8
[CANCER RESEARCH58, 283-289, January 15. 19981 ABSTRACT This study was designed to assess the efficacy of a new antimelanoma therapeutic strategy that relies on the use of a c-myc antisense 15-mer phosphorothioate oligodeoxynucleotide ([SJODN), in combination with cisplatin (cis-dlamminedichloroplatinwn; DDP), which is currently used in the clinical management ofmelanoma patients. Proliferation and colony formation of melanoma cells were both inhibited by the DDP/c-myc anti sense [SIODN combination to a greater extent than that observed with either agent alone. Inhibition was most effective when DDP was followed by c-myc antisense [SJODNs. Cell cycle flow cytometric analysis of cells exposed to the two agents either alone or In combination demonstrated that (a) c-myc antisense [SIODNs Induced an accumulation of cells in S phase and apoptosis in a fraction of the cells, detectable at day 5 after the beginning of treatment (b) DDP induced a block in G2-M phase detect able at day 1, which was partially recovered, and apoptosis similar in extent to that induced by c-myc antisense [SJODNs; and (c) DDP and c-myc antisense [S]ODNs together Induced arrest in G2-M phase, which was maximum at day 3, Le., delayed as compared to the block induced by DDP. The combination induced a higher percentage of apoptosis, evident at day 3 from the Start of treatment, that correlated with a marked reduction in Bcl-2 expression. Mice bearing human melanoma xenografts and treated sequentially with DDP and c-myc antisense (SIODNs showed a higher inhibition of tumor growth, reduction In the number of lung metastases,and increasem life span comparedwith those treatedwith either agent alone. Together, these data lend support to the development of anticancer therapies involving oncogene-targeted antisense ODNs and conventional antineoplastic drugs INTRODUCTION Recent advances in delineating the genetic and molecular bases of cancer have raised interest in the possibility of correcting the genetic alterations responsible for the phenotype of neoplastic cells (1, 2). The identification of activated oncogenes and inactivated tumor suppres sor genes as the fundamental genetic difference between tumor and normal cells has made it possible to consider these genes as potential targets for antitumor therapy. An attractive approach to controlling gene expression is represented by the â€oeantisense― strategy, which, by restoring normal regulatory functions in cancer cells, could render them more susceptible to standard chemotherapy. Systemic administration of exogenous anti sense oligodeoxynucleotides complementary to oncogene-encoded mRNAs inhibits gene expression by forming RNA-DNA duplexes, thereby reducing the activity of the targeted gene products (3, 4). Antisense oligodeoxynucleotides have been reported to inhibit a va Received 7/2/97; accepted 11/10/97. 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 Ths work was partially supported by grants from Associazione Italiana per Ia Ricerca sul Cancro (to G. C.), Mimstero della Sanità (to G. Z.), Consiglio Nazionale delle Ricerche (to G. Z.), and Italy-Usa Project on Therapy of Tumors (to G. Z.). 2 To whom requests for reprints should be addressed, at Laboratory of Experimental Chemotherapy, Experimental Research Center, Regina Elena Cancer Institute, Via delle Messi d'Oro n. 156, 00158 Rome, Italy. Phone: 39-6-49852537; Fax: 39-6-49852537; E-mail: [email protected]. riety of oncogenes, including ras, bcr/abl, bcl-2, c-myb, and c-myc in tumors of different histotypes (5—7). We reported previously that c-myc antisense [S]ODNs3 are effec tive inhibitors of human melanoma cell growth in vitro and in vivo (6). Mice bearing human melanoma xenografts and treated with c-myc antisense [S]ODNs showed a marked inhibition of tumor growth, a reduction in the number of lung metastases, and an increased life span. This antitumor activity was associated with an early down-regulation of c-Myc expression, followed by inhibition of cell proliferation and induction of apoptosis. However, despite the significantly longer survival time, all treated animals died of their disease, indicating that inhibiting the expression of a single dysregulated oncogene is insuf ficient to completely suppress tumor cell proliferation. Therefore, the use of ODNs in combination with antineoplastic drugs might result in more complete control of cancer growth (8, 9). To determine whether an antisense strategy might enhance the efficacy of antitumor therapy, we investigated the effects of c-myc antisense [SIODNs in combination with cisplatin (DDP) on human melanoma cells constitutively expressing the c-myc oncogene. DDP was chosen based on clinical studies showing that, in combination with other drugs, it improves the therapeutic efficacy in metastatic melanoma. In addition, other authors have demonstrated that the combination of DDP and c-myc antisense [S]ODNs determines a synergistic inhibitory effect on bladder tumor cells in vitro (10). However, no studies have been reported on the efficacy of such therapy in experimental models in vivo. In the present study, we show that the combination of DDP and c-myc antisense [S]ODNs exerts significantly greater antiproliferative and antitumor effects than either agent used alone on human mela noma cells in vitro or xenografted to mice. Cell cycle perturbations and apoptosis might underlie the enhanced efficacy of this combina tion. MATERIALS AND METHODS Tumor Cell Lines and Tumor Implants. Human melanoma cell line Ml4 was maintained as a monolayer culture in RPM! supplemented with 10% heat-inactivated FCS, antibiotics, and L-glutamine (2 mM) at 37°C in a 5% C02-95% air atmosphere in a humidified incubator. Human primary melanoma NG was obtained and transplanted as described (6). CD-l male nude (nu/nu) mice, 6—8 weeks old, were injected in the hind leg muscles with a cell suspension of 2.5 x 106NO cells. A tumor mass of --70 mg was evident in all mice on day 4 after implant. All experiments were carried out between the fifth and eighth passages of the NO tumor in nude mice. Drug and [SJODNs. Clinical grade DDP (Pronto Platamine) was obtained from Pharmacia (Milan, Italy). DDP dilutions were freshly prepared before each experiment. A l5-mer antisense [S]ODN (5'-AACGTTGA0000CAT-3') complemen (sty to the translation initiation region of c-myc mRNA (Lynx Therapeutics Inc., Hayward, CA) was used. The scrambled sequence [S]ODN containing the 3 The abbreviations used are: [SIODN, phosphorothioate oligodeoxynucleotide; DDP, cis-diamminedichloroplatinum; EBSS, Earle's balanced salt solution; TWI, tumor weight inhibition. 283 c-myc Antisense Oligodeoxynucleotides Enhance the Efficacy of Cisplatin in Melanoma Chemotherapy in Vitro and in Nude Mice' Gennaro Citro, Igea D'Agnano, Carlo Leonetti, Roberto Perini, Barbara Bucci, Gerald Zon, Bruno Calabretta, and Gabriella Zupi2 Laboratory of Erperimental Chemotherapy, Regina Elena Cancer institute, Centro Ricerca Sperimentale. 00158 Rome, Italy fG. C., i. D.. C. L, R. P.. B. B., G. ii: Biomedical Technology institute, Consiglio Nazionale delle Ricerche, 00161 Rome, italy Ii. Di: Lynx Therapeutics, Hayward, California 94545 1G. ZJ; and Kimmel Cancer institute, Thomas Jefferson University, Philadelphia. Pennsylvania 19107-6799 (B. C.J on April 1, 2016. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

