7
[CANCER RESEARCH 61, 2220 –2225, March 1, 2001] Altered Expression of Ape1/ref-1 in Germ Cell Tumors and Overexpression in NT2 Cells Confers Resistance to Bleomycin and Radiation 1 Kent A. Robertson, Heather A. Bullock, Yi Xu, Renee Tritt, Erika Zimmerman, Thomas M. Ulbright, Richard S. Foster, Lawrence H. Einhorn, and Mark R. Kelley 2 Herman B. Wells Center for Pediatric Research, Departments of Pediatrics [K. A. R., H. A. B., Y. X., R. T., E. Z., M. R. K.], Biochemistry and Molecular Biology [M. R. K.], Pathology [T. M. U.], Medicine [L. H. E.], and Urology [R. S. F.], James Whitcomb Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana 46202 ABSTRACT The human AP endonuclease (Ape1 or ref-1) DNA base excision repair (BER) enzyme is a multifunctional protein that has an impact on a wide variety of important cellular functions including oxidative signaling, tran- scription factor regulation, and cell cycle control. It acts on mutagenic AP (baseless) sites in DNA as a critical member of the DNA BER repair pathway. Moreover, Ape1/ref-1 stimulates the DNA-binding activity of transcription factors (Fos-Jun, nuclear factor-kB, Myb, ATF/cyclic AMP- responsive element binding protein family, HIF-1a, HLF, PAX, and p53) through a redox mechanism and thus represents a novel component of signal transduction processes that regulate eukaryotic gene expression. Ape1/ref-1 has also been shown to be closely linked to apoptosis associated with thioredoxin, and altered levels of Ape1/ref-1 have been found in some cancers. In a pilot study, we have examined Ape1/ref-1 expression by immunohistochemistry in sections of germ cell tumors (GCTs) from 10 patients with testicular cancer of various histologies including seminomas, yolk sac tumors, and malignant teratomas. Ape1/ref-1 was expressed at relatively high levels in the tumor cells of nearly all sections. We hypoth- esized that elevated expression of Ape1/ref-1 is responsible in part for the resistance to therapeutic agents. To answer this hypothesis, we overex- pressed the Ape1/ref-1 cDNA in the GCT cell line NT2/D1 using retroviral gene transduction with the vector LAPESN. Using an oligonucleotide cleavage assay and immunohistochemistry to assess Ape1/ref-1 repair activity and expression, respectively, we found that the repair activity and relative Ape1/ref-1 expression in GCT cell lines are directly related. NT2/D1 cells transduced with Ape1/ref-1 exhibited 2-fold higher AP endonuclease activity in the oligonucleotide cleavage assay, and this was reflected in a 2–3-fold increase in protection against bleomycin. Lesser protection was observed with g-irradiation. We conclude that: (a) Ape1/ ref-1 is expressed at relatively high levels in some GCTs; (b) elevated expression of Ape1/ref-1 in testicular cancer cell lines results in resistance to certain therapeutic agents; and (c) Ape1/ref-1 expression in GCT cell lines determined by immunohistochemistry and repair activity assays parallels the level of protection from bleomycin. We further hypothesize that elevated Ape1/ref-1 levels observed in human testicular cancer may be related to their relative resistance to therapy and may serve as a diagnostic marker for refractory disease. To our knowledge, this is the first example of overexpressing Ape1/ref-1 in a mammalian system result- ing in enhanced protection to DNA-damaging agents. INTRODUCTION Therapy for disseminated testicular cancer/GCT 3 has been success- ful, with 70 – 80% of patients being cured with front-line chemother- apy (1). However, for those 20 –30% of patients with extragonadal primaries or relapsed/refractory disease, the response to therapy is poor, with only 3–30% surviving disease free after second-line agents (1). Active chemotherapeutic agents against GCTs include vinblas- tine, etoposide, bleomycin, cisplatin, cyclophosphamide, and ifos- famide (2). Except for vinblastine and etoposide, each of these agents exerts their toxic effect on tumor cells by directly reacting with DNA, inducing damage including baseless mutagenic AP sites, and ulti- mately cell death. Although little is known about the expression and role of DNA repair systems in GCTs and their response to therapeutic agents, it is of interest that biopsies of GCTs reveal that patients with tumors responding (complete remission) to cisplatin have a higher number of platinum-DNA adducts/tumor cell than patients who re- spond poorly (partial remission; Ref. 3). This observation suggests that resistant cells are able to repair cisplatin-induced DNA damage more effectively than sensitive GCT cells. Preliminary investigations have been performed examining GCTs that are refractory to cisplatin and by establishing cell lines from these cells (4). These initial studies have demonstrated that cisplatin-resist- ant GCT cell lines are able to repair DNA-platinum adducts and cross-links better than cisplatin-responsive cell lines. These two stud- ies suggested a hypothesis that resistance to cisplatin may be related to the relative efficacy of DNA repair (5). More recently, cisplatin adducts have been found to inhibit DNA glycosylases (6), which may alter the BER pathway. The human AP endonuclease (Ape1 or ref-1) DNA BER enzyme is a multifunctional protein that has an impact on a wide variety of important cellular functions, including oxidative signaling, transcrip- tion factor regulation, cell cycle control, and cancer (7). It acts on mutagenic AP (baseless) sites in DNA as a major member of the DNA BER repair pathway. Simple glycosylases, such as methylpurine DNA glycosylase, excise damaged/alkylated bases, resulting in AP sites that are subsequently incised 59 to the AP site by Ape1/ref-1, allowing repair to be completed by deoxyribose phosphatase activity, provided by DNA b-polymerase, to remove the deoxyribose phosphate termini, followed by insertion of the correct base and ligation. Additionally, Ape1/ref-1 is able to repair the 39 phosphate and phosphoglycolate lesions generated in single-strand breaks by ionizing radiation and bleomycin (8). Moreover, Ape1/ref-1 stimulates the DNA-binding activity of transcription factors (Fos-Jun, NFkB, Myb, ATF/cyclic AMP-responsive element binding protein family, HIF-1a, HLF, PAX, and p53) through a redox mechanism and thus represents a novel component of signal transduction processes that regulate eukaryotic gene expression (9 –15). Ape1/ref-1 has also been shown to be closely linked to apoptosis (16), associated with thioredoxin (17, 18), and Received 8/28/00; accepted 12/27/00. 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. 1 Supported in part by NIH Grants CA76643 (to K. A. R., M. R. K., and Y. X.), NS38506 (to M. R. K.), R43 CA83507 (to M. R. K.), P30-DK49218, PO1-CA74295 (to T. M. U. and L. H. E.), and P01-CA75426 (to M. R. K. and R. T.); the Lance Armstrong Foundation (to K. A. R. and M. R. K.); the Gynecologic Oncology Group (to M. R. K.); and the Riley Memorial Association (to K. A. R. and M. R. K.). 2 To whom requests for reprints should be addressed, at The Herman B. Wells Center for Pediatric Research, Riley Hospital for Children, Room 2600, 702 Barnhill Drive, Indianapolis, IN 46202. 3 The abbreviations used are: GCT, germ cell tumor; Ape1/ref-1, apurinic/apyrimidinic endonuclease; BER, base excision repair; IHC, immunohistochemistry; AP, apurinic/ apyrimidinic; THF, tetrohydrofuran; NT2, NTERA-2/D1 human embryonal carcinoma. 2220 on April 2, 2021. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Altered Expression of Ape1/ref-1 in Germ Cell Tumors and ... · component of signal transduction processes that regulate eukaryotic gene expression (9–15). Ape1/ref-1 has also been

