felt to be related to

    loading dose. Endogenous expression of p53 rnRNA is seen in

    Lanes 2 and 3 in the mutated p53 cell line, Iu-138. Lanes 6 and

    7 show no endogenous p53 rnRNA in the 686-LN cell line,

    which is wild-type for the p53 gene. These data suggest that the

    AdCMV-p53-FLAG virus, like the AdCMV-p53 virus, is sue-

    cessfully transduced and efficiently transcribed. Northern anal-

    ysis did not reveal evidence of AdCMV-p53 DNA contarnina-

    tion.

    Expression of Exogenous p53 Protein in AdCMV-p53-

    and AdCMV-p53-FLAG-infected SCCHN Cell Lines.

    Western blot analysis was performed to compare the amount of

    protein expressed by the AdCMV-p53- and AdCMV-p53-

    FLAG-infected cells. Protein bands were identified using the

    monospecifie p53 antibody (PAbl8Ol) and the anti-FLAG M2

    antibody (1B13002) on two simultaneously run gels. Fig. 2 (top)

    showed, using the p53 antibody (PAbl8Ol ), a similar high level

    of p53 protein expression in both cell lines that were infected

    with the AdCMV-p53 and AdCMV-p53-FLAG. Lanes 3 and 7

    correspond to the To- I 38 and MDA 686-LN cells infected with

    AdCMV-p53, respectively. Lanes 4 and S of the blot represent

    protein expression from those cells infected with the AdCMV-

    p53-FLAG. No change in p53 protein expression was noted in

    either the replication-defective, adenovirus-infeeted cells or in

    the mock group. The bottom blot demonstrates a similarly

    executed gel that was probed with the mouse anti-FLAG M2

    antibody. The level of p53-FLAG protein expression appeared

    to be similar to that expressed following p53 antibody probing.

    but no detectable band was noted in those cells infected with the

    AdCMV-p53 virus (Fig. 2. lanes 3 and 7). The mock and

    DL3I2-infeeted cells exhibited no detectable level of the im-

    munoreactive p53 protein in either cell line.

    on July 2, 2021. © 1997 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • A

    0 2

    B

    6 8

    tn

    L

    .nE

    2:

    LI

    Days

    Fig. 3 Inhibition of SCCHN cell growth in vitro. A, Tu-138; B, 686-LN. At each indicated time point, three dishes of cells were trypsinizedand counted. The mean cell counts per triplicate wells following infec-

    tion were plotted against the number of days since infection; bars, SE.

    0, mock; A, DL312; EL PS3; U, p53-FLAG.

    2 4 6 8

    188 p53-FLAG: Potential Marker for Gene Therapy Trials

    A

    B

    I 2345678

    �*p53

    ,�. actin

    .� FLAG

    ..�- actin

    Fig. 2 Expression of p53 and

    p53-FLAG fusion protein fromcellular extracts isolated 24 h

    after infection with AdCMV-

    p53 or AdCMV-p53-FLAG.

    Lanes 1-4, Tu-138 (mutated

    pS3) cell line; Lanes 5-8,

    686-LN (wild-type p53) cell

    line. Lanes I and 5, mock in-

    fection. Lanes 2 and 6, DL3 12infection. Lanes 3 and 7,

    AdCMV-p53 infection. Lanes4 and 8, AdCMV-p53-FLAGinfection. A, probed with p53(PAbl8Ol) antibody; B,

    probed with FLAG M2 anti-

    body.

    Effect of AdCMV-p53 and AdCMV-p53-FLAG on

    SCCHN Cell Growth. We have described previously the

    cytotoxic effect of p53 therapy in Tu-l38 and 686-LN cell lines.

    The Iu-l38 cell line has an endogenously mutated p53 gene,

    and the 686-LN cell line possesses the wild-type p53 gene. This

    study sought to determine if any difference in efficacy would be

    seen after manipulation of the AdCMV-p53 virus by inserting

    the FLAG sequence.

    Cells infected with the replication-defective adenovirus had a

    similar growth rate to the mock infection cells. A mild cytotoxic

    effect may be seen with the replication-defective adenovirus (Fig.