c-myc Antisense Oligodeoxynucleotides Enhance the Efficacy of Cisplatin in Melanoma Chemotherapy in Vitro and in Nude Mice

  • Upload
    cnr-it

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

[CANCER RESEARCH58, 283-289, January 15. 19981

ABSTRACT

This study was designed to assess the efficacy of a new antimelanomatherapeutic strategy that relies on the use of a c-myc antisense 15-merphosphorothioate oligodeoxynucleotide ([SJODN), in combination withcisplatin (cis-dlamminedichloroplatinwn; DDP), which is currently usedin the clinical management ofmelanoma patients. Proliferation and colonyformation of melanoma cells were both inhibited by the DDP/c-myc antisense [SIODN combination to a greater extent than that observed witheither agent alone. Inhibition was most effective when DDP was followedby c-myc antisense [SJODNs. Cell cycle flow cytometric analysis of cellsexposed to the two agents either alone or In combination demonstratedthat (a) c-myc antisense [SIODNs Induced an accumulation of cells in Sphase and apoptosis in a fraction of the cells, detectable at day 5 after thebeginning of treatment (b) DDP induced a block in G2-M phase detectable at day 1, which was partially recovered, and apoptosis similar inextent to that induced by c-myc antisense [SJODNs; and (c) DDP andc-myc antisense [S]ODNs together Induced arrest in G2-M phase, whichwas maximum at day 3, Le., delayed as compared to the block induced byDDP. The combination induced a higher percentage of apoptosis, evidentat day 3 from the Start of treatment, that correlated with a markedreduction in Bcl-2 expression. Mice bearing human melanoma xenograftsand treated sequentially with DDP and c-myc antisense (SIODNs showeda higher inhibition of tumor growth, reduction In the number of lungmetastases,and increasem life span comparedwith those treatedwitheither agent alone. Together, these data lend support to the developmentof anticancer therapies involving oncogene-targeted antisense ODNs andconventional antineoplastic drugs

INTRODUCTION

Recent advances in delineating the genetic and molecular bases ofcancer have raised interest in the possibility of correcting the geneticalterations responsible for the phenotype of neoplastic cells (1, 2). Theidentification of activated oncogenes and inactivated tumor suppressor genes as the fundamental genetic difference between tumor andnormal cells has made it possible to consider these genes as potentialtargets for antitumor therapy.

An attractive approach to controlling gene expression is representedby the “antisense―strategy, which, by restoring normal regulatoryfunctions in cancer cells, could render them more susceptible tostandard chemotherapy. Systemic administration of exogenous antisense oligodeoxynucleotides complementary to oncogene-encodedmRNAs inhibits gene expression by forming RNA-DNA duplexes,

thereby reducing the activity of the targeted gene products (3, 4).Antisense oligodeoxynucleotides have been reported to inhibit a va

Received 7/2/97; accepted 11/10/97.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 with18 U.S.C. Section 1734 solely to indicate this fact.

1 Ths work was partially supported by grants from Associazione Italiana per Ia Ricerca

sul Cancro (to G. C.), Mimstero della Sanità(to G. Z.), Consiglio Nazionale delleRicerche (to G. Z.), and Italy-Usa Project on Therapy of Tumors (to G. Z.).

2 To whom requests for reprints should be addressed, at Laboratory of Experimental

Chemotherapy, Experimental Research Center, Regina Elena Cancer Institute, Via delleMessi d'Oro n. 156, 00158 Rome, Italy. Phone: 39-6-49852537; Fax: 39-6-49852537;E-mail: [email protected].

riety of oncogenes, including ras, bcr/abl, bcl-2, c-myb, and c-myc intumors of different histotypes (5—7).

We reported previously that c-myc antisense [S]ODNs3 are effective inhibitors of human melanoma cell growth in vitro and in vivo (6).Mice bearing human melanoma xenografts and treated with c-mycantisense [S]ODNs showed a marked inhibition of tumor growth, areduction in the number of lung metastases, and an increased life span.

This antitumor activity was associated with an early down-regulationof c-Myc expression, followed by inhibition of cell proliferation andinduction of apoptosis. However, despite the significantly longersurvival time, all treated animals died of their disease, indicating thatinhibiting the expression of a single dysregulated oncogene is insufficient to completely suppress tumor cell proliferation. Therefore, theuse of ODNs in combination with antineoplastic drugs might result inmore complete control of cancer growth (8, 9).

To determine whether an antisense strategy might enhance theefficacy of antitumor therapy, we investigated the effects of c-mycantisense [SIODNs in combination with cisplatin (DDP) on humanmelanoma cells constitutively expressing the c-myc oncogene. DDPwas chosen based on clinical studies showing that, in combinationwith other drugs, it improves the therapeutic efficacy in metastaticmelanoma. In addition, other authors have demonstrated that thecombination of DDP and c-myc antisense [S]ODNs determines a

synergistic inhibitory effect on bladder tumor cells in vitro (10).However, no studies have been reported on the efficacy of suchtherapy in experimental models in vivo.

In the present study, we show that the combination of DDP andc-myc antisense [S]ODNs exerts significantly greater antiproliferativeand antitumor effects than either agent used alone on human melanoma cells in vitro or xenografted to mice. Cell cycle perturbationsand apoptosis might underlie the enhanced efficacy of this combination.

