+ All Categories
Home > Documents > Synergy of Topical Toll-like Receptor 7 Agonist with Radiation and Low-Dose Cyclophosphamide in a...

Synergy of Topical Toll-like Receptor 7 Agonist with Radiation and Low-Dose Cyclophosphamide in a...

Date post: 13-Nov-2023
Category:
Upload: sacklerinstitute
View: 0 times
Download: 0 times
Share this document with a friend
12
2012;18:6668-6678. Published OnlineFirst October 9, 2012. Clin Cancer Res M. Zahidunnabi Dewan, Claire Vanpouille-Box, Noriko Kawashima, et al. Breast Cancer Low-Dose Cyclophosphamide in a Mouse Model of Cutaneous Synergy of Topical Toll-like Receptor 7 Agonist with Radiation and Updated Version 10.1158/1078-0432.CCR-12-0984 doi: Access the most recent version of this article at: Material Supplementary http://clincancerres.aacrjournals.org/content/suppl/2012/10/09/1078-0432.CCR-12-0984.DC1.html Access the most recent supplemental material at: Cited Articles http://clincancerres.aacrjournals.org/content/18/24/6668.full.html#ref-list-1 This article cites 40 articles, 18 of which you can access for free at: Citing Articles http://clincancerres.aacrjournals.org/content/18/24/6668.full.html#related-urls This article has been cited by 1 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] Department at To order reprints of this article or to subscribe to the journal, contact the AACR Publications Permissions . [email protected] To request permission to re-use all or part of this article, contact the AACR Publications Department at American Association for Cancer Research Copyright © 2012 on December 17, 2012 clincancerres.aacrjournals.org Downloaded from Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984
Transcript

  

2012;18:6668-6678. Published OnlineFirst October 9, 2012.Clin Cancer Res M. Zahidunnabi Dewan, Claire Vanpouille-Box, Noriko Kawashima, et al. Breast CancerLow-Dose Cyclophosphamide in a Mouse Model of Cutaneous Synergy of Topical Toll-like Receptor 7 Agonist with Radiation and  

  

Updated Version 10.1158/1078-0432.CCR-12-0984doi:

Access the most recent version of this article at:

MaterialSupplementary

http://clincancerres.aacrjournals.org/content/suppl/2012/10/09/1078-0432.CCR-12-0984.DC1.htmlAccess the most recent supplemental material at:

  

Cited Articles http://clincancerres.aacrjournals.org/content/18/24/6668.full.html#ref-list-1

This article cites 40 articles, 18 of which you can access for free at:

Citing Articles http://clincancerres.aacrjournals.org/content/18/24/6668.full.html#related-urls

This article has been cited by 1 HighWire-hosted articles. Access the articles at:

  

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

SubscriptionsReprints and

[email protected] atTo order reprints of this article or to subscribe to the journal, contact the AACR Publications

[email protected]

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

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

Cancer Therapy: PreclinicalSee related commentary by Kohrt, p. 6571

Synergy of Topical Toll-like Receptor 7 Agonist withRadiation and Low-Dose Cyclophosphamide in a MouseModel of Cutaneous Breast Cancer

M. Zahidunnabi Dewan1, Claire Vanpouille-Box1, Noriko Kawashima1, Sara DiNapoli1, James S. Babb2,Silvia C. Formenti3, Sylvia Adams4, and Sandra Demaria1

AbstractPurpose: This study tested the hypothesis that topical Toll-like receptor (TLR) 7 agonist imiquimod

promotes antitumor immunity and synergizes with other treatments in a model of skin-involving breast

cancer.

Experimental Design: TSA mouse breast carcinoma cells were injected s.c. into syngeneic mice.

Imiquimod 5% or placebo cream was applied topically on the shaved skin overlying tumors three

times/wk. In some experiments, local ionizing radiation therapy (RT) was delivered to the tumor in three

fractions of 8 Gy, given on consecutive days. Cyclophosphamide was given intraperitoneally (i.p.) in one

dose of 2 mg/mouse. Mice were followed for tumor growth and survival.

Results: Treatment with imiquimod significantly inhibited tumor growth, an effect that was associated

with increased tumor infiltration by CD11cþ, CD4þ, and CD8þ cells, and abolished by depletion of CD8þ

cells. Administration of imiquimod in combination with RT enhanced significantly tumor response

compared with either treatment alone (P < 0.005), and 11% to 66% of irradiated tumors completely

regressed. Importantly, the addition of topical imiquimod also resulted in growth inhibition of a secondary

tumor outside of the radiation field. Low-dose cyclophosphamide given before start of treatment with

imiquimod and RT further improved tumor inhibition and reduced tumor recurrence. Mice that remained

tumor-free rejected a tumorigenic inoculum of TSA cells, showing long-term immunologic memory.