  • Upload
    others

  • View
    1

  • Download
    0

Embed Size (px)

Citation preview

  • [CANCER RESEARCH 61, 2220–2225, March 1, 2001]

    Altered Expression of Ape1/ref-1 in Germ Cell Tumors and Overexpression in NT2Cells Confers Resistance to Bleomycin and Radiation1

    Kent A. Robertson, Heather A. Bullock, Yi Xu, Renee Tritt, Erika Zimmerman, Thomas M. Ulbright,Richard S. Foster, Lawrence H. Einhorn, and Mark R. Kelley2

    Herman B. Wells Center for Pediatric Research, Departments of Pediatrics [K. A. R., H. A. B., Y. X., R. T., E. Z., M. R. K.], Biochemistry and Molecular Biology [M. R. K.],Pathology [T. M. U.], Medicine [L. H. E.], and Urology [R. S. F.], James Whitcomb Riley Hospital for Children, Indiana University School of Medicine, Indianapolis, Indiana46202

    ABSTRACT

    The human AP endonuclease (Ape1 or ref-1) DNA base excision repair(BER) enzyme is a multifunctional protein that has an impact on a widevariety of important cellular functions including oxidative signaling, tran-scription factor regulation, and cell cycle control. It acts on mutagenic AP(baseless) sites in DNA as a critical member of the DNA BER repairpathway. Moreover, Ape1/ref-1 stimulates the DNA-binding activity oftranscription factors (Fos-Jun, nuclear factor-kB, Myb, ATF/cyclic AMP-responsive element binding protein family, HIF-1a, HLF, PAX, and p53)through a redox mechanism and thus represents a novel component ofsignal transduction processes that regulate eukaryotic gene expression.Ape1/ref-1 has also been shown to be closely linked to apoptosis associatedwith thioredoxin, and altered levels of Ape1/ref-1 have been found in somecancers. In a pilot study, we have examined Ape1/ref-1 expression byimmunohistochemistry in sections of germ cell tumors (GCTs) from 10patients with testicular cancer of various histologies including seminomas,yolk sac tumors, and malignant teratomas. Ape1/ref-1 was expressed atrelatively high levels in the tumor cells of nearly all sections. We hypoth-esized that elevated expression of Ape1/ref-1 is responsible in part for theresistance to therapeutic agents. To answer this hypothesis, we overex-pressed the Ape1/ref-1 cDNA in the GCT cell line NT2/D1 using retroviralgene transduction with the vector LAPESN. Using an oligonucleotidecleavage assay and immunohistochemistry to assess Ape1/ref-1 repairactivity and expression, respectively, we found that the repair activity andrelative Ape1/ref-1 expression in GCT cell lines are directly related.NT2/D1 cells transduced with Ape1/ref-1 exhibited 2-fold higher APendonuclease activity in the oligonucleotide cleavage assay, and this wasreflected in a 2–3-fold increase in protection against bleomycin. Lesserprotection was observed withg-irradiation. We conclude that: (a) Ape1/ref-1 is expressed at relatively high levels in some GCTs; (b) elevatedexpression of Ape1/ref-1 in testicular cancer cell lines results in resistanceto certain therapeutic agents; and (c) Ape1/ref-1 expression in GCT celllines determined by immunohistochemistry and repair activity assaysparallels the level of protection from bleomycin. We further hypothesizethat elevated Ape1/ref-1 levels observed in human testicular cancer maybe related to their relative resistance to therapy and may serve as adiagnostic marker for refractory disease. To our knowledge, this is thefirst example of overexpressing Ape1/ref-1 in a mammalian system result-ing in enhanced protection to DNA-damaging agents.

    INTRODUCTION

    Therapy for disseminated testicular cancer/GCT3 has been success-ful, with 70–80% of patients being cured with front-line chemother-apy (1). However, for those 20–30% of patients with extragonadalprimaries or relapsed/refractory disease, the response to therapy ispoor, with only 3–30% surviving disease free after second-line agents(1). Active chemotherapeutic agents against GCTs include vinblas-tine, etoposide, bleomycin, cisplatin, cyclophosphamide, and ifos-famide (2). Except for vinblastine and etoposide, each of these agentsexerts their toxic effect on tumor cells by directly reacting with DNA,inducing damage including baseless mutagenic AP sites, and ulti-mately cell death. Although little is known about the expression androle of DNA repair systems in GCTs and their response to therapeuticagents, it is of interest that biopsies of GCTs reveal that patients withtumors responding (complete remission) to cisplatin have a highernumber of platinum-DNA adducts/tumor cell than patients who re-spond poorly (partial remission; Ref. 3). This observation suggeststhat resistant cells are able to repair cisplatin-induced DNA damagemore effectively than sensitive GCT cells.

    Preliminary investigations have been performed examining GCTsthat are refractory to cisplatin and by establishing cell lines from thesecells (4). These initial studies have demonstrated that cisplatin-resist-ant GCT cell lines are able to repair DNA-platinum adducts andcross-links better than cisplatin-responsive cell lines. These two stud-ies suggested a hypothesis that resistance to cisplatin may be relatedto the relative efficacy of DNA repair (5). More recently, cisplatinadducts have been found to inhibit DNA glycosylases (6), which mayalter the BER pathway.