MATERIALS AND METHODS

Tumor Cell Lines and Tumor Implants. Human melanoma cell line Ml4

was maintained as a monolayer culture in RPM! supplemented with 10%heat-inactivated FCS, antibiotics, and L-glutamine (2 mM) at 37°C in a 5%

C02-95% air atmosphere in a humidified incubator. Human primary melanoma

NG was obtained and transplanted as described (6). CD-l male nude (nu/nu)mice, 6—8 weeks old, were injected in the hind leg muscles with a cell

suspension of 2.5 x 106NO cells. A tumor mass of --70 mg was evident inall mice on day 4 after implant. All experiments were carried out between thefifth and eighth passages of the NO tumor in nude mice.

Drug and [SJODNs. Clinical grade DDP (Pronto Platamine) was obtainedfrom Pharmacia (Milan, Italy). DDP dilutions were freshly prepared beforeeach experiment.

A l5-mer antisense [S]ODN (5'-AACGTTGA0000CAT-3') complemen(sty to the translation initiation region of c-myc mRNA (Lynx Therapeutics

Inc., Hayward, CA) was used. The scrambled sequence [S]ODN containing the

3 The abbreviations used are: [SIODN, phosphorothioate oligodeoxynucleotide; DDP,

cis-diamminedichloroplatinum; EBSS, Earle's balanced salt solution; TWI, tumor weightinhibition.

283

c-myc Antisense Oligodeoxynucleotides Enhance the Efficacy of Cisplatin inMelanoma Chemotherapy in Vitro and in Nude Mice'

Gennaro Citro, Igea D'Agnano, Carlo Leonetti, Roberto Perini, Barbara Bucci, Gerald Zon, Bruno Calabretta, andGabriella Zupi2Laboratory of Erperimental Chemotherapy, Regina Elena Cancer institute, Centro Ricerca Sperimentale. 00158 Rome, Italy fG. C., i. D.. C. L, R. P.. B. B., G. ii: BiomedicalTechnology institute, Consiglio Nazionale delle Ricerche, 00161 Rome, italy Ii. Di: Lynx Therapeutics, Hayward, California 94545 1G. ZJ; and Kimmel Cancer institute,Thomas Jefferson University, Philadelphia. Pennsylvania 19107-6799 (B. C.J

on April 1, 2016. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Table 1Therapeutic efficacy ofc-myc (S1ODNs andDDP in NG melanoma-bearingmice1'WI

at firstTWI at secondTVs'I at thirdMetastasisIncrease inlifeSchedule'cycle.%cycle, %cycle, %T —C―Toxic deathsreduction, %Cspan,%ADDP(day4)61605210Oofl6835B

DDP (day 7)5251467.50 of10829CAntisense45505070 of I82528DScrambled43001 of1008EDDP (AntisenseY'879293360 of185275FDDP(Scrambled)63624581 of 10837

C-MYC ANTISENSEODNS AND DDP IN MELANOMA TREATMENT

“0-quartet―motif (5'-AAGCATACGGGGTGT-3') was used as control.[SJODNs were resuspended at 5 mg/mI in sterile EBSS.

In Vitro ISIODNs and DDP Treatment. M14 cells were seeded in 60-mmPets-idishes at a density of 3 X l0@cells per dish. After 24 h, [SJODNs wereadded at concentrations of 100 @g/mlon day 1 and 50 @ig/mlon days 2—4.Cells were exposed to DDP at a dose of 1 @g/mlon day 1 or day S of growth.Equimolar amounts of c-myc scrambled [S]ODNs were used in control experiments. In combination experiments, cells were pre- or posttreated with DDP

for 2 h and exposed to c-myc antisense [S]ODNs for 4 days. For eachcombination, parallel analyses with each agent alone were performed andprocessed by changing the medium at the same time. Antiproliferative effectswere evaluated daily based on cell number and viability as assessed in aCoulter counter (model ZM; Kontron Instruments, Luton, United Kingdom)and by trypan blue dye exclusion, respectively.

To evaluate cell colony-forming ability, aliquots of cell suspension fromeach sample were seeded into 60-mm Petri dishes with RPM! 1640 andincubated at 37°Cin a 5% C02-95% air atmosphere for 10—12days. Colonieswere stained with 2% methylene blue in 95% ethanol and counted (1 colony 50 cells). Surviving fractions were calculated as the ratio of absolutesurvival of the treated sample:absolute survival of the control sample. Allexperiments were repeated four times in triplicate.

Western Blotting. Protein isolation and Western blot analyses were asdescribed (6). Briefly, proteins (30 @g)were separated by 10% SDS-PAGE,transferred to nitrocellulose filters, and reacted with monoclonal antibodiesspecific for human p62 c-Myc (clone 9ElO; Santa Cruz Biotechnology, SantaCruz, CA), p53 (clone Ab2; Oncogene Science, Manhasset, NY), and bcl-2(clone 124; DAKO A/S. Glostrup, Denmark). After stripping, filters wereincubated with antihuman @3-actinantibody (clone JLA 20; Oncogene Science),and reactivity was detected by enhanced chemiluminescence (Amersham International, Little Chalfont, Buckinghamshire, United Kingdom) according tothe manufacturer's instructions. c-Myc, p53, and bcl-2 levels were quantifiedby scanning densitometry (Bio-Rad 0700) of the autoradiography films andnormalized to @3-actinlevels.

Propidlum Iodide Staining and Flow Cytometry. Cell cycle analysis ofM14 cells exposed to DDP and [SIODNs, either separately or in combination,was performed by flow cytometry as described (8) during treatment and aftertreatment (0.5, 1, 2, 3, 4, 5, and 6 days after treatment). Apoptosis wasevaluated as described (6) at days 3, 5, and 6, and an apoptotic index was

calculated by dividing the percentage of apoptotic cells in treated samples by

that in the control cells as determined by flow cytometry. Morphology ofuntreated and treated cells was analyzed in May-Grunwald/Giemsa-stainedcytospins.

In Vivo Treatments. CD-I male nude mice (6—8weeks old and 22—24gin body weight) were obtained from Charles River Laboratories (Calco, Italy).Animals were kept under pathogen-free conditions and given acidified, autoclaved water and y-irradiated commercial diet ad libitum. All manipulationswere performed under sterile conditions in a laminar air-flow hood. Allprocedures involving mice and their care were in accord with institutional

guidelines in compliance with national and international laws and policies(European Economic Community Council Directive 86/109, OJL 358, Dec. 1,1987, and with the NIH Guide for the Care and Use of Laboratory Animals(11). Each experimental group included at least 10 mice, with a maximum of18 animals.