Conclusions: Topical imiquimod inhibits tumor growth and synergizes with RT. Addition of cyclo-

phosphamide further increases the therapeutic effect and induces protective immunologic memory,

suggesting that this combination is a promising strategy for cutaneous breast cancer metastases. Clin Cancer

Res; 18(24); 6668–78. �2012 AACR.

IntroductionBreast cancer usually arises from the mammary ducts,

which are ectodermal appendages. This may account for itspredilection for cutaneous metastases (epidermotropism).While some locally advanced tumors may directly infiltratethe adjacent skin, true cutaneous metastases are defined ascancers involving dermal or subcutaneous tissue that is

not contiguous with the primary tumor. Skin metastasesoccur in up to 10%ofmetastatic tumors, with breast cancershaving the second highest incidence/prevalence of skininvolvement after melanoma (1). A retrospective analysisof 420 patients with cutaneous metastases identified pri-mary breast carcinoma in 212 patients (30%), a meta-analysis reported an incidence of 24% (457 of 1,903; refs. 1,2). These metastases frequently occur in the vicinity of theoriginal tumor, such as at the chest wall or its overlying skinfollowing amastectomy or on the skin of the residual breasttissue after a segmental mastectomy. Initially, skin lesionsare generally managed by surgical resection and postoper-ative radiation, but they frequently recur. Chest wall recur-rences of breast cancer often herald or are associated withmetastatic disease in vital organs. Overall, the prognosisis relatively poor with a mean survival of 31 monthsfrom the diagnosis of skin metastases (1). Importantly,chest wall lesions often progress to become ulcerated, withcommon bleeding and infection. These complications havea detrimental impact on quality of life and particularlywarrant novel therapies. Toll-like receptor (TLR) agonistsare included in the ranked National Cancer Institute list of

Authors' Affiliations: Department of 1Pathology, 2Radiology, 3RadiationOncology, and 4Medicine, New York University School of Medicine, andNYU Langone Medical Center, New York, New York

Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

S. Adams and S. Demaria contributed equally to this work and share seniorauthorship.

Corresponding Author: Sandra Demaria, Department of Pathology,MSB-521, NYU Langone Medical Center, 550 First Avenue, New York,NY 10016. Phone: 212-263-7308; Fax: 212-263-8211; E-mail:[email protected]

doi: 10.1158/1078-0432.CCR-12-0984

�2012 American Association for Cancer Research.

ClinicalCancer

Research

Clin Cancer Res; 18(24) December 15, 20126668

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

immunotherapeutic agents with the highest potential totreat cancer (3, 4). Among them, the TLR7 agonist imiqui-mod is U.S. Food and Drug Administration–approved as atopical treatment for premalignant and early skin cancersand genital warts. Imiquimod is a very effective treatmentfor superficial basal cell carcinoma, achieving completetumor eradication in the majority of patients, mediated bythe activation of innate and adaptive immune responses (5,6). Activation of antitumor immune responses by imiqui-mod is mediated via TLR7 signaling which enhances den-dritic cell (DC)maturation andantigenpresentation ability.Imiquimod also promotes T-helper (TH)1 skewing andincreasedT-cell homing to the tumor, in part, by the indirecteffect of type I IFN derived from plasmacytoid DC (pDC)activated by imiquimod (7–9). In addition, direct pro-apoptotic effects on cancer cells that could contribute totumor responses have been described (10).The success of imiquimod in treating superficial skin

cancers has prompted its testing in other tumors which areamenable to topical therapy, such as primary andmetastaticmelanoma, with some encouraging but limited results (11–13). Because of their accessibility, unresectable breast cancerskinmetastases provide the ideal experimental setting to testnovel combinations of local treatments andoffer the oppor-tunity to test immune-mediated effects on systemic disease.The combination of TLR stimulation with other treat-

ments is an area of active investigation. Synthetic agonists ofTLR9 were shown to synergize with local ionizing radiationtherapy (RT) by inducing antitumor immune responses inmice models and early clinical trials (14–16). The combi-nation of TLR9 agonist and cyclophosphamide (CY) hasalso been shown to improve survival compared to eachtreatment alone in a model of embryonal rhabdomyosar-coma (17). Cyclophosphamide was also tested in combi-

nation with 2 TLR7/8 agonists designed for systemic ratherthan topical administration and showed additive effects in amousemodel of colon carcinoma (18). The combination oftopical imiquimodwith local RT and/or cyclophosphamidefor skin metastases of breast or other cancers has not beentested before.