    3). In contrast, those cells infected with either the AdCMV-p53 or

    AdCMV-p53-FLAG experienced virtual total tumor cell death by

    day three. Histological examination revealed bleb formation by the

    plasma membrane, which is one of the characteristic features of

    apoptosis. As noted previously in our laboratory, the effect was

    more prominent for the Tu-138 cell line (mutated p53) than it was

    for the MDA 686-LN cell line (wild-type p53). Growth curve

    assays were reproducible in three repeated experiments without a

    significant difference being noted between the effect of the Ad-

    CMV-p53 and the AdCMV-p53-FLAG viruses, suggesting that theaddition of the FLAG peptide did not affect the ability of p53 in

    suppression of cell growth.

    Immunohistochemical Staining of SCCHN Cell Lines

    Infected with Adenovirus. Infected cell monolayers were corn-

    pared for expression of the p53 arid p53-FLAG protein using

    standard immunohistochemical techniques. As controls, both the

    mock-infected cells and the DL3 12-infected cells showed similar

    staining (Fig. 4). Neither the p53 nor FLAG protein could be

    clearly identified in the control group. However, when cells were

    infected with the AdCMV-p53 virus, a strong staining was noted.

    Those cells infected with the AdCMV-p53-FLAG virus showed

    virtually identical intensity of staining and number of positive cells

    with PAbl8Ol for antibody as compared to the cells infected with

    the AdCMV-p53 virus. The cells infected with the AdCMV-p53-

    FLAG virus also showed strong immunohistochemical positivity

    with the M2 FLAG antibody. The quality of staining was different,

    although both were within the nucleus and to a lesser degree in the

    cytoplasm.

    In Vivo Suppression of Growth. Dose-response studies

    using 106, l0�, and 108 pfu of AdCMV-p53-FLAG virus com-

    pared to a control flap that was either PBS or DL3 12 were

    performed using the microscopic model method (10). The mean

    tumor size for the mock infection was 1205 mm (3). Tumor size

    decreased in a linear fashion with increasing concentration of

    virus used in the molecular intervention. Mean tumor size was

    on July 2, 2021. © 1997 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • Clinical Cancer Research 189

    A .. .‘ . 1’ ;-: .� #{149}..‘, I � #{149}� � �‘ �, �, ‘ -

    - � �A � �

    � �‘ ,‘-.-�.. -- .. ,1 #{149}� � :‘,,.�‘T)I.,..

    -‘. . I ‘�: � � �1

    .(a. � � .‘s%�., ‘#{149}-.

    . s-... .-‘‘4 .% . : . L��#{149} #{149}‘:�� . �:

    0#{149}0

    0�

    c� �

    #{149}.:‘� � � g’:�t-��e .

    Fig. 4 In vitro immunohistochemical staining of 686-LN cells infected

    with AdCMV-p53-FLAG. A, endogenous p53 staining. B, p53 antibody(PAbl8Ol). C, FLAG M2 antibody (1B13002). B, nuclear p53 stainingis prominent. C’, cytoplasmic component to staining noted with the

    FLAG antibody. x 100.

    637 mm (3), 392 mm (3), and 193 mm (3) for those flaps treated

    with 106, l0�, and l0� pfu of the AdCMV-p53-FLAG, respec-

    tively. Each animal was compared against itself using a paired

    t test, and a significant dose-response effect was noted at P <

    0.05 in all comparisons, except between the flap treated with l0�

    and l0� pfu. Clearly, the greater the amount of virus, the greater

    the tumor growth inhibition. In an additional study, the in vito

    effects of AdCMV-p53 were compared to that of AdCMV-p53-

    FLAG. No difference in growth suppression activity was noted

    (data not shown).

    Immunohistochemical Demonstration of Exogenous

    Tumor Suppression Effect in the Microscopic Residual Dis-

    ease Animal Model. After proving comparable in vitro and in

    t,ivo activity of AdCMV-p53 and the AdCMV-p53-FLAG. we

    applied immunohistoehemical techniques to demonstrate p53-

    FLAG fusion protein product in vito. Microscopic residual

    disease flaps were harvested 48 h after treatment, fixed in

    formalin, and paraffin embedded. On neighboring sections of

    tumor cells treated with the AdCMV-p53-FLAG virus, staining

    for both the p53 and FLAG protein was applied. Staining

    intensity and the number of cells staining positively was directly.� . proportional to the amount of virus used in the infection. Con-

    trols were negative for staining with both p53 and FLAG anti-

    bodies. Fig. S shows a histological specimen stained with H&E

    (A) the p53 antibody (B) and the FLAG antibody (C). The

    characteristic intranuclear staining with the p53 antibody is

    similarly expressed as the intranuclear staining with the FLAG

    throughout multiple layers of the gene transfer site. This also

    demonstrates that the FLAG M2 antibody is effective on paraf-

    fin-embedded fixed tissue. The staining demonstrates that the

    tumor-suppressive effect is directed by the exogenous therapy,

    and that in an in vito model, one can identify the exogenous

    therapy using the applied FLAG system. lime course protein

    expression experiments using Western blotting show peak pro-

    tein at 3 days and low detectable levels at 15 days. When

    animals were sacrificed at 21 days, tissue sections were, there-

    fore, uninformative using immunohistoehemical methods (data

    not shown).