[S]ODNs were injected i.v. in alternating doses of 1 and 0.5 mg/day for 8consecutive days. This schedule was chosen based on previous studies (6).DDP was administered i.p. at 10 mg/kg (10% of the lethal dose; LDJ0)in threeconsecutive daily injections (3.3 mg/kg). Preliminary experiments demon

strated that this scheduling was less toxic than the single injection in terms ofbody weight loss and drug deaths. Three cycles at 7-day intervals wereadministered according to the following schedules. In schedule A, DDP (3.3mg/kg/day) was administered at days 4—6,22—24,and 40—42,and in scheduleB, DDP (3.3 mg/kg/day) at days 7—9,25—27,and 43—45was administered. Inschedules C (c-mycantisense) and D (scrambled [S]ODNs), the cycles were asfollows: 1 mg/mouse/day at days 7, 9, 11, and 13; 0.5 mg/mouse/day at days8, 10, 12, and 14; 1 mg/mouse/day at days 25, 27, 29, and 31 ; 0.5 mg/mouse/day at days 26, 28, 30, and 32; 1 mg/mouse/day at days 43, 45, 47, and 49; and0.5 mg/mouse/day at days 44, 46, 48, and 50. In the combination experiments,schedules A plus C (schedule E) and schedules A plus D (schedule F) wereadministered (see Table I). As a control, tumor-bearing mice treated withEBSS diluent were used.

The tumor weight was calculated from caliper measurements according tothe method of Geran et a!. (12). To evaluate the metastatic potential of NOmelanoma, tumor-bearing mice were killed 28 days after tumor cell implantation, their lungs were removed and fixed in Bouin's solution to distinguish

tumor nodules from lung tissue, and the number of metastases was determinedunder a dissecting microscope. Toxicity was assessed on the basis of apparentdreg-related deaths and net body weight loss. Death in a treated mouse waspresumed to be treatment related if the mouse died within 7 days after the lasttreatment. Net body weight loss was calculated as a percentage of the mean netbody weight of untreated mice.

Therapeutic efficacy of treatments was evaluated using the following endpoints: (a) percentage of TWI calculated as 1 —(mean tumor weight of treated

mice/mean tumor weight of controls) X 100; (b) tumor growth delay, evaluated as T —C, where T and C are the median times for treated and controltumors, respectively, to reach the same size (i.e., 700 mg); (c) percentage ofreduction in the number of lung metastases; and (d) percentage of increase inlife span calculated as [1 —survival time (in days) of treated mice/mediansurvival time (in days) of control mice] X 100.

Statistical Analysis. Results were analyzed by the Mann-Whitney nonparametric test. Differences were considered significant at P < 0.05 (twosided). Linear regression analysis was performed on in vivo tumor growth

curves, and the slopes were compared to evaluate the statistical significance of

the differences.

RESULTS

Effects of c-myc Antisense [SIODNs and DDP on M14 Cells.Fig. 1 shows the proliferation of Ml4 melanoma cells treated withc-myc antisense [SJODNs and DDP used alone or in combination.Only the scrambled [S]ODN sequence containing the G-quartet motifwas used as a control, because sense, scrambled, and scrambled0-quartet sequences were all completely ineffective on Ml4 and NGmelanoma cells both in vitro and in vivo (6). The scrambled [S]ODNcontrol sequence did not exert any antiproliferative effect either aloneor in combination with DDP. Proliferation ofcells exposed to DDP for

a@ cycles at 7-day intervals were administered according to the following schedules: A, DDP 3.3 mg/kg/day at days 4—6, 22—24, and 40—42; B, DDP 3.3 mg/kg/day at days

7—9,25—27,and 43—45;C, c-myc antisense; and D, scrambled [SJODNs 1 mg/mouse/day at days 7, 9, 11, 13 and 0.5 mg/mouse/day at days 8, 10, 12, and 14; 1 mg/mouse/day at days25, 27, 29, and 3 1 and 0.5 mg/mouse/day at days 26, 28, 30, and 32; and 1 mg/mouse/day at days 43, 45, 47, 49, and 0.5 mg/mouse/day at days 44, 46, 48, and 50. In the combinationexperiments, schedule A + C (schedule E) and schedule A + D (schedule F) were administered. Tumor-bearing mice treated with EBSS diluent were used as a control.

b Tumor growth delay (median times in days for treated and control tumors to reach the size of 700 mg)C The median number of metastases in the untreated group was 29 (15—35).

d All end point values were significantly different (P < 0.0001) among schedule (E) and the other schedules.

284

on April 1, 2016. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

C-MYC ANTISENSE ODNS AND DDP IN MELANOMA TREATMENT

100

75

50

25

0

100-

75

50

25

0-

Fig. 1. Effect of c-myc antisense [S]ODNs and DDP alone or incombination on M14 melanoma cell proliferation. Cells were treatedbeginning 1 day after cell seeding with the following schedules: A,scrambled (X) or c-myc antisense (0) [S]ODNs for 4 days (100 @gfmlon the 1st day and 50 @g/mlon days 2—4),and DDP (•) for 2 h (I@Lg/ml)and DDP for 2 h followed by scrambled (A) or c-myc antisense(0) [SJODNsfor4 daysat thesameconcentrations.Statisticalanalysis:D—D curve versusO—@Ocurve, P = 0.004; D—D curveversus •—•curve, P 0.04; B, symbols and concentrations of thetwo agents are the same as in A. DDP was administered for 2 h at daySafter cell seeding. In the combination, DDP followed [SJODN exposure.Statistical analysis: D—D curve versus O—O curve, P = 0.026;curve D—D curve versus •—• curve, P = 0.0022. Data in A andB are representative of four experiments with similar results; values aremeans (bars, SE) of triplicate samples within the same experiment.Whennot shown,the SE is smallerthanthe symbols.