Here, we evaluated the antitumor efficacy of imiquimodin the syngeneic and poorly immunogenic TSA mousemodel of breast cancer. We show that topical imiquimoddirectly applied to established tumors is an effective localtreatment for TSA cells, which are TLR7-negative, and cansynergize with local RT. Moreover, addition of low-dosecyclophosphamide, a strategy that has been shown to haveimmunomodulatory function in both preclinical and clin-ical studies (19–21), increased the strength anddurability ofthe antitumor immune response.

Materials and MethodsMice

Six to 8-week-old BALB/c mice were obtained from Taco-nic Animal Laboratory and maintained under pathogen-free conditions in the animal facility at NewYorkUniversityLangoneMedical Center (NewYork, NY). All animal experi-ments were carried out according to protocols approved bythe Institutional Animal Care and Use Committee of NewYork University.

Cells and reagentsTSA is a BALB/c mouse–derived poorly immunogenic

mammary carcinoma cell line (22). Cells were cultured inDulbecco’s Modified Eagle’s Medium (DMEM; InvitrogenCorporation) with 2 mmol/L L-glutamine, 100 U/mL pen-icillin, 100 mg/mL streptomycin, 2.5 � 10�5 mol/L 2-mer-capthoethanol, and 10% FBS (Gemini Bio-Products; com-plete medium), and routinely monitored and found to befree of contamination by mycoplasma by the MycoplasmaDetection Kit (Roche Diagnostics). TLR7 expression wasdetermined by reverse transcriptase PCR (RT PCR) usingprimers specific for mouse TLR7 and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) as previouslydescribed (23). Five percent imiquimod (Aldara) and aplacebo control creamwere supplied by Graceway Pharma-ceuticals. Cyclophosphamide monohydrate (Sigma-Aldrich) was dissolved in water at 20 mg/mL and preparedfresh before use.

In vitro toxicity assayTSA (1.5 � 104 per well) cells were cultured in a 96-well

plate in complete mediumwith reduced serum (1% FBS) inthe presence of imiquimod or vehicle control, and viabilitywasmeasured at 24, 48, or 72hours using theCell CountingKit-8 (Dojindo Molecular Technologies, Inc.).

Tumor challenge and treatmentBALB/c mice were injected s.c. with 1 � 105 TSA cells in

the right flank (primary tumor) on day 0. On day 10, whentumors were palpable, animals were randomly assigned toplacebo or imiquimod treatment. A small amount (�35mL)

Translational RelevanceToll-like receptor agonists can alter the tolerogenic

and immunosuppressive tumor microenvironment andpromote antitumor immune responses. TLR7 agonistimiquimod is a powerful immune response modifierwithproven anticancer activitywhen applied topically toprimary skin tumors. Although breast cancer metastasesto the skin can be extensive, difficult to treat, and asso-ciatedwith significant patient discomfort andmorbidity,their superficial location lends them to topical therapy.In a mouse breast cancer model, we show that topicalimiquimod inhibits the growth of the treated tumor andof a secondary untreated tumor. Importantly, therapeu-tic synergy was seen between imiquimod and localradiotherapy, which is frequently used in the clinic totreat breast cancer skin metastases. Addition of a singlelow dose of cyclophosphamide reduced tumor recur-rence and further promoted a protective immunologicmemory response, supporting the future investigation ofthis triple combination in patients.

TLR7 Stimulation and Radiotherapy for Breast Cancer

www.aacrjournals.org Clin Cancer Res; 18(24) December 15, 2012 6669

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

of cream was applied on the shaved skin over the tumor(corresponding to�87.5 mg/kg of the active compound) 3times/wk and gently rubbed in until completely adsorbed.In some experiments, TSA cells were injected in the left flankon day 2 (secondary tumor) as previously described (24).Perpendicular tumor diameters were measured with a Ver-nier caliper, and tumor volumes were calculated as length�width2 � 0.52. In some experiments, mice were followeduntil the tumor reached an average diameter of 12 mm(corresponding to �5% body weight), according to insti-tutional guidelines.

RT was administered as previously described (24, 25).Briefly, all mice (including mice receiving mock radiation)were lightly anesthetized by intraperitoneally (i.p.) injec-tion of Avertin (240mg/kg), and positioned on a dedicatedplexiglass tray. Radiotherapy was delivered in 3 fractions of8 Gy given on days 12, 13, and 14 to a field including thetumor with 5 mmmargins using a Clinac 2300 C/D LinearAccelerator (Varian Medical Systems) fitted with a 25-mmRadioSurgery conical collimator (BrainLAB AG), which isdesigned to deliver very sharp and limited radiation dosefields. Superflab bolus (1.5-cm tissue equivalent material)was placed over the tumor, and a source-to-skin distance(SSD) of 100 cm was set. RT was delivered at 600 cGy/minwith 6 MV X-rays. Cyclophosphamide was given i.p. at 2mg/mouse on day 9.