    The human AP endonuclease (Ape1 or ref-1) DNA BER enzyme isa multifunctional protein that has an impact on a wide variety ofimportant cellular functions, including oxidative signaling, transcrip-tion factor regulation, cell cycle control, and cancer (7). It acts onmutagenic AP (baseless) sites in DNA as a major member of the DNABER repair pathway. Simple glycosylases, such as methylpurine DNAglycosylase, excise damaged/alkylated bases, resulting in AP sites thatare subsequently incised 59to the AP site by Ape1/ref-1, allowingrepair to be completed by deoxyribose phosphatase activity, providedby DNA b-polymerase, to remove the deoxyribose phosphate termini,followed by insertion of the correct base and ligation. Additionally,Ape1/ref-1 is able to repair the 39phosphate and phosphoglycolatelesions generated in single-strand breaks by ionizing radiation andbleomycin (8). Moreover, Ape1/ref-1 stimulates the DNA-bindingactivity of transcription factors (Fos-Jun, NFkB, Myb, ATF/cyclicAMP-responsive element binding protein family, HIF-1a, HLF, PAX,and p53) through a redox mechanism and thus represents a novelcomponent of signal transduction processes that regulate eukaryoticgene expression (9–15). Ape1/ref-1 has also been shown to be closelylinked to apoptosis (16), associated with thioredoxin (17, 18), and

    Received 8/28/00; accepted 12/27/00.The costs of publication of this article were defrayed in part by the payment of page

    charges. This article must therefore be hereby markedadvertisementin accordance with18 U.S.C. Section 1734 solely to indicate this fact.

    1 Supported in part by NIH Grants CA76643 (to K. A. R., M. R. K., and Y. X.),NS38506 (to M. R. K.), R43 CA83507 (to M. R. K.), P30-DK49218, PO1-CA74295 (toT. M. U. and L. H. E.), and P01-CA75426 (to M. R. K. and R. T.); the Lance ArmstrongFoundation (to K. A. R. and M. R. K.); the Gynecologic Oncology Group (to M. R. K.);and the Riley Memorial Association (to K. A. R. and M. R. K.).

    2 To whom requests for reprints should be addressed, at The Herman B. Wells Centerfor Pediatric Research, Riley Hospital for Children, Room 2600, 702 Barnhill Drive,Indianapolis, IN 46202.

    3 The abbreviations used are: GCT, germ cell tumor; Ape1/ref-1, apurinic/apyrimidinicendonuclease; BER, base excision repair; IHC, immunohistochemistry; AP, apurinic/apyrimidinic; THF, tetrohydrofuran; NT2, NTERA-2/D1 human embryonal carcinoma.

    2220

    on April 2, 2021. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • altered levels of Ape1/ref-1 have been found in some cancers (19–21).More recently, in a B-lymphocyte system, Ape1/ref-1 has demon-strated a response to reactive oxygen species by rapid translocationfrom the cytoplasm to the nucleus (22).

    In a pilot study, we found Ape1/ref-1 to be expressed at a relativelyhigh level in a small group of GSTs (23). Because of the multifacetedrole of Ape1/ref-1 in cells, we have begun to try and determinewhether the elevated level of Ape1/ref-1 in GCTs is responsible for anincrease in repair, redox, or both activities. We hypothesized thatelevated expression of Ape1/ref-1 is responsible in part for the resist-ance to therapeutic agents. To answer this hypothesis, we overex-pressed the Ape1/ref-1 cDNA in the GCT cell line NT2/D1 usingretroviral gene transduction with LAPESN. Using an oligonucleotidecleavage assay and IHC to assess Ape1/ref-1 repair activity andexpression, respectively, we found that the repair activity and relativeApe1/ref-1 expression in GCT cell lines to be directly related.NT2/D1 cells transduced with Ape1/ref-1 exhibited 2-fold higher APendonuclease activity in the oligonucleotide cleavage assay, and thiswas reflected in a 2–3-fold increase in protection against bleomycin.Lesser protection was observed withg-irradiation. We conclude thatelevated expression of Ape1/ref-1 in testicular cancer cell lines resultsin resistance to certain therapeutic agents. We further hypothesize thatelevated Ape1/ref-1 levels observed in human testicular cancer maybe related to their relative resistance to therapy and may serve as adiagnostic marker for refractory disease. This is the first example ofoverexpressing Ape1/ref-1 in a mammalian model system that leads tocellular protection from chemotherapeutic agents.

    MATERIALS AND METHODS

    Patients and Tumors. Tissue sections of biopsy material from treated anduntreated patients with disseminated GCTs were obtained from the IndianaUniversity Medical Center, University Hospital, under an Indiana UniversityInstitutional Review Board approved protocol (IU Study No. 9908-47) as 4%buffered formaldehyde-fixed tissues embedded in paraffin blocks, which weresectioned at 3mm and fixed onto slides. Diagnosis was made from morpho-logical examination of H&E-stained sections of biopsy material.

    IHC. Tissue sections were stained for Ape1/ref-1 expression using ananti-Ape1 monoclonal antibody that has been characterized extensively (19,20) and is available commercially (Novus Biologicals, Littleton, CO). Thestaining process used was identical to that described previously (19, 20), withthe exception that a Dako Universal Staining System was used to automate theprocess (Dako Corp., Carpinteria, CA). Sections were treated with a 10-minincubation in 3% H2O2 to block endogenous peroxidases, incubated with theanti-Ape1 antibody (1:1000) for 25 min, the biotinylated goat antimouse IgGsecondary antibody for 10 min, streptavidin-horseradish peroxidase for 10 min,and diaminobenzidine for 5 min, per Dako recommendations and empiricdetermination. Preimmune IgG was used as a control for antibody specificityin place of the antihuman Ape1 antibody at a concentration of 50mg/ml. Noantigen unmasking techniques were used, nor are they necessary with thisantibody.

    Cell Lines. NT2 cells and the PG13 GALV-based retrovirus packaging cellline were obtained from the American Type Culture Collection (Rockville,MD). The 833K GCT cell line was obtained from George Sledge (IndianaUniversity). Both cell lines were maintained as adherent cells in polystyrene,six-well Costar tissue culture trays (Costar, Corning, NY) in DMEM tissueculture media (Life Technologies, Inc., Rockville, MD) plus 10% Cosmic Calfserum (Hyclone, Logan, UT) plus antibiotics. Cells were maintained in ahumidified incubator at 37°C in an atmosphere of 5% CO2 and 95% air.