    DISCUSSION

    The tumor suppressor gene p.53 has been shown previously

    to be an effective molecular therapy against SCCHN in vitro and

    in a nude mouse model (10). In addition, another gene therapy

    strategy using the HSV-TK gene has demonstrated effectiveness

    in this epithelial carcinoma (24). Despite rapid progress directed

    toward the development of gene therapy, an effective unique

    marker, which may establish the transduction oftargeted tissues,

    has not been established. A potential marker gene product is the

    FLAG peptide. So et ai. (25) used this method to isolate a

    TNF-a protein. The FLAG M2 monoclonal antibody was ap-

    plied to identify the FLAG-INF-a fusion protein. Then the

    FLAG peptide was cleaved using enterokinase, leaving thespecific NH2 terminus to the TNF-a molecule. Importantly, the

    fusion FLAG-TNF-a protein and the TNF-a protein alone had

    similar biological function in cytotoxic assays. We hypothesized

    that we could co-deliver the p53-FLAG fusion gene via an

    adenovirus vector, identify it using the FLAG M2 monoclonal

    antibody, and express the tumor suppressor gene product, p53.

    At the same time, identification of those cells transduced by the

    virus could be identified independently of the endogenous p53

    expression of the infected cells by determining the expression of

    the FLAG gene product.

    Initial studies sought to establish the induction of p53

    mRNA following infection with either the AdCMV-p53 or

    AdCMV-p53-FLAG and clearly demonstrate similar exogenous

    mRNA induction. Western blot analysis of p53 and FLAG

    protein expression demonstrated similar levels of p53 protein

    expression in cells infected with the AdCMV-p53 and AdCMV-

    on July 2, 2021. © 1997 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • B� “ “ . �.

    . .

    190 p53-FLAG: Potential Marker for Gene Therapy Trials

    ‘� “ ‘ ‘ .‘:“ “.- ‘ ‘ . . “ � ,�4 ‘

    .. . . ..‘ .. . . .. :: : , . . � � t.1��P;t�� � .‘ ‘�. �

    ‘ ‘ . .. - .: ,‘. �

    �. �‘. I �‘ .: . ; , . ,.- ., .. � .‘-.� , C ��“�‘�-\ A

    �;J�.1C.-.’ ‘ ‘ ‘ : , � . , , .

    w

    � :‘�;‘ � �N�I�’ � :� .� ,, . ,‘.� ‘.- -�‘ . ...� � ‘� � � , �

    Fig. 5 Immunohistochemical staining of formalin-fixed, paraffin-em-bedded tissue from in vito Tu-I38 tumor treated with AdCMV-p53-FLAG. A, H&E. B, p53 antibody (PAbl8Ol). C. FLAG M2 antibody(IBI3002). B, strong nuclear staining noted with p53 antibody. C. strongnuclear staining is also seen utilizing the FLAG antibody. X 100.

    p53-FLAG, respectively. Furthermore, the FLAG antibodydemonstrated that the protein expressed by the cells infected

    with the AdCMV-p53-FLAG was a unique fusion protein ex-

    pressing the FLAG octapeptide moiety that could be differenti-

    ated from the native p53 protein.

    Northern analysis confirmed that the cells infected with

    either the AdCMV-p53 or AdCMV-p53-FLAG exhibited simi-

    lar exogenous mRNA induction. Our previous studies using the

    AdCMV-p53 had demonstrated marked induction of apoptosis

    among infected cancer cell lines ( 16, 26); however, nonmalig-

    nant cell lines were spared. The next question was whether the

    addition of the FLAG sequence to the putative p53 protein

    would alter its inherent growth-inhibitory effect. To this end.

    cell growth assays were performed, and no significant difference

    could be demonstrated between AdCMV-p53 and AdCMV-p53-

    FLAG. As we had described previously, induction of apoptosiswas not observed following mock or replication-defective virus

    infection.