In vivo T-cell subset depletionDepletion of T-cell subsets was achieved by injecting anti-

mouse CD4 GK1.5 mAb or anti-mouse CD8 2.43 mAb(BioXCell) i.p. at 100 mg/mouse on day 7, 8, and 9 post-tumor cell inoculation. The depletion was maintained byrepeated weekly injections of mAb. Depletion was con-firmed by testing spleen cells for the presence of CD4þ andCD8þ T-cells using non–cross-reactive FITC-RMA4-4 andPE-anti-CD8b mAb (BD PharMingen).

Immunostaining of tumor sectionsTumors were harvested at day 26 postinoculation, fixed

for 1 hour at 4�C in 4% paraformaldehyde followed byovernight incubation in 30% sucrose, and frozen in opti-mum cutting temperature (OCT) medium. Sections (8 mm)were stained as previously described (24) with phycoery-thrin (PE)–Texas Red–conjugated rat anti-mouse CD4 orPE-conjugated rat anti-mouse CD8a (Caltag) or PE-conju-gated Hamster anti-mouse CD11c (BD Pharmingen) andcounterstained with 5 mg/mL 40,6-diamidino-2-phenylin-dole (DAPI; Sigma). Images were obtained with the use of aNikon Eclipse 800 deconvolution microscope. The num-bers of CD4þ, CD8þ T cells and CD11cþDCs were countedin 3 randomly selected fields (�20) in each tumor.

Measurements of interleukin-10 by ELISATumors, serum, tumor-draining lymph node (TDLN),

and spleen were harvested at day 26 from TSA tumor–bearing mice (n ¼ 6/group) treated with topical placeboor imiquimod. Tumor lysates were prepared by mincingtumors in lysis buffer containing 10 mmol/L Tris (pH 8.0,)

0.5%Nonidet P-40, 250mmol/LNaCl, 10mmol/L sodiumorthovanadate, 100 mmol/L phenylmethylsulfonylfluoride,1 mg/mL leupeptin, 1 mg/mL pepstatin, and 1 mg/mL apro-tinin (all from Sigma-Aldrich). After sonication and incu-bation on ice for 30 minutes, lysate supernatants wereobtained by centrifugation at 13,000 rpm for 20 minutesat 4�C. Spleen and TDLN cells (4 � 106 cells/mL) werecultured in 96-well tissue culture plates for 24hours in T-cellmedium [RPMI-1640 supplemented with 2 mmol/L L-glu-tamine, 100 U/mL penicillin, 100 mg/mL streptomycin, 5�10�5 mol/L 2-mercapthoethanol, 10% FBS, and 10 U/mLhuman recombinant interleukin (IL)-2] in the presence ofphorbol-12-myristate 13-acetate (PMA; 5 ng/mL) and iono-mycin (500 ng/mL). Cell-free supernatants were collectedand stored at �80�C. IL-10 was measured by ELISA (R&DSystems). Samples from 2 mice were pooled to obtain 3independent samples of the 6 mice analyzed in each treat-ment group.

Flow cytometrySingle-cell suspensions were obtained by digestion of

treated and untreated tumors with 0.2 mg/mL DNase and1.67 W€unschU/mL Liberase (Roche) as previouslydescribed (26). The Fixable Viability Dye eFLuor 660(eBioscience) was used to distinguish live and dead cells.Aliquots of 106 cellswere incubatedwith anti-mouseCD16/32 (Fc block) for 10 minutes followed by staining for 30minutes with Pacific Blue-anti-CD45 mAb, PE-anti-H2-Kd,FITC-anti-H2-Ld, and FITC-anti-ICAM-1 (BD Pharmingen).Lymphocytes isolated from spleen and peripheral bloodwere stainedwith FITC-anti-CD4mAb,PE-Cy-5-anti-CD25,and PE-anti-FoxP3 mAb using the Fixation/Permeabiliza-tionConcentrate andDiluent Kit (eBioscience). All sampleswere analyzed using a LSRII flow cytometer and FlowJoversion 7.6.3 (Tree Star).

Tumor-specific production of IFN-g by TDLN cellsTDLN cells (0.5 � 106) were cultured in 48-well tissue

culture plates with 1 mmol/L AH1 peptide (SPSYVYHQF), aCD8 T-cell epitope known to be an immunodominantantigen in TSA cells (22), or control peptide derived fromMCMV (YPHFMPTNL; GenScript) for 72 hours in T-cellmedium. IFN-g concentration was measured in cell-freesupernatant of duplicate well using flowcytomix kit(eBioscience).