0

0C)

‘4-0

2 4 6 8 10 5 7 9

Growth days

50%. The survival curve in cells exposed to DDP followed by c-mycantisense [S]ODNs was biphasic: after an exponential decrease in cellsurvival similar to that obtained from administering DDP alone, aplateau phase was observed, indicating that the cells are unable torecover from the DDP-induced damage.

Cell Cycle Perturbations and Apoptosis in M14 Cells. To elucidate the mechanisms by which the DDP plus c-myc antisense[S]ODN combination determines a significant inhibition of cell proliferation and survival, we analyzed cell cycle distribution of treated

and untreated M14 cells. Histograms of DNA content (Fig. 4) revealed that antisense treatment induced an accumulation of cells in Sphase, evident at day 3 after the start of treatment and concomitant

with a decrease in the percentage of 0@ cells. This result is consistentwith a prolonged staying of the antisense-treated cells in S phase asevaluated by bromodeoxyuridine incorporation (not shown). DDPtreatment induced G2-M-phase accumulation detectable at 12 h after

treatment and partially recovered within the following 3 days. However, DDP followed by c-myc antisense [S]ODNs produced significantly greater G2-M-phase accumulation still markedly evident at day3 from the beginning of treatment than that induced by DDP alone. Atday 6, cells exposed to either DDP or c-myc antisense [S}ODNsshowed a sub-01 peak, typical of apoptotic cells, with a concomitantloss of cells from the G2-M and S phases, respectively. The cellfraction in the sub-G1 peak increases after exposure to the DDP plus

c-Myc -@- —@— —

f3-act @:=@ _@ __::@

Fig. 2. p62 c-Myc protein levels in M14 cells treated with scrambled [SIODN5 (S),c-myc antisense [SIODN5 (AS), DDP (DDP), and DDP followed by c-myc antisense[SJODNs(DDP/AS;seeFig. 1legend);C,untreatedcells.Analysiswasperformedatday2 from the beginning of treatments.

100

0

@0l0

00

2 4 6 8 10

Days

Fig. 3. Survival curves of Ml4 cells exposed to c-myc antisense [SJODNs (O—O),DDP ( •—•), and DDP followed by c-myc antisense [SJODNs (D—D). Cells weretreated as described in the Fig. 1 legend. and, at the indicated times during the growth,aliquots ofcell suspension from each sample were seeded in Petri dishes and incubated for10—12days. Data are representative of four experiments with similar results. Values aremeans (bars, SE) of triplicate samples. When not shown, the SE is smaller than thesymbol. Statistical analysis: (D—D) curve versus (O—O) curve. P = 0.0006;(D—D) curveversus(•—•)curve,P 0.01.

285

C S AS DDP DDP/AS

2 h followed by c-myc antisense [S]ODN treatment was inhibited to agreater extent than that observed after treatment with either agentalone (Fig. 1A). This effect was already evident 2 days after the startof treatment (day 3 of growth), and cell number decreased furtherbetween days 6-10 of culture (more than 90% growth inhibition withrespect to control cells). DDP induced a maximal inhibition of cellproliferation of about 65%, and the antisense [S]ODNs determined areduction in cell number evident from day 4 of growth, with amaximum inhibition of 55% as compared to control cells. In cellstreated first with c-myc antisense [S]ODNs for 4 days followed byDDP for 2 h, cell number was also markedly reduced (75%), especially compared to the effect of DDP, which reduced cell number byabout 40% (Fig. 1B). As expected, DDP was less effective whenadministered in the plateau growth phase than during exponential

growth.All subsequent experiments were performed using the DDP fol

lowed by the c-myc antisense [S]ODN protocol, which is the mosteffective.

Immunoblot analysis of c-Myc protein levels in lysates from M14cells exposed to the different treatments to confirm the sequencespecific effect of [S]ODNs (Fig. 2) demonstrated that c-myc antisense[S]ODN treatment reduced c-Myc protein levels by about 40% in cellstreated with antisense [S]ODNs alone or in combination with DDP, ascompared to control cells. By contrast, c-Myc protein levels detectedin lysates from both scrambled [S]ODN- and DDP-treated cells weresuperimposable to levels of control cells.

Comparison of survival curves of M14 cells exposed to DDP

followed by c-myc antisense [S]ODNs and to each compound usedalone (Fig. 3) revealed a constant inhibition of colony formationelicited by antisense [S]ODNs alone starting from day 4 of culture,with a surviving fraction of about 50%, whereas DDP administeredalone induced a decrease in cell survival of about 85% observed at day4 of growth, with a partial recovery within the following days to about

on April 1, 2016. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

C-MYC ANTISENSE ODNS AND DDP IN MELANOMA TREATMENT

a b c

C

S

AS

DDP

DDP/AS

Fig. 4. Effect of DDP and [S]ODN5alone or in combination on the Ml4 cellcycle. Cell cycle analysis after propidium iodide staining was performed byflow cytometry at days 0.5 (a), 3 (b), and 6 (c) after the start of treatments.Untreated cells (C) and cells treated with scrambled [SJODNs (5), c-mycantisense [SJODNs (AS), DDP (DDP), and DDP followed by c-myc antisenseISIODN5 (DDP/AS) were analyzed.

DNA content

c-myc [SJODN combination, concomitant with a depletion of cellsfrom both S and G2-M phase. The scrambled [SJODN sequence didnot affect cell cycle phase distribution. Daily analysis to evaluateprogression through the cell cycle during and immediately after thetreatment (Fig. 5) revealed no detectable changes in the percentage of

cells in G2-M phase after exposure to c-myc antisense [S]ODNs alonecompared to untreated cells. Consistent with the data in Fig. 4, theDDP plus c-myc antisense [S]ODN combination induced an arrest inG2-M phase, reaching a maximum value with a delay of 2 dayscompared to the block induced by DDP. Apparently, the block induced by DDP alone is completely recovered at day 5, whereas theDDP plus c-myc antisense [S]ODN combination prevents the cellsfrom progressing through the cell cycle. These results suggest that theDDP reversible G2-M phase arrest becomes irreversible on DDP plusc-myc antisense [S]ODN treatment.

On the basis of DNA histograms at day 6 from the beginning oftreatment (Fig. 4) and based on previous data indicating the inductionof apoptosis by c-myc antisense [S]ODNs and by DDP (6, 13, 14), weanalyzed apoptosis in M14 cells (Fig. 6A).