    In vitro immunohistochemical studies were performed to

    establish that the FLAG protein could be identified among

    infected SCCHN cell lines. We were able to show a strong

    nuclear staining for the p53 protein in those cells infected by

    either the AdCMV-p53 or the AdCMV-p53-FLAG. Further-

    more, using the FLAG antibody, the novel FLAG octapeptide

    sequence was demonstrated in those cells that were infected

    with the AdCMV-p53-FLAG only. There appeared to be a slight

    qualitative difference in the staining with a probable cytoplas-

    mic component when probing with the FLAG monoclonal

    antibody.

    in t’it’() experiments were carried out to further explore the

    efficacy of FLAG immunohistochemical staining. Using the

    microscopic residual disease model described previously. s.c.

    tumor sites were infected with increasing concentrations of the

    AdCMV-p53-FLAG vector. A dose-response curve was notedwith greater tumor kill as the virus concentration was increased.

    Flaps infected with AdCMV-p53-FLAG could be effectively

    immunohistoehemically analyzed for both the p53 and FLAG

    proteins using formalin-fixed, paraffin-embedded tissues. The

    large pS3 protein was noted to be primarily intranuclear. as

    documented previously. However, the FLAG protein was noted

    additionally within the cytoplasm. Because the nuclear translo-

    cation signal was altered in the p53 3’-end deletion in this

    FLAG construct, both nuclear and cytoplasniic localization of

    the transgene product were found (27). We were not able to

    demonstrate staining for the FLAG protein outside the region of

    treatment, confirming the local expression of these virions and

    lack of systemic expression when delivered in this model.

    FLAG appears to be a potential marker for tracking proteinproduct expression in gene therapy. We currently have the

    technology to use virus-specific promoters and PCR techniques

    to analyze the presence ofdelivered virions. As a screening tool,

    this would be an effective and sensitive test for identifyi�ig virus.

    However, it does not answer the more important question: is the

    protein product encoded by the delivered virus being expressed?

    Herein lies the major advantage of the FLAG marker. Fixed

    tissue can be analyzed for the presence of the delivered gene

    product. This is particularly important in tumor biology, where

    endogenous heterogeneity for the genes of interest might mask

    locations where a specific gene therapy has been delivered.

    Specifically, in tumors with a mutated p53 gene, exogenous

    expression of the vector-delivered p53 can be differentiated

    from the overexpression of the endogenous p53 of the tumor.

    Such a marker would also demonstrate the expression and effect

    imparted on normal bystander cells. Both of these lend them-

    selves to carryover into human trials, where such questions must

    be addressed.

    Potential criticisms of FLAG as a marker for gene therapy

    are 2-fold: (a) a minimal protein expression is required to

    on July 2, 2021. © 1997 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • Clinical Cancer Research 191

    immunohistochemically identify the gene product. For simple

    documentation of the presence of virion, the sensitivity is far

    less than that of PCR. However, the transduction efficiency of

    the replication-defective adenovirus in SCCHN is excellent, and

    p53 protein expression is high. The added advantage of docu-

    menting expression of the viral product and its histological

    location distinguishes this technique; (b) introduction of novel

    genes has the potential to cause deleterious downstream effects.

    Using a retroviral vector, these potential effects would be irre-

    versible, but with the transient episomal expression of the ade-

    novirus, this hazard is avoided. Lastly, the p53-FLAG and

    p53-adenoviral vectors both show evidence of inflammatory

    infiltrates in animal studies at viral doses of l0� pfu and greater.

    Local inflammatory responses may, however, be beneficial in

    the local tumor environment. The potential systemic immune

    response from viral products and exogenous gene products will

    require further investigations.

    In conclusion, we feel that the co-delivery of the FLAG

    protein along with the desired gene therapy offers potential

    utility as a marker of gene therapy. We were able to show that

    it was simultaneously promoted along with the p53 gene and

    that expression of the mRNA and protein were not decreased.

    More importantly, the biological activity of the delivered tumor

    suppressor gene was not altered. For the first time, the FLAG

    antibody was proven effective when immunohistoehemical

    analysis was perf’ormed on formalin-fixed, paraffin-embedded

    tissue. These factors suggest the utility of this novel protein as

    a tracer in further gene therapy studies.