Statistical analysisThe Student t test was used to compare the effect of

imiquimod versus vehicle control in in vitro toxicity assays,levels of IL-10 and surface molecules in placebo versusimiquimod-treated mice. Mixed model ANOVA was usedto compare treatment arms with respect to mean tumorvolumes at each day of measurement. The dependent var-iable was composed of the tumor volumes observed for allanimals at all time points of observation. The modelincluded treatment and time as fixed classification factors.The Tukey multiple comparison procedure was used tocompare treatment arms with respect to tumor volume at

Dewan et al.

Clin Cancer Res; 18(24) December 15, 2012 Clinical Cancer Research6670

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

each day of measurement while keeping the family-wisetype I error rate for the set of comparisons below thenominal 5% level. Random coefficients regression was usedto assess the effect of treatment on tumor growth. Thedependent variable was the natural log of tumor volumeat all available time points; log volumes were used in placeof observed volumes as the change in log volume over timewas well approximated as linear. For both the ANOVA andrandom coefficients analyses, the correlation structure wasmodeled by assuming observations to be correlated onlywhen acquired from the same animal with the strength ofcorrelation between observations inversely dependent onthe elapsed time between the observations. Treatment armswere compared in terms of survival, defined as time to deathdue todisease or sacrifice, using log-rank tests. All reportedPvalues are 2-sided and were declared statistically significantat the 5% level. The statistical computationswere carriedoutusing SAS for Windows, version 9.3 (SAS Institute).

ResultsImiquimod toxicity in vitro is minimal for TLR7-negative TSA mouse breast cancer cellsTLRs are expressed by many epithelial tumors, including

breast cancer (27). Expression of TLR7 has been reported insome human breast cancer cell lines, although the func-tional consequences remain undefined (28). To determinewhether imiquimod has therapeutic activity against cuta-neous metastases of breast cancer, we used a mouse modelof poorly immunogenic breast cancer, which mimics thesituation of patients with cancer with chest wall recurrenceas TSA tumor cells form subcutaneous nodules that invadethe dermis and ulcerate through the skin. By RT-PCR anal-ysis, expression of TLR7 was not detected in TSA cells (datanot shown). TSA cells did not show a significant reductionin viability at 24 hours when cultured in the presence ofimiquimod (Supplementary Fig. S1). However, a modestbut significant toxicity (P < 0.005) was seen at 48 hours inthe presence of 10 mg/mL of imiquimod, and at 48 hours (P< 0.05) in the presence 5 mg/mL of imiquimod. Overall,results suggest that imiquimod is toxic for TSA cells only atrelatively high concentrations and this effect is independentfrom TLR7 expression, consistent with the previouslyreported pro-apoptotic effect of imiquimod in somehumanepithelial cancer cells (10).

Topical imiquimod inhibits TSA tumor growth in vivoby a CD8-dependent mechanismTo determine the effect of imiquimod in vivo, imiquimod

orplacebo creamwas applied onto the skinover TSA tumorsstarting at day 10 postinjection of tumor cells, when tumorsbecame palpable (Fig. 1A). Imiquimod significantly inhib-ited tumor growth (P < 0.0001 compared with placebo) butdid not induce any complete tumor regression (Fig. 1B,top). Analysis at day 26 showed increased infiltration byCD11cþ DC and CD4þ and CD8þ T cells in imiquimod-treated tumors (Fig. 1C and D). The antitumor effect oftopical imiquimod was abrogated by depletion of CD8þ

cells (Fig. 1B, bottom), indicating that it was mediated by

activation of antitumor CD8þ T cells and/or CD8þ pDCs(29). Conversely, CD4þ T cells may play a suppressive roleas their depletion improved significantly (P < 0.005) tumorinhibition. A marked increase in the levels of the immu-nosuppressive cytokine IL-10, produced by CD4þ T cells,was recently reported in the spontaneous tumors of neu-transgenic mice upon topical imiquimod treatment (30).Measurements of IL-10 levels showed no significant changein the tumor and TDLNs of imiquimod-treated mice (Fig.1E). However, a modest but statistically significant increasewas detected in the serum, as well as the supernatants of exvivo stimulated spleen cells, consistent with the increasedserum levels reported in neu-transgenic mice (30). Overall,in the TSAmodel, there was no evidence of IL-10 inductionat the site of topical imiquimod treatment, possibly becauseIL-10 was already elevated in placebo-treated tumors (Fig.1E). However, increased IL-10 production in ex vivo stim-ulated spleen cultures indicates that a population of IL-10–producing T cells is expanded in imiquimod-treated mice.