Apoptosis was more extensive in M14 cells exposed to the DDPplus c-myc antisense [S]ODNs than after treatment with either agentalone. DDP plus c-myc antisense [S]ODN-induced apoptosis wasevident at day 3 after treatment with an apoptotic index ranging from13 to 20 up to day 6 from the beginning of treatment, whereas DDPand c-myc antisense [S]ODNs alone induced a lower level of apoptosis even at day 6 (maximum apoptotic index, 10; Fig. 6A). Apoptosiswas confirmed by morphological analysis of untreated and treatedcells (data not shown). These data are in agreement with the Westernblot analysis of p53 and Bcl-2 proteins associated with the apoptoticprogram (Fig. 6B). Densitometric analysis of the blots indicated that

286

121)

91)

61)

31)

0

(5

L)0

0

1) 0,5 I 2 3 4 5

Time after treatment (days)

Fig. S. Effect of DDP (0), DDP followed by c-myc antisense [SJODN combination(s), andc-mycantisenseESIODN5(@)onG2-MphasedistributioninMl4cells.Thepercentage of change in the G2-M phase fraction was calculated by dividing the percentage of treated cells in G2-M phase by that of untreated cells at the indicated time after thestart of treatments.

C,'(5C)

C.0

2:

e a@@ió.1 @éia@s

on April 1, 2016. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

C S AS DDPDDP/AS

p53@@

bcl2

p53@@@@

bct2@@

C-MYC ANTISENSE ODNS AND DDP IN MELANOMA TREATMENT

A

Fig. 6. A, apoptotic index in M14 melanoma cellstreated with c-myc antisense [S]ODNs (1), DDP(0), and DDP plus c-myc antisense [SJODNs (0).Apoptoticindiceswerecalculatedby dividingtheapoptotic cell percentage of treated cells by that ofuntreated cells as determined by flow cytometry.Data are means; bars, SE. B, Western blot analysisof p53 and Bc1-2 proteins in control cells (C) andcells treated with scrambled [S]ODNs (S), c-mycantisense [S]ODNs (AS), DDP (DDP), and DDPplus c-myc antisense [SIODN5 (DDP/AS) as evaluated at days 2 and 3 after treatment.

B

L@ day2

HF@dII@

30

:@

a.<

03

Time after treatment (days)6

c-myc antisense [S]ODNs, DDP, and the combination of the agents allinduced an increase in cellular levels of p53 (—2-fold) evaluated ateither day 2 or day 3. Bcl-2 levels decreased slightly at day 2 in alltreated cells; at day 3, DDP and the c-myc antisense [S]ODNs eachdetermined a 50% reduction in Bcl-2 expression, whereas the combination reduced Bcl-2 expression by —75%.

In Vivo Antitumor Effect of DDP and c-myc Antisense[SJODNs.To evaluatethe efficacy of DDP and c-myc antisense[S]ODNs in vivo, mice bearing NO melanoma were treated with thetwo agents, alone or in combination, at day 4 after cell injection, and

tumor weight was monitored (Fig. 7). DDP and c-myc antisense[S]ODNs administered separately caused a significant decrease intumor growth of —50% after the end of the third treatment cycle,whereas the scrambled sequence did not affect tumor growth. DDPplus c-myc antisense [S]ODNs in combination, however, inhibitedtumor growth by —90%even after the first cycle of treatment, withcomplete tumor regression evident from day 10 up to day 18 ofgrowth. The antitumoral effect elicited by the combination of DDPplus c-myc antisense [S]ODN treatment was significantly more potent(P < 0.0001) than that obtained using the two agents separately.

Table 1 summarizes the end points evaluated after the varioustreatments. DDP was administered at day 4 (schedule A) and at day 7(schedule B) to compare the efficacy of DDP versus the c-myc antisense[S]ODNs administered at day 7 and versus the combination started at day4. DDP given alone (schedule A) induced a significant inhibition oftumor weight (P < 0.0001 versus untreated mice) after the first (61%),second (60%), and third (52%) cycles of administration. A marked delay

in tumor growth (10 days) was also observed (P < 0.0001 versusuntreated mice) accompanied by a significant increase in survival(P < 0.0001 versus untreated mice). No significant reduction in thenumber of lung metastases was observed. As expected, DDP was lesseffective when administered at day 7, in a relatively late stage of tumorgrowth. Repeated cycles of c-myc antisense [S]ODNs (schedule C)started at day 7 also produced a significant inhibition oftumor weight anda growth delay of 7 days as compared to untreated mice (P < 0.0001).Moreover, c-myc antisense [SJODN treatment caused a marked reductionin the number oflung metastases compared to DDP-treated and untreatedmice, whereas treatment with c-myc scrambled [S]ODNs showed noantitumor activity. The combination of DDP and c-myc antisense

[S]ODNs was significantly more effective in delaying (36 days) and ininhibiting (P < 0.0001) tumor growth (ranging from 87% after the firstcycle to 93% at the end of treatment) than DDP or c-myc antisense[S}ODNs alone or DDP in combination with scrambled [S]ODNs. Also,the number oflung metastases was reduced significantly. Finally, mediansurvival, which represents the most important end point in the evaluationof the efficacy of a new therapeutic regimen, was increased markedly(75%).

DISCUSSION

Melanoma is a highly malignant cancer, and despite some efficacyof many chemotherapeutic agents used alone or in combination (15,16), no agent has made a significant impact on the survival of patientswith metastatic melanoma.

Fig.7. Effectof DDPand [S]ODNsaloneor in combinationon thegrowth of the NO melanoma tumor implanted in nude mice. Treatmentswith three cycles of administration of antisense [SJODNs, scrambled[S)ODNs, and DDP alone or in combination were performed as described in “Materialsand Methods.―s, @j@fl@j mice; A, mice treatedwith scrambled [S]ODNs; 0, mice treated with c-myc antisense[SJODNs;0, mice treatedwith DDPat day 4; U, mice treatedwithDDP/scrambled [S]ODNs; •,mice treated with DDP/c-myc antisense[SJODNs.Arrowsindicatethestartofeachcycleof treatment.Pointsaremeans (bars, SD). Slope values of the different tumor growthcurves were evaluated as reported in “Materialsand Methods,―as follows: DDP/antisense (•),slope value 11 (P < 0.0001 versus all of thecurves); DDP (0), slope value 77 (P < 0.0001 versus * and tx);Antisense (0), slope value 92 (P < 0.0001 versus * and @);DDP/scrambled (W@slope value 113 (P < 0.0001 versus@ and z@);scrambled(ti), slope value 204; and untreated (a), slope value 177.