    REFERENCES

    1. Calhoun, K. H., Fulmer, P., Weiss, R., and Hokanson, J. A. Distant

    metastases from head and neck squamous cell carcinomas. Laryngo-

    scope, 104: 1199-1205, 1994.

    2. Brennan, J. A., Mao, L., Hruban, R. H., Boyle, J. 0., Eby, Y. J..Koch, W. M., Goodman, S. N., and Sidransky, D. Molecular assessmentof histopathological staging in squamous-cell carcinoma of the head and

    neck. N. EngI. J. Med., 332: 429-435. 1995.

    3. Boyle. J. 0.. Hakim, J., Koch, W., van der Riet, P., Hruban, R. H.,

    Roa, R. A., Correo, R., Eby, Y. J., Ruppert, M., and Sidransky, D. Theincidence of p53 mutation increases with progression of head and neck

    cancer. Cancer Res., 53: 4477-4480, 1993.

    4. Field, J. K.. Pavelic, Z. P., Spandidos, D. A.. Stambrook, P. J.. Jones.

    A. S., and Gluckman. J. L. The role of the p53 tumor suppressor genein squamous cell carcinoma of the head and neck. Arch. Otolaryngol.

    Head Neck Surg., 119: 1 1 18-1 122, 1993.

    5. Watling, D. L., Gown, A. M., and Coltrera, M. D. Overexpression of

    p53 in head and neck cancer. Head Neck, 14: 437-444, 1992.

    6. Nylander. K., Stenling, R., Gustafsson, H., Zackrisson, B., and Root,G. p53 expression and cell proliferation in squamous cell carcinomas of

    the head and neck. Cancer (Phila.), 75: 87-93, 1995.

    7. Pavelic, Z. P.. Li, Y-Q., Stambrook, P. J., McDonald, J. S., Munck-

    Wikland, E., Pavelic, K., Dacic, S., Danilovic, Z., Pavelic, L., Mugge,

    R. E., Wilson, K., Nguyen, C.. and Gluckman, J. L. Overexpression ofp53 protein is common in premalignant head and neck lesions. Anti-

    cancer Res., 14: 2259-2266, 1994.

    8. Maestro, R., Doleetti, R., Gasparatto, D., Doglioni, C., Pelucchi, S.,

    Barzan, L., Grandi, E., and Boiocchi, M. High frequency of p53 genealterations associated with protein overexpression in human squamous

    cell carcinoma of the larynx. Oncogene, 7: 1 159-1 166, 1992.

    9. Liu, T-J., Zhang, W-W., Taylor, D. L., Roth, J. A., Goepfert, H., and

    Clayman. G. L. Growth suppression of human head and neck cancercells by the introduction of a wild-type p53 gene via a recombinant

    adenovirus. Cancer Res., 54: 3662-3667, 1994.

    10. Clayman, G. L., El-Naggar, A. K., Roth, J. A.. Zhang. W-W.,Goepfert. H.. Taylor. D. L., and Liu, 1-i. In vito molecular therapy withp53 adenovirus for microscopic residual head and neck squamous car-

    cinoma. Cancer Res., 55: 1-6, 1995.

    1 1 . Baker, S. J., Markowitz, S., Fearon, E. R., Wilson, J. R., and

    Vogelstein, B. Suppression of human colorectal carcinoma cell growth

    by wild-type p53. Science (Washington DC), 249.’ 912-915, 1990.

    12. Mercer, W. E., Shields, M. 1., Amin, M., Sauve, G. J., Apella, E.,

    Romano, J. W., and Ulrich, S. J. Negative growth regulation in aglioblastoma tumor cell line that conditionally expresses human wild-

    Type p53. Proc. NatI. Acad. Sci. USA, 87: 6166-6170, 1990.

    13. Diller, L., Kassel, J., Nelson, C. E., Gryka, M. A., Litwank, G.,

    Gebhardt, M., Bressac, B., Ozturk, M., Baker, J. J., and Vogelstein, B.p53 functions as a cell cycle control protein in osteosarcoma. Mol. Cell.

    Biol.. 10: 5772-5781. 1990.

    14. Shaw, P., Bovey, R., Tardy, S., Sahli, R., Sordet, B., and Costa,

    J. Induction of apoptosis by wild-type p53 in human colon tumor-derived cell line. Proc. NatI. Acad. Sci. USA, 89: 4495-4499, 1992.