Topical imiquimod synergizes with local radiotherapyin inhibition of the irradiated tumor and of a tumoroutside of the radiation field

We have previously shown that RT of established TSAtumors given as a single dose of 20Gy, 3 fractions of 8Gy, or5 fractions of 6 Gy caused a similar tumor growth inhibi-tion, but no tumor eradication was achieved by any of theseradiation regimens, and there was no effect on a secondarytumor outside of the radiation field (24). Importantly, onlyfractionated RT showed synergy with anti-CTLA-4 antibodyin inducing antitumor immune responses leading to erad-ication of the majority of the irradiated tumors and oftumors outside of the radiation field (abscopal effect), withthe best synergy seen with 8 Gy � 3 (24). Therefore, theregimen of 8 Gy� 3 was chosen for testing in combinationwith imiquimod. In the first experiment, mice bearing asingle tumor were treated with topical imiquimod startingat day 10, and RT was delivered to the tumor at days 12, 13,and 14 (Fig. 2A). As expected, RT and imiquimod asmonotherapy delayed tumor growth but did not inducecomplete regression. In contrast, the majority of tumorsreceiving imiquimod þ RT had completely regressedbetweendays 25 and30 (Fig. 2B andC). Randomcoefficientregression analysis confirmed a significant interactionbetween RT and imiquimod (P ¼ 0.0115), consistent witha synergistic effect of this combination.

To determine whether the antitumor effect of imiqui-mod, alone or in combination with RT, is localized to thetreated area or could extend to other skin/chest wall sitesinvolved by tumor, TSA cells were injected at 2 separatedsites, defined as "primary" and "secondary" tumors (Fig. 3Aand B). At start of treatment both tumors were palpable, butthe secondary tumors were smaller than primary tumors(mean volume, 8.1 � 4.7 vs. 21.8 � 7.5 mm3) as TSA cellswere injected with 2-day delay. RT was delivered only to theprimary tumor, and we have previously shown that RTalone does not affect growthof the secondary tumor outsideof the radiation field (24). Imiquimod applied to the

TLR7 Stimulation and Radiotherapy for Breast Cancer

www.aacrjournals.org Clin Cancer Res; 18(24) December 15, 2012 6671

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

primary tumor caused a significant tumor inhibition thatwas enhanced by administration of RT, leading to completeregression of 30% of the primary tumors by day 25 (Fig.3C), confirmingdata shownabove (Fig. 2). Treatment of thesecondary tumor with imiquimod did not alter the effect ofimiquimod alone or with RT at the primary tumor. Inter-estingly, treatment of the primary tumor with imiquimodinhibited the growth of the secondary tumor, an effectthat was not further enhanced by treating also the sec-ondary tumor with imiquimod (Fig. 3D). However, appli-cation of imiquimod to the secondary tumor significantlyenhanced inhibition of the secondary tumor when theprimary tumor was treated with imiquimod þ RT. Overallresults suggest that imiquimod has both direct (i.e., at thesite of application) and indirect antitumor effects. Part ofthe direct effects of imiquimod may result in sensitizationof a tumor outside of the radiation field to antitumor

immune responses triggered by concomitant use of RTand imiquimod at another tumor site. To determinewhether the expression of molecules known to facilitatetumor rejection by T cells was increased in tumors treatedwith topical imiquimod, TSA cells freshly isolated fromplacebo and imiquimod-treated mice were analyzed exvivo by flow cytometry. Imiquimod induced a statisticallysignificant increase in the levels of 2 MHC class I allelesand of intercellular adhesion molecule-1 (ICAM-1; Fig.4A). Analysis of TDLNs from mice treated with imiqui-mod þ RT confirmed the development of T-cell responsesto the CTL epitope AH1, which is derived from an immu-nodominant antigen in TSA cells and presented by H2-Ld

(ref. 22; Fig. 4B). These data support the hypothesis thatimiquimod induces immune-mediated tumor inhibitionby promoting both priming and effector phases of anti-tumor T-cell responses.