10.000

00E i.000

000)

I-

E100

4 10 20 30 40 50 60

Daysafterinjection

t t

287

on April 1, 2016. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

C-MYC ANTISENSE ODNS AND DDP IN MELANOMA TREATMENT

In this study, we demonstrate that the use of antisense [S]ODNs incombination with a conventional anticancer drug such as DDP represents an attractive cancer treatment modality. The combination ofDDP with antisense c-myc antisense [S]ODNs, independent of theorder of administration, produced a greater inhibitory effect on cellproliferation of melanoma cells than did either agent given alone.However, administration of DDP followed by c-myc antisense[S]ODNs appeared to be more effective, because it produced a 90%cell growth inhibition compared to the 70% induced by the agentsused in the reverse order. In turn, the inhibition of cell proliferationwas accompanied by a decrease in colony-forming capacity, with only10% of the population surviving the combined treatment. Analysis ofthe survival curves clearly indicated that the tumor cells partiallyrecovered from the cytotoxic effect induced by DDP treatment; infact, cell survival increased from 25 to 50%, whereas the addition ofc-myc antisense [S]ODNs blocked the ability of the cells to recoverfrom the DDP damage. Our findings are in accord with those of aprevious study, in which c-myc antisense ODNs were shown toenhance the cytotoxic activity of DDP when used in combination inbladder tumor cells in vitro (10). In addition, combined treatmentsusing other [S]ODNs and cytotoxic drugs proved more effective ininhibiting leukemia cell growth in severe combined immunodeficientmice than the agents used individually (17).

The enhanced cytotoxic effect elicited by the combined treatmentmight rest in the different mechanisms of action of the two agents. Infact, cell cycle analysis after DDP treatment revealed the characteristic G2-M phase block after 1 day, although this block was in part

circumvented, as evidenced by the cell cycle repopulation during thedays following the treatment. Instead, the targeting of c-myc mRNAby the antisense [S]ODNs in melanoma cells constitutively expressingc-myc, although not arresting the cells in the G@)/G)phases (18, 19),did prevent cell progression through the S phase, suggesting that atleast in melanoma cells c-myc plays a role in the S to G2-M transition.Additional studies are needed to assess the precise function of c-mycin melanoma cell cycle phases. On the other hand, the S-phase arrestobserved after exposure of the cells to the c-myc antisense [S]ODNsis in agreement with the reported ability of ectopic c-myc to rescue

S-phase arrest of interleukin 3-dependent hematopoietic cells stimulated by epidermal growth factor (20). A similar S-phase arrest induced by the c-myc antisense sequence, identical to that used herein,has been reported in a leukemia model (5). When the melanoma cellswere treated with DDP followed by antisense [S]ODNs, accumulationin G2-M phase was greater than after treatment with each agent used

separately. In addition, maximum G2-M accumulation was delayed by2 days, probably due to the antisense-induced block in S phase in cellsinitially damaged by DDP. This block appears to be irreversible,because even at day 3 from the start of treatment, a fraction of thecells, which is unable to repopulate the cell cycle, died from apoptosis.The proportion of apoptotic cells increased dramatically during thefollowing days (—75%), with a marked loss of cells with S and G2-MDNA content. However, DDP or c-myc antisense [S]ODNs alone canalso induce apoptosis. In fact, a fraction of the cell population arrestedin G2-M phase by DDP was apoptotic at day 6 from the start oftreatment, concomitant with a loss of cells with G2-M DNA content.This is consistent with the reported apoptosis induced by DDP treatment in leukemia cells, as well as in solid tumors (14, 21). Furthermore, we showed previously that c-myc antisense [S]ODN treatmentinduces apoptosis in M 14 cells (6), in association with a significantdepletion of the S-phase compartment. Apoptosis after c-myc antisense treatment has also been described for BV173 leukemia cells (5).The increased proportion of apoptotic cells observed after treatmentwith DDP followed by c-myc antisense [S]ODNs is consistent with

the marked decrease in levels of the antiapoptotic Bcl-2 protein afterthe combined treatment.

The effects of DDP/[S]ODN treatment were examined in nude micebearing human melanoma (NG). This tumor spontaneously gives riseto reproducible lung metastases, providing a good model for evaluation of new pharmaceutical drugs. Using the same experimentalmodel, we demonstrated previously that two repeated cycles of c-mycantisense [S]ODNs effectively inhibits melanoma growth. In the present study, we also demonstrate that c-myc antisense [S]ODNs caninhibit tumor growth at a relatively late stage when the tumor mass is— 180 mg. These in vivo effects were sequence specific, given that the

scrambled [S]ODNs did not affect tumor growth. Our in vivo resultssuggest that the therapeutic index of c-myc antisense [S]ODNs issomewhat higher than that of repeated cycles of DDP, because,although tumor weight inhibition and lack of toxicity were similar inboth treatments, c-myc antisense [S}ODNs also induced a significantdecrease in the number of lung metastases. Antitumoral efficacy wasenhanced when DDP was followed by the c-myc antisense [S]ODNsfor three repeated cycles at 7-day intervals. In fact, tumor weight wassignificantly inhibited, and growth was delayed by 36 days. This lastresult is particularly impressive, considering that the life span of micebearing melanoma is —60days. The significant increase in animal lifespan (75%), which exceeds that reported to date for other combinationregimens with antineoplastic drugs or immunomodulatory therapy(22), suggests this combination as a promising therapeutic approach inmelanoma treatment.

ACKNOWLEDGMENTS

We thank Antonio Candiloro for his technical help. We thank Maria LetiziaFolino and Simona Righi for their helpful assistance in preparing the manu

script.

REFERENCES

1. Gottesman, M. M. Report of a meeting: molecular basis of cancer therapy. J. NatI.Cancer Inst., 86: 1277—1285,1994

2. Mastrangelo, M. J., Berd, D., Nathan, F. E., and Lattime, E. C. Gene therapy forhuman cancer: an assay for clinicians. Semin. Oncol., 23: 4—21,1996.