    Molecular Analysis. Southern, Northern, and Western blotting were per-formed as described previously (16, 20). Southern blots were probed with a32P-labeled random primed 1-kb human Ape1/ref-1 cDNA. Northern blotswere hybridized with32P-labeled Ape1/ref-1, Neo, and humanb-actin, asdescribed previously (16, 20).

    Retroviral Construction, Production, and Infection of Cell Lines. The1-kb human Ape1/ref-1 cDNA was cloned into theEcoRI (59) andXhoI (39)

    sites of the retroviral vector LXSN as described previously (16). Virus wasgenerated from the LXSN plasmid harboring Ape1/ref-1 (now designatedLAPESN), as performed previously (24). Briefly, 10mg of agarose gel purifiedLAPESN were transfected into PE501 producer cells, followed by selection inG418 (Life Technologies, Inc.) at 1 mg/ml. Supernatants were harvested andused to infect PG13 GALV-based amphotrophic packaging cells (25)and selected in G418. Total RNA was extracted from the G418-resistant cellsand analyzed by Northern blot hybridization with an Ape1/ref-1 probe todemonstrate the 1.7-kb Ape1/ref-1 retroviral transcript. Supernatants fromG418-resistant PG131 LAPESN cells were used to transduce NT2 cells. Afterselection in G418 (1 mg/ml), individual clones were isolated and expanded.Screening by Southern blot analysis revealed two clones with distinct singleinserts that were selected for further study. NT2 cells transduced with theempty LXSN vector were produced in a similar manner as controls.

    Cell Culture Studies. NT2, NT2-LXSN, and NT2-LAPESN cells wereseeded (3–53 10 4 cells/well) in six-well trays and allowed to adhereovernight. The following day, cells were treated with bleomycin (Blenoxane;Bristol Myers Squib Company, Princeton, NJ) as indicated or radiation. Bleo-mycin was dissolved in sterile water (250mg/ml) and filter-sterilized througha 0.22mm filter prior to being added to cultures, and radiation was deliveredfrom a Cesium 137 irradiator at a rate of 11.1 cGy/min. After 3, 5, and 7 days,control and treated cells were enumerated by trypsinizing the adherent cells,pipetting up and down to generate a single-cell suspension in trypan blue, andcounting using a hemocytometer.

    The determination of AP endonuclease activity was performed using anoligonucleotide cleavage assay as described previously (20, 26, 27). Briefly,cell extracts were incubated with a 59-32 P-end-labeled 26-mer oligonucleotidecontaining a single THF artificial AP site at position 14, which, in the presenceof AP endonuclease activity, cleaves the32P-labeled 26-mer to a 14-mer. Theabasic analogue is resistant to cleavage by 39-acting AP lyase activity, whichis generally possessed by DNA glycosylases/AP lyases. Therefore, this assayis specific for Ape1/ref-1 activity in cells. Reaction mixtures (10ml) containingcell extracts of interest, 2.5 pmol of 5932P end-labeled, double-stranded THFoligonucleotide, 50 mM HEPES, 50 mM KCl, 10 mM MgCl2, 1 mg/ml BSA, and0.05% Triton X-100 (pH 7.5) were allowed to proceed for 15 min in a 37°Cwater bath. Reactions were halted by adding 10ml of 96% formamide, 10 mMEDTA, xylene cytanol, and bromphenol blue. AP assay products (5ml) wereseparated on a 20% polyacrylamide gel containing 7M urea. Gels werewrapped in saran wrap and exposed to film for visualization. The amount of14-mer to 26-mer was determined by scanning the exposed film into SigmaScan (Jandel Scientific, San Rafael, CA) for analysis.

    RESULTS

    Expression of Human Ape1/ref-1 in GCTs.We examined theexpression of Ape1/ref-1 in 10 GCT cases from a variety of histolog-ical subtypes. Four tumors were yolk sac tumors, four were metastaticteratomas, including one with primitive neuroectodermal tumor ele-ments and one with cystic trophoblastic elements, and two wereseminomas. Normal cells, including fibroblasts and vascular endothe-lium, had a predominant nuclear staining pattern with occasional cellsdemonstrating a mixed nuclear-cytoplasmic staining pattern (Fig. 1).Infiltrating lymphocytes had little Ape1/ref-1 expression. Tumor cellsidentified in each section displayed an increased level of expression ofnuclear Ape1/ref-1 compared with stromal elements on the sameslides (Fig. 1 and Table 1). The distribution of Ape1/ref-1 staining inthe GCTs was predominantly nuclear in 50% of the cases and mixednuclear-cytoplasmic in the other 50% (Table 1). There appeared to bea trend for the more differentiated subtypes (teratomas) to have lesscytoplasmic expression of Ape1/ref-1 compared with other subtypes(Table 1). Given the small number of samples examined in this pilotstudy, no statistical analysis was performed; however, a larger trialhas been initiated that should permit more conclusive analysis ofApe1/ref-1 expression in GCTs and its relationship to clinical param-eters, such as relapse and chemotherapy resistance.

    Retroviral Gene Transfer of Ape1/ref-1 into NT2 Cells. Asdetailed in “Materials and Methods,” we inserted a cDNA fragment

    2221

    APE1 PROTECTS NT2 CELLS FROM BLEOMYCIN/RADIATION

    on April 2, 2021. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • harboring the complete coding sequence of human Ape1/ref-1 into theLXSN retroviral vector (Ref. 28; Fig. 2A). This vector includes aconvenient cloning site downstream from the Moloney murine leuke-mia virus long terminal repeat as well as the neomycin phosphotrans-ferase gene (neo) for use as a selectable marker. NT2 clones trans-duced with LAPESN were selected in 10-cm dishes and isolated withcloning rings. Screening by Southern blot (Ape1/ref-1 probe) identi-

    fied two clones (data not shown) with single inserts, which were usedfor further study. It was difficult to distinguish on Northern blotanalysis the mRNA from NT2, NT2-LXSN, and NT2-LAPESNclones 1 and 2 of the endogenous Ape1/ref-1 (1.6 kb) from theretroviral-derived transcript (1.7 kb); however, probing forneo re-vealed the full-length, 2.9-kb LXSN transcript in the LXSN vector-only transduced cells and the truncated 1.6-kb Neo transcript from the