Figure 1. Topical imiquimodinhibits growth of TSA tumorsin vivo. A, treatment schema.Placebo (PLA) or imiquimod (IMQ)5% cream was applied topicallyas indicated. B, tumor growth inmice treated with PLA (closedcircles), IMQ (closed squares),IMQ and CD4þ cell depletion(closed triangles), or IMQand CD8þ cell depletion(closed diamonds). Top, datashow the mean � SEM of10 mice/group. Bottom, CD4þ

and CD8þ cell depletion wasstarted at day 7. Data show themean �SEM of 6 mice/group. Cand D, immune cell infiltration oftumors from mice treated withPLA or IMQ at day 26. C,representative fields (�200)showing CD11cþ, CD8þ,and CD4þ cells (orange). Nucleiwere stained with DAPI (green).D, number of CD11cþ, CD8þ, andCD4þ cells infiltrating TSA tumorsquantified in 3 mice/group. E,IL-10 concentration measured atday 26 in tumor lysates, serum,and supernatants of TDLNsand spleen cells stimulatedex vivowithPMAand ionomycin in6 mice/group. Data arerepresentative of 2 experiments.���, P < 0.0005; ��, P < 0.005;�, P < 0.05.

Dewan et al.

Clin Cancer Res; 18(24) December 15, 2012 Clinical Cancer Research6672

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

Low-dose cyclophosphamide improves local controland reduces recurrence of tumors treated with RT andtopical imiquimodThe inhibition of tumor growth by imiquimod treat-

ment alone was reported to be only temporary in neu-transgenic mice as tumors resumed growth after topicalimiquimod was stopped (30). To determine whether thecomplete tumor regression observed after treatment withthe combination of imiquimod þ RT was durable, micefrom the experiment in Fig. 2 were followed over time. Alltumors recurred between 20 and 30 days after discontin-uation of imiquimod treatment at day 35, with thelongest tumor-free period of 40 days, indicating that theimmune response triggered by treatment was unable toeliminate all cancer cells and not potent and/or durableenough to prevent tumor regrowth. An attractive strategyto improve antitumor immune responses is the use oflow-dose cyclophosphamide, a chemotherapy drug thathas been shown to have immunomodulatory function inboth preclinical and clinical studies (19–21). The optimaldose and schedule of cyclophosphamide to enhance theeffect of vaccination have been previously defined byMachiels and colleagues to be 100 mg/kg administered1 day before vaccination in a mouse breast cancer model(19). At this dose, cyclophosphamide caused a significantreduction of regulatory T (Treg) cells detectable in thespleen and peripheral circulation 3 days after adminis-tration to tumor-bearing mice (Fig. 5A and B). Therefore,we tested the effect of cyclophosphamide administered at100 mg/kg 1 day before the first dose of imiquimod.Cyclophosphamide by itself caused a significant tumorgrowth delay (P < 0.0005 cyclophosphamide þ placebovs. placebo) and enhanced tumor growth inhibition byimiquimod, although the difference was not significant (P

> 0.05; imiquimodþ cyclophosphamide vs. imiquimod),and no complete tumor regression was achieved (Fig.5C). Interestingly, IFN-g responses to the tumor-specificCTL epitope AH1 were the highest in TDLNs of micetreated with the combination of imiquimod þ cyclophos-phamide than single treatment with cyclophosphamideor imiquimod, alone (Fig. 5D). The increase showed atrend to significance when compared with imiquimodalone (P ¼ 0.087). A single dose of cyclophosphamidehad no effect on serum levels of IL-10 but diminishedsignificantly IL-10 production induced by imiquimodtreatment in ex vivo stimulated spleen cells (Fig. 5E andF). Overall, data suggest that cyclophosphamide mitigatesat least some of the immunosuppressive mechanismsresulting in improved antitumor T-cell responses.

Next, the effect of cyclophosphamide was tested in micereceiving RT with or without imiquimod (Fig. 6A). Micereceiving cyclophosphamide in combination with RT andimiquimod showed a significant improvement in tumorinhibition compared to all other groups (P < 0.0005;cyclophosphamide þ imiquimod þ RT vs. placebo andimiquimod þ RT, and P < 0.005 vs. cyclophosphamide þRT; Fig. 6B). Median survival was significantly increased inall treated mice compared with placebo (P < 0.0005) andwas 31 days for placebo, 52 days for imiquimod þ RT, 49days for cyclophosphamide þ RT, and 73 days for cyclo-phosphamide þ imiquimod þ RT (Fig. 6C). Completetumor regression occurred between days 24 and 31 andwas seen in 50% of mice treated with cyclophosphamideþimiquimod þ RT, 30% of mice treated with cyclophospha-mideþ RT, and 11% ofmice treated with imiquimodþ RT.Although some tumors continued regressing after termina-tion of imiquimod administration at day 24 posttumorinoculation, tumor recurrence was eventually seen in all

Figure 2. RT and imiquimodsynergize in inducing TSA tumorregression. A, treatment schema.PLA or IMQ 5% cream was appliedtopically and RT was given in 3fractions of 8 Gy as indicated. B,tumor growth delay in mice treatedwith PLA (closed circles), IMQ (opencircles), RT þ PLA (closed squares),and RT þ IMQ (open squares). Dataare the mean � SEM. The number ofmicewith complete tumor regressionover the total number of mice pergroup is indicated. C, representativephotographs of tumors duringtreatment and 4 days after end oftreatment, showing progressiveulcerated tumors that are smaller inmice treated with monotherapy butonly showcomplete regression in theRT þ IMQ combination group. Dataare representative of 2 experiments.���, P < 0.0005.