3. Wagner, R. W. Gene inhibition using antisense oligodeoxynucleotides. Nature(Lond.), 372: 333—335,1994.

4. Stein, C. A., and Cheng, Y. C. Antisense oligonucleotides as therapeutic agents: is thebullet really magical? Science (Washington DC), 261: 1004—1012,1993.

5. Skorski, T., Nieborowska-Skorska, M., Campbell, K., lozzo, R. V., Zon, G.,Darzynkiewicz, Z., and Calabrena, B. Leukemia treatment in severe combinedimmunodeficiency mice by antisense oligodeoxynucleotides targeting cooperatingoncogenes. J. Exp. Med., 182: 1645—1653,1995.

6. Leonetti, C., D'Agnano, I., Lozupone, F., Valentini, A., Geiser, T., Zon, G.,Calabretta, B., Citro, G., and Zupi, G. Antitumor effect of c-myc antisense phosphorothioate oligodeoxynucleotides on human melanoma cells in vitro and in mice.J. NatI. Cancer Inst., 88: 419—429, 1996.

7. Citro, G., Perrotti, D., Cucco, C., D'Agnano, I., Sacchi, A., Zupi, G., andCalabretta, B. Inhibition of leukemia cell proliferation by receptor-mediateduptake of c-myb antisense oligodeoxynucleotides. Proc. Natl. Acad. Sci. USA, 89:7031—7035, 1992.

8. Del Bufalo, D., Cucco, C., Leonetti, C., Citro, G., D'Agnano, I., Benassi, M., Geiser,T., Zon, G., Calabretta, B., and Zupi, G. Effect of cisplatin and c-myb antisensephosphorothioate oligodeoxynucleotides combination on a human colon carcinomacell line in vitro and in vivo. Br. 3. Cancer, 74: 387—393,1996.

9. Nieborowska-Skorska, M., Nakashima, M., Ratajczak, M., Steplewski, Z., Calabretta,B., and Skorski, T. Oncogene-targeted antisense oligodeoxynucleotides combinedwith chemotherapy or immunotherapy: a new approach for tumor treatment? FoliaHistochem. Cytochem., 32: 35—40,1994.

10. Mizutani, Y., Fukumoto, M., Bonavida, B., and Yoshida, 0. Enhancement of sensitivity of urinary bladder tumor cells to cisplatin by c-myc antisense oligonucleotide.Cancer (Phila.), 74: 2546—2554,1994.

11. Guide for the Care and Use of Laboratory Animals. NIH Publ. No. 85-23. Bethesda,MD: NIH, 1985.

12. Geran, R. I., Greenberg, N. H., Macdonald, M. M., Shumacher, A. M., and Abbott,B. J. Protocols for screening chemical agents and natural products against animaltumors and other biological systems, Ed. 3. Cancer Chemother. Rep., 3: 1—88,1972.

288

on April 1, 2016. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

C-MYC ANTISENSE ODNS AND DDP IN MELANOMA TREATMENT

13. Alarcon, R. M., Rupnow, B. A., Graeber, T. G., Knox, S. J., and Giaccia, A. J.Modulation of c-Myc activity and apoptosis in vivo. Cancer Res., 56: 43 15—4319,1996.

14. Onnerod, M. G., Orr, R. M., and Peacock, J. H. The role of apoptosis in cell killingby cisplatin: a flow cytometric study. Br. J. Cancer, 69: 93—100,1994.

15. Wines, R. E., Wiftes, J. T., and Golbey, R. B. Combination chemotherapy inmetastatic malignant melanoma: a randomized study of three DTIC-containing combinations. Cancer (Phila.), 41: 415—421, 1978.

16. Luikart,S. D., and Kirkwoo,J. M. A comparisonof vinblastine,bleomycinandcisplatin (VBD) with dacarbazine (DTIC) in metastatic melanoma. Proc. Am. Soc.Clin. Oncol., 1: 176, 1982.

17. Skorski, T., Nieborowska-Skorska, M., Wlodarski, P., Perrotti, D., Hoser, G.,Kawiak, 3, Majewski, M., Christensen, L., lozzo, R. V., and Calabreua, B. Treatmentof Philadelphia leukemia in severe combined immunodeficient mice by combinationof cyclophosphamide and bcr/abl antisense oligodeoxynucleotides. J. Nail. CancerInst., 89: 124—133,1997.

18. Skouteris, G. G., and Schroder, C. H. C-myc is required for the G,JG,-S transition ofprimary hepatocytes stimulated with a deleted form of hepatocyte growth factor.Biochem. J., 316: 879—886,1996.

19. Shichiri, M., Hanson, K. D., and Sedivy, J. M. Effects of c-myc expression onproliferation, quiescence, and the G,@to G, transition in nontransformed cells. CellGrowth Differ., 4: 93—104,1993.

20. Shibuya, H., Yoneyama, M., Ninomiya-Tsuji, J., Matsumoto, K., and Taniguchi, T.IL-2 and EGF receptors stimulate the hematopoietic cell cycle via different signalingpathways: demonstration of a novel role for c-myc. Cell, 70: 57—67,1992.

21. Barry, M. A., Behnke, C. A., and Eastman, A. Activation of programmed cell death(apoptosis) by cisplatin, other anticancer drugs, toxins and hyperthermia. Biochem.Pharmacol., 40: 2353—2362,1990.

22. Feun, L. G., Savaraj, N., Moffat, F., Robinson, D., Liebman, A., Hurley, J, Raub,w. A., Jr., and Richman,S. P. PhaseII trial of recombinationinterferon-awithBCNU, cisplatin, DTIC and tamoxifen in advanced malignant melanoma. MelanomaRes., 5: 273—276,1995.

289

on April 1, 2016. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

1998;58:283-289. Cancer Res   Gennaro Citro, Igea D'Agnano, Carlo Leonetti, et al.   Mice

and in Nudein Vitroof Cisplatin in Melanoma Chemotherapy Antisense Oligodeoxynucleotides Enhance the Efficacymycc-

  Updated version

  http://cancerres.aacrjournals.org/content/58/2/283

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

  [email protected] at

To request permission to re-use all or part of this article, contact the AACR Publications

on April 1, 2016. © 1998 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from