    Table 1 Ten clinical GCT samples scored for Ape1/ref-1 expression by IHC including both tumor cells and surrounding normal tissuesa

    GCT no. Histology% APE-tumor

    cells% APE-

    Tumor Nuclei% APE-TumorCell Cytoplasm

    Tumor nuclearintensity (1–3)

    Tumor cytoplasmicintensity (1–3)

    % APE-Stromal Cells

    Stromal cellintensity (1–3)

    011 Solid and glandular yolk sac tumor 90% 90% 10% 2–3 1 50% 2012 Seminoma 95% 80% 80% 1–2 2 40% 1–2013 Glandular yolk sac tumor 90% .95% 30% 3 1 60% 2–3014 Solid yolk sac tumor 90% 90% 50% 2–3 1 70% 3015 Teratoma with PNET elements 75% 80% 0% 3 0 70% 3016 Seminoma 80% 60% 50% 2–3 2 50% 2017 Myxoid yolk sac tumor 90% 90% 1% 3 2 40% 2–3018 Cystic trophoblast 90% 90% 40% 3 2 20% 2–3019 Teratoma 70% 70% 0% 3 0 50% 2–3020 Teratoma 60% 60% 0% 3 0 70% 3

    a Any appreciable staining is considered positive and graded for intensity: 1, barely detectable; 2, fine granules diffusely present throughout the nucleus or cytoplasm; 3, dark coarsegranules are observed.

    Fig. 1. H&E staining (A,C, andE) and Ape1/ref-1 IHC (B, D, and F) of a glandular yolk sactumor, 310 and with inset, 340 (A and B); aseminoma,340 (C andD); and a malignant tera-toma, 340 (E and F). Sections demonstrate therelatively intense Ape1/ref-1 staining of tumor cells(arrows) from different GCTs compared with stromalelements (arrowheads), including fibroblasts, vascu-lar elements (B), and infiltrating lymphocytes (D).

    2222

    APE1 PROTECTS NT2 CELLS FROM BLEOMYCIN/RADIATION

    on April 2, 2021. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • LAPESN-expressing cells (Fig. 2B). The absence of a full-lengthLAPESN transcript likely results from an occult polyadenylationsignal within the 39sequence of the Ape1/ref-1 cDNA, similar to thetranscript pattern observed with other LXSN constructs (24).

    Expression of Human Ape1/ref-1 in GCT Cell Lines. Immuno-histochemical staining experiments using the antihuman Ape1 anti-body were performed on the human GCT cell lines NTD2 and 833Kto begin to characterize the role that Ape1/ref-1 might play in the

    response of GCTs to therapeutic agents. The NTD2 cell line is derivedfrom a human testicular embryonal carcinoma/teratoma lung metas-tasis (29), and the 833K cell line is derived from an abdominalmetastasis of a human testicular GCT with teratoma, embryonalcarcinoma, choriocarcinoma, and seminoma elements (30). In bothcell lines, there was a predominance of nuclear staining with littlecytoplasmic staining (Fig. 3A), similar to the pattern described abovein sections of primary teratomas (Table 1). The nuclei displayed adistinctive punctate staining pattern that we observed previously incervical carcinoma cells (19). The NT2 cells overexpressing Ape1/ref-1 demonstrated an elevated level of nuclear staining for Ape1/ref-1 again with little cytoplasmic staining, similar to the wild-typeNT2 cells (Fig. 3A). Vector-only transduced cells were identical to thewild-type NT2 cells in Ape1/ref-1 staining, and morphologically, thetransduced cells (LXSN and LAPESN) were indistinguishable fromthe wild-type cells (Fig. 3A).

    Ape1/ref-1 Repair Function in GCT Cell Lines. To determinewhether the increased expression of Ape1/ref-1 detected by IHCcorrelated with increased AP site repair activity, we used an oligonu-cleotide cleavage assay (20, 27). The assay uses a radiolabeled oli-gonucleotide (26-mer; Fig. 3B) containing an artificial THF AP site,which when cleaved by AP endonuclease produces a labeled 14-mer(Fig. 3B). Sequential dilutions of cell extracts from each of the celltypes were assayed for endonuclease activity and are shown in Fig.3B. The increasing expression of Ape1/ref-1 noted in parental NT2cells, 833K cells, and NT2-LAPESN cells (Fig. 3A) was reflected inincreasing AP endonuclease activity, as measured with the oligonu-cleotide cleavage assay. Thus, Ape1/ref-1 repair activity appears tocorrelate with the degree of Ape1/ref-1 expression by IHC.

    Ape1/ref-1 Overexpression Protects GCT Cells from Bleomycinand Radiation. To determine whether Ape1/ref-1 expression corre-lated with sensitivity to therapeutic agents, we quantitated the AP-sitecleavage activity of parental NT2 cells and NT2-LAPESN clones 1and 2 (Fig. 4,A–C). Both clones as well as the bulk-infected NT2 cellsfrom which the clones were selected displayed a 2-fold higher endo-nuclease activity compared with NT2 cells (Fig. 4D). We then exam-ined the response of NT2 and NT2-LAPESN cells to bleomycin, anantitumor antibiotic known for its effectiveness in treating GCTs.Cells transduced with the empty vector, LXSN, and the parental NT2cells responded similarly to treatment (Fig. 5). The NT2 cells over-

    Fig. 2. Retroviral Ape1/ref-1 construct and Northern blot expression.A, schematic ofthe LXSN retroviral vector and the LAPESN construct.B, Northern blots of NT2 cells(Lane 1), NT2-LXSN (Lane 2), and NT2-LAPESN clones 1 and 2 (Lanes 3and 4,respectively). Probes are as indicated.

    Fig. 3. Ape1/ref-1 expression (A) and activity(B) in NT2 (1), 833K (2), and NT2-LAPESN (3)cells. A, IHC staining of Ape1/ref-1 expressiondemonstrating increased expression in 833K cells(2) compared with wild-type NT2 cells (1) andfurther increased expression in NT2-LAPESN cells(3). NT2-LXSN cells were identical to parentalNT2 cells (data not shown).B, AP endonucleaseactivity using the oligonucleotide cleavage assay(see “Materials and Methods”) with cell extractsfrom NT2 (1), 833K (2), and NT2-LAPESN (3)cells. Sequential dilutions of cell extracts (Lanes2–8 were assayed for AP site cleavage activity).Lane 1, recombinant Ape1/ref-1 as a positive con-trol.