    15. Fujiwara. 1., Grimm, E. A., Murkhopadhyay, 1., Cal, D. W.,Owen-Schaub, L. B., and Roth, J. A. A retroviral wild-type p53 expres-sion vector penetrates human lung cancer spheroids and inhibits growth

    by inducing apoptosis. Cancer Res., 53: 4129-4133, 1993.

    16. Liu, T-J., El-Naggar, A. K., McDonnell, 1. J., Steck, K. D., Wang,M., Taylor. D. L.. and Clayman. G. L. Apoptosis induction mediated by

    wild-type p53 adenovirus gene transfer in squamous cell carcinoma ofthe head and neck. Cancer Res., 55: 31 17-3122, 1995.

    I 7. Hartwell, L. H., and Kastan, M. B. Cell cycle control and cancer.

    Science (Washington DC), 266: 1821-1828, 1994.

    18. Hopp, 1. P., Prickett, K. S., Price, V., Libby, R. 1., March, C. J.,

    Cerretti, P., Urdal, D. L., and Conlon, P. J. A short polypeptide marker

    sequence useful for identification and purification of recombinant pro-

    teins. Biotechnology, 7: 1205-1210, 1988.

    19. Clayman, G. L., Wang, S. W., Nicholson, G. L., El-Naggar, A. K..Mazar, A.. Henkins, J., Blasi, F., Goepfert, H., and Boyd, D. D.Regulation of urokinase-type plasminogen activator expression in squa-

    mous-cell carcinoma ofthe oral cavity. Int. J. Cancer, 54: 73-80, 1993.

    20. Sacks, P. G., Parnes, S. M., Gallick, G. E., Mansouri, Z., Lichtner,

    R., Satya-Prakash. K. L.. Pathak, S., and Parsons, D. F. Establishmentand characterization of two new squamous cell carcinoma cell lines

    derived From tumors of the head and neck. Cancer Res., 48: 2858-

    2866, 1988.

    21. Zhang. W-W.. Fang, X., Mazur, W., French, B. A., Georges, R. N.,

    and Roth, J. A. High-efficiency gene transfer and high-level expression

    of wild-type p53. I. Human lung cancer cells mediated by recombinant

    adenovirus. Cancer Gene Ther., I: 1-10, 1994.

    22. Chomczynski, P., and Sacchi, N. Single-step method of RNA iso-

    lation by guanidinium thiocyanate-phenol-chloroform extraction. Anal.

    Biochem., 162: 156-159, 1987.

    23. Hsu, S. M., Raine, L., and Fanger, H. Use of avidin-biotin-perox-

    idase complex (ABC) in immunoperoxidase techniques: a comparison

    between ABC and unlabeled antibody (PAP) procedures. J. Histochem.Cytochem.. 29: 577-580, 1981.

    24. O’Malley. B. W., Jr., Chen, S-H., Schwartz, M. R., and Woo,

    S. L. C. Adenovirus-mediated gene therapy for human head and neck

    squamous cell cancer in a nude mouse model. Cancer Res., 55: 1080-

    1085, 1995.

    25. So, X., Prestwood, A. K., and McGraw, R. A. Production of

    recombinant porcine tumor necrosis factor a in a novel E. cohi expres-sion system. Biotechniques, /3: 756-761, 1992.

    26. Wang. J., Bucana, C. D., Roth, J. A., and Zhang, W-W. Apoptosisinduced in human osteosarcoma cells is one of the mechanisms for the

    cytocidal effect of AdCMV-p53. Cancer Gene Ther., 2: 1-9, 1995.

    27. Shaulsky, G., Goldfinger, N., Ben-Ze’ev, A., and Rotter, V. Nuclear

    accumulation of p53 protein is mediated by several localization signals

    and plays a role in tumorigenesis. Mol. Cell Biol.. 10: 6565-6577, 1990.

    on July 2, 2021. © 1997 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/

  • 1997;3:185-191. Clin Cancer Res S M Overholt, T J Liu, D L Taylor, et al. trials.adenovirus-p53-FLAG: a potential marker for gene therapy Head and neck squamous cell growth suppression using

    Updated version

    http://clincancerres.aacrjournals.org/content/3/2/185

    Access the most recent version of this article at:

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/3/2/185To request permission to re-use all or part of this article, use this link

    on July 2, 2021. © 1997 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    http://clincancerres.aacrjournals.org/content/3/2/185http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/3/2/185http://clincancerres.aacrjournals.org/