    2223

    APE1 PROTECTS NT2 CELLS FROM BLEOMYCIN/RADIATION

    on April 2, 2021. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • expressing Ape1/ref-1 demonstrated a 2-fold increase in protectionfrom bleomycin in a dose-dependent fashion (Fig. 5A), paralleling the2-fold increase in endonuclease activity observed in the oligonucleo-tide cleavage assay (Fig. 4). A similar trend for protection fromg-irradiation was observed but with not quite the same degree ofprotection that was afforded to bleomycin (Fig. 5B). Thus, the APendonuclease activity of Ape1/ref-1 as measured by the oligonucleo-tide cleavage assay appears to reflect the degree of protection frombleomycin and radiation.

    DISCUSSION

    We have made the observation that a key enzyme in the DNA BERpathway, Ape1/ref-1, is expressed at a high level in GCTs of varyinghistological subtypes. The implications of this observation are thatcells expressing a higher level of Ape1/ref-1 may be more efficient atrepair of chemotherapy-induced damage and thereby become resistantto the therapeutic agents. To approach this question, we used thehuman embryonal carcinoma cell line, NT2, and overexpressed Ape1/ref-1 using retroviral gene transduction. Although alteration in Ape1/ref-1 levels in various cell types has resulted in altered sensitivity tocytotoxic agents, few studies have been performed in mammaliancells, and almost none have been performed in human-derived cells(26, 31–33). IHC has been an effective technique to identify andcharacterize expression of molecular cellular components in clinicalsamples, particularly for Ape1/ref-1 (20, 34–36). IHC for Ape1/ref-1expression in GCT clinical samples in this study clearly demonstratedan elevated expression of Ape1/ref-1. To determine whether thiselevated expression was meaningful in terms of repair, we evaluatedthe expression of Ape1/ref-1 in transduced NT2 cells using IHC andcorrelated the expression with BER activity in the same cells using anoligonucleotide cleavage assay. Overexpression of Ape1/ref-1 re-

    sulted in a 2-fold increase in repair activity, which correlated with a2–3-fold higher protection against bleomycin. Although we frequentlysee results in molecular biology on the level of orders of magnitude,a 2-fold increase in the resistance of a tumor to therapy is significant.There was less of an effect of protection with radiation likely becausedamage induced by radiation is complex, with many different types ofDNA lesions being produced, many of which are not amenable torepair via the BER pathway. Bleomycin produces single-strand breaksand 49-oxidized AP sites, which are recognized and repaired byApe1/ref-1 primarily through its phosphodiesterase function. Further-more, some of the damage created by bleomycin results in oxidizedAP sites in the form of C-4-keto-C-1-aldehydes in the intact DNAstrands, as well as 39-phosphoglycolate esters that terminate strandbreaks (8). In previous studies, Ape1/ref-1 catalyzed incision at theC-4-keto-C-1-aldehyde sites at a rate similar to its hydrolytic incisionof AP sites. Ape1/ref-1 also incised DNA at hydrolyzed 39-phospho-glycolates, albeit more slowly than incisions at C-4-keto-C-1-alde-hydes (8). Other studies have documented similar types of damagecaused by bleomycin or ionizing radiation (8, 37–39).g-Irradiationproduces a variety of damage to DNA, including oxidative damage tobases resulting in base excision and the formation of mutagenic/cytotoxic AP sites, which are substrates for Ape1/ref-1 (33). Thus, weconclude that elevated expression of Ape1/ref-1 in GCTs likely con-tributes to resistance to therapeutic agents that induce damage respon-sive to BER.

    However, we must recognize that Ape1/ref-1 is a complex enzyme

    Fig. 5. Overexpression of Ape1/ref-1 protects NT2 cells from bleomycin and radiation.Cell numbers expressed as percentage of untreated controls 5–7 days after exposure toincreasing doses of bleomycin (A) or radiation (B).Bars, 95% confidence interval (SE).

    Fig. 4. Quantification of Ape/ref-1 endonuclease activity.A–C, sequential dilutions ofcell extracts (Lanes 2–9) of NT2-LXSN (A), NT2-LAPESN #1 (B), and NT2-LAPESN #2(C) demonstrating higher AP endonuclease activity in the Ape1/ref-1 transduced clones.Lane 1, recombinant Ape1/ref-1 as a positive control.D, quantification of AP endonu-clease activity fromLane 5of A–C. Percentages indicated are the means of three differentexperiments reflecting the percentage of cleavage of the radiolabeled AP site-containingoligonucleotide.

    2224

    APE1 PROTECTS NT2 CELLS FROM BLEOMYCIN/RADIATION

    on April 2, 2021. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • with multiple functions including a well-documented redox function (7).Although Ape1/ref-1 appears to protect from bleomycin andg-irradiationpresumably by means of repair activity, another aspect, and somewhatcontrary to these results, is its potential role in augmentation of theresponse to certain therapeutic agents. Ape1/ref-1 involvement in activa-tion of p53 and induction of cyclin G may be integral to the apoptoticresponse to cisplatin in GCTs (9, 40). The high level expression ofApe1/ref-1 in GCTs in conjunction with its role in activation of apoptosisthrough p53/cyclin G may account in part for the sensitivity of GCTs totherapy. Additionally, elevation of Ape1/ref-1 may be important in main-taining the malignant phenotype through redox activation of transcriptionfactors such as Fos, Jun, and HIF-1a (7). One way to sort out these issuesis the use of site-specific Ape1/ref-1 mutants to characterize the role ofthe repair/redox domains in response of GCTs to specific therapeuticagents. Mechanistic studies should be complemented with the examina-tion of Ape1/ref-1 expression in a large sample of GCT patients todetermine the correlation, if any, of expression with response to therapy.We have an ongoing protocol to collect such data retrospectively andprospectively, with the objective to analyze the relationship of Ape1/ref-1expression to histological subtypes of GCTs, risk of relapse, response totherapy, and role in tumor progression. Other questions also exist, suchas: Does the cytoplasmic distribution of Ape1/ref-1 observed in someGCTs result from an alteration in the nuclear localization signal harboredin the 59-end of Ape1/ref-1? How does this change in distribution affectrepair and redox function? In one primary teratoma of the testis, weexamined only the intratubular GCT cells marked with antihuman Ape1/ref-1 antibody, sparing the rest of the normal cellular elements, suggest-ing that in certain settings, staining with anti-Ape1 may be useful indetecting tumor cells. Similar observations have been made in stainingsections of cervical carcinomain situ for Ape1/ref-1 (19).

    ACKNOWLEDGMENTS

    We thank Steve Parsons for technical assistance.

    REFERENCES

    1. Nichols, C. R., Roth, B. J., Loehrer, P. J., Williams, S. D., and Einhorn, L. H. Salvagechemotherapy for recurrent germ cell cancer. Semin. Oncol.,21: 102–108, 1994.

    2. Broun, E. R., Nichols, C. R., Gize, G., Cornetta, K., Hromas, R. A., Schacht, B., andEinhorn, L. H. Tandem high dose chemotherapy with autologous bone marrowtransplantation for initial relapse of testicular germ cell cancer. Cancer (Phila.),79:1605–1610, 1997.

    3. Reed, E., Ozols, R. F., Tarone, R., Yuspa, S. H., and Poirier, M. C. The measurementof cisplatin-DNA adduct levels in testicular cancer patients. Carcinogenesis (Lond.),9: 1909–1911, 1988.

    4. Reilly, P. A., Heerema, N. A., Sledge, G. W., Jr., and Palmer, C. G. Unusualdistribution of chromosome 12 in a testicular germ-cell tumor cell line (833K) and itscisplatin-resistant derivative (64CP9). Cancer Genet. Cytogenet.,68: 114–121, 1993.

    5. Kurie, J. M., Bosl, G. J., and Dmitrovsky, E. The genetic and biologic aspects oftreatment response and resistance in male germ cell cancer. Semin. Oncol.,19:197–205, 1992.

    6. Kartalou, M., Samson, L. D., and Essigmann, J. M. Cisplatin adducts inhibit 1N6-ethenoadenine repair by interacting with the human 3-methyladenine DNA gly-cosylase. Biochemistry,39: 8032–8040, 2000.

    7. Evans, A. R., Limp-Foster, M., and Kelley, M. R. Going APE over ref-1. Mutat. Res.,461: 83–108, 2000.

    8. Xu, Y. J., Kim, E. Y., and Demple, B. Excision of C-49-oxidized deoxyribose lesionsfrom double-stranded DNA by human apurinic/apyrimidinic endonuclease (Ape1protein) and DNA polymeraseb. J. Biol. Chem.,273: 28837–28844, 1998.

    9. Jayaraman, L., Murthy, K. G. K., Zhu, C., Curran, T., Xanthoudakis, S., and Prives,C. Identification of redox/repair protein Ref-1 as a potent activator ofp53. GenesDev., 11: 558–570, 1997.

    10. Gaiddon, C., Moorthy, N. C., and Prives, C. Ref-1 regulates the transactivation andpro-apoptotic functions ofp53 in vivo. EMBO J.,18: 5609–5621, 1999.

    11. Xanthoudakis, S., Miao, G., Wang, F., Pan, Y. C., and Curran, T. Redox activation ofFos-Jun DNA binding activity is mediated by a DNA repair enzyme. EMBO J.,11:3323–3335, 1992.

    12. Huang, R. P., and Adamson, E. D. Characterization of the DNA-binding properties ofthe early growth response-1 (Egr-1) transcription factor: evidence for modulation bya redox mechanism. DNA Cell Biol.,12: 265–273, 1993.

    13. Mansouri, A., Hallonet, M., and Gruss, P.Paxgenes and their roles in cell differen-tiation and development. Curr. Opin. Cell Biol.,8: 851–857, 1996.

    14. Tell, G., Pellizzari, L., Cimarosti, D., Pucillo, C., and Damante, G. Ref-1 controls pax-8DNA-binding activity. Biochem. Biophys. Res. Commun.,252: 178–183, 1998.

    15. Lando, D., Pongratz, I., Poellinger, L., and Whitelaw, M. L. A redox mechanismcontrols differential DNA binding activities of hypoxia-inducible factor (HIF) 1a andthe HIF-like factor. J. Biol. Chem.,275: 4618–4627, 2000.

    16. Robertson, K. A., Hill, D. P., Xu, Y., Liu, L., VanEpps, S., Hockenbery, D. M., Park,J. R., Wilson, T. M., and Kelley, M. R. Down-regulation of AP endonucleaseexpression is associated with the induction of apoptosis in differentiating myeloidleukemia cells. Cell Growth Differ.,8: 443–449, 1997.

    17. Hirota, K., Matsui, M., and Yodoi, J. AP-1 transcriptional activity is regulated by adirect association between thioredoxin and Ref-1. Proc. Natl. Acad. Sci. USA,94:3633–3638, 1997.

    18. Qin, J., Clore, G. M., Kennedy, W. P., Kuszewski, J., and Gronenborn, A. M. Thesolution structure of human thioredoxin complexed with its target from Ref-1 revealspeptide chain reversal. Structure,4: 613–620, 1996.

    19. Xu, Y., Moore, D. H., Broshears, J., Liu, L., Wilson, T. M., and Kelley, M. R. Theapurinic/apyrimidinic endonuclease (APE/ref-1) DNA repair enzyme is elevatedin premalignant and malignant cervical cancer. Anticancer Res.,17: 3713–3719,1997.

    20. Moore, D. H., Michael, H., Tritt, R., Parsons, S. H., and Kelley, M. R. Alterations inthe expression of the DNA repair/redox enzyme APE/ref-1 in epithelial ovariancancers. Clin. Cancer Res.,6: 602–609, 2000.

    21. Kelley, M. R., Moore, D. H., Cheng, L., Foster, R., Michael, H., Xu, Y., Parsons, S.,Tritt, R., and Koch, M. Altered expression of the multifunctional DNA base excisionrepair and redox enzyme ape/ref-1 in ovarian and prostate cancers.In: DNA RepairDefects, AACR Special Conference, San Diego, CA, 2000.

    22. Tell, G., Zecca, A., Pellizzari, L., Spessotto, P., Colombatti, A., Kelley, M. R.,Damante, G., and Pucillo, C. An “environment to nucleus” signaling system operatesin B lymphocytes: redox status modulates BSAP/Pax-5 activation through Ref-1nuclear translocation. Nucleic Acids Res.,28: 1099–1105, 2000.

    23. Kelley, M. R., Xu, Y., Tritt, R., and Robertson, K. A. The multifunctional DNA baseexcision repair and redox protein, AP endonuclease (APE/ref-1), and its role in germcell tumors. In: I. A. W. G. Jones, P. Harnden, and J. K. Joffe (eds.), Germ CellTumors IV, Vol. 4, pp. 81–86. London: John Libbey & Co., 1998.

    24. Robertson, K. A., Emami, B., Mueller, L., and Collins, S. J. Multiple members of theretinoic acid receptor family are capable of mediating the granulocytic differentiationof HL-60 cells. Mol. Cell. Biol.,12: 3743–3749, 1992.

    25. Bauer, T. R., Jr., Miller, A. D., and Hickstein, D. D. Improved transfer of theleukocyte integrin CD18 subunit into hematopoietic cell lines by using retroviralvectors having a gibbon ape leukemia virus envelope. Blood,86: 2379–2387, 1995.

    26. Hansen, W. K., Deutsch, W. A., Yacoub, A., Xu, Y., Williams, D. A., and Kelley,M. R. Creation of a fully functional human chimeric DNA repair protein. J. Biol.Chem.,273: 756–762, 1998.

    27. Yacoub, A., Kelley, M. R., and Deutsch, W. A. The DNA repair activity of humanredox/repair protein APE/Ref-1 is inactivated by phosphorylation. Cancer Res.,57:5457–5459, 1997.

    28. Miller, A. D., and Rosman, G. J. Improved retroviral vectors for gene transfer andexpression. Biotechniques,7: 980–982, 984–986, 989–990, 1989.

    29. Andrews, P. W., Damjanov, I., Simon, D., Banting, G. S., Carlin, C., Dracopoli,N. C., and Fogh, J. Pluripotent embryonal carcinoma clones derived from the humanteratocarcinoma cell line Tera-2. Lab. Investig.,50: 147–162, 1984.

    30. Bronson, D. L., Amdrews, P. W., Solter, D., Cervenka, J., Lange, P. H., and Fraley,E. E. Cell line derived from a metastasis of a human testicular germ cell tumor.Cancer Res.,40: 2500–2506, 1980.

    31. Robson, C. N., and Hickson, I. D. Isolation of cDNA clones encoding a humanapurinic/apyrimidinic endonuclease that corrects DNA repair and mutagenesis defectsin E. coli xth (exonuclease III) mutants. Nucleic Acids Res.,19: 5519–5523, 1991.

    32. Wilson, D. M., III, Bennett, R. A., Marquis, J. C., Ansari, P., and Demple, B.Trans-complementation by human apurinic endonuclease (Ape) of hypersensitivity toDNA damage and spontaneous mutator phenotype in apn1-yeast. Nucleic Acids Res.,23: 5027–5033, 1995.

    33. Herring, C. J., Deans, B., Elder, R. H., Rafferty, J. A., MacKinnon, J., Barzilay, G.,Hickson, I. D., Hendry, J. H., and Margison, G. P. Expression levels of the DNArepair enzyme HAP1 do not correlate with the radiosensitivities of human or HAP1-transfected rat cell lines. Br. J. Cancer,80: 940–945, 1999.

    34. Thomson, B., Tritt, R., Davis, M., Perlman, E. J., and Kelley, M. R. Apurinic/apyrimidinic endonuclease expression in pediatric yolk sac tumors. J. Pediatr. Oncol.,in press, 2000.

    35. Kakolyris, S., Kaklamanis, L., Engels, K., Turley, H., Hickson, I. D., Gatter, K. C.,and Harris, A. L. Human apurinic endonuclease 1 expression in a colorectal adenoma-carcinoma sequence. Cancer Res.,57: 1794–1797, 1997.

    36. Kakolyris, S., Kaklamanis, L., Engels, K., Fox, S. B., Taylor, M., Hickson, I. D.,Gatter, K. C., and Harris, A. L. Human AP endonuclease 1 (HAP1) protein expressionin breast cancer correlates with lymph node status and angiogenesis. Br. J. Cancer.,77: 1169–1173, 1998.

    37. Haring, M., Rudiger, H., Demple, B., Boiteux, S., and Epe, B. Recognition of oxidizedabasic sites by repair endonucleases. Nucleic Acids Res.,22: 2010–2015, 1994.

    38. Johnson, A. W., and Demple, B. Yeast DNA 39-repair diesterase is the major cellularapurinic/apyrimidinic endonuclease: substrate specificity and kinetics. J. Biol. Chem.,263: 18017–18022, 1988.

    39. Levin, J. D., and Demple, B.In vitro detection of endonuclease IV-specific DNAdamage formed by bleomycinin vivo. Nucleic Acids Res.,24: 885–889, 1996.

    40. Meira, L. B., Cheo, D. L., Hammer, R. E., Burns, D. K., Reis, A., and Friedberg,E. C. Re: “Genetic interaction between HAP1/REF-1 andp53.” Nat Genet.,17:145, 1997.

    2225

    APE1 PROTECTS NT2 CELLS FROM BLEOMYCIN/RADIATION

    on April 2, 2021. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/

  • 2001;61:2220-2225. Cancer Res Kent A. Robertson, Heather A. Bullock, Yi Xu, et al. and RadiationOverexpression in NT2 Cells Confers Resistance to Bleomycin Altered Expression of Ape1/ref-1 in Germ Cell Tumors and

    Updated version

    http://cancerres.aacrjournals.org/content/61/5/2220

    Access the most recent version of this article at:

    Cited articles

    http://cancerres.aacrjournals.org/content/61/5/2220.full#ref-list-1

    This article cites 37 articles, 14 of which you can access for free at:

    Citing articles

    http://cancerres.aacrjournals.org/content/61/5/2220.full#related-urls

    This article has been cited by 24 HighWire-hosted articles. Access the articles 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://cancerres.aacrjournals.org/content/61/5/2220To request permission to re-use all or part of this article, use this link

    on April 2, 2021. © 2001 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    http://cancerres.aacrjournals.org/content/61/5/2220http://cancerres.aacrjournals.org/content/61/5/2220.full#ref-list-1http://cancerres.aacrjournals.org/content/61/5/2220.full#related-urlshttp://cancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://cancerres.aacrjournals.org/content/61/5/2220http://cancerres.aacrjournals.org/