TLR7 Stimulation and Radiotherapy for Breast Cancer

www.aacrjournals.org Clin Cancer Res; 18(24) December 15, 2012 6673

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

mice except 3mice in cyclophosphamideþ RT group and 3mice in cyclophosphamide þ imiquimod þ RT group,which remained tumor-free 65 days after termination ofimiquimod administration. Mice that remained tumor-freeat day 90were rechallengedwith a tumorigenic inoculumofTSA cells togetherwith a groupof na€�vemice.While all na€�vemice developed tumors by day 10 postchallenge, all mice

H2-Kd

H2-Ld

ICAM-1

FL2

Count

0

200

400

600

800

1,000

PLA IMQ

MF

I ±

SD

0

60

120

180

240

300

360

420

PLA IMQ

MF

I ±

SD

0

40

80

120

160

200

240

280

PLA IMQ

MF

I ±

SD

*

*

*

A

B 150

125

100

75

50

25

0

IFN

-γ p

g/m

L

PLA RT+IMQ

*

Figure 4. Topical imiquimod upregulates MHC class I moleculeand ICAM-1 on tumor cells and induces tumor-specific T-cellresponses in TDLN in combination with RT. A, mice were injectedwith TSA cells on day 0. PLA or IMQ 5% cream was applied topicallyon days 10, 12, 14, and 17, and tumors were harvested on day 18.Obtained single-cell suspensions were stained with Pacific Blue-anti-CD45, PE-anti-H2-Kd, FITC-anti-H2-Ld, and FITC-anti-ICAM-1.Histograms show the expression of H2-Kd, H2-Ld, and ICAM-1, onthe gated CD45-negative TSA cells treated with PLA (gray lines) orIMQ (black lines). Cells stained with isotype control (dashed graylines). Bar graphs show mean fluorescence intensity (MFI) of3 samples �SD. B, production of IFN-g by TDLN cells stimulatedwith the tumor-specific CTL epitope AH1 (full circles) or controlpeptide (empty circles). TDLN cells were harvested at day 26 fromLN draining the primary tumor of mice treated RT þ imiquimod, as inFig. 3. Each symbol represents one mouse. Horizontal lines indicatethe mean of AH1 (solid line) or control (dotted line) peptide-stimulated cultures. �, P < 0.05.

Figure 3. Topical imiquimod sensitizes a secondary tumor to the abscopaleffect induced by treatment with IMQ þ RT of a primary tumor. A,treatment schema. Mice were injected s.c. with TSA cells into the right(defined as "primary" tumor) and left (defined as "secondary" tumor) flankon days 0 and 2, respectively. PLA or IMQ 5% cream was appliedtopically as indicated. RT was administered locally exclusively to theprimary tumor as indicated. B, schematic representation of the tumormodel. Primary (C) andsecondary (D) tumor volumesat endof experiment(day 25) in mice receiving IMQ to the primary only or to both primary andsecondary tumors, as indicated. Number above the columns indicatesmice with complete tumor regression over the total number of micetreated. Data show themean� SEM of 10 to 12mice/group and are from2 independent experiments combined. ���, P < 0.0005; ��, P < 0.005.

Dewan et al.

Clin Cancer Res; 18(24) December 15, 2012 Clinical Cancer Research6674

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

that had survived tumor-free after treatment with cyclo-phosphamide þ imiquimod þ RT and 2 of 3 mice treatedwith cyclophosphamide þ RT rejected the challenge andremained healthy until the end of the follow-up period 2months after re-challenge.Collectively, these results indicate that a single low-dose

cyclophosphamide promotes the development of long-term protective immunity when combined with RT andthat addition of imiquimod improves local tumor controland survival in mice treated with cyclophosphamide þ RT,supporting further testing of combinations of TLR7 agonistswith chemotherapy and radiation.

DiscussionIn this study, we show in an experimental breast cancer

model of skin metastasis that a local immune response

modifier can be combined with local radiotherapy andsystemic low-dose chemotherapy to induce effective anti-tumor immunity with the potential to eliminate the tumor.The appeal of this approach is that it exploits the accessi-bility of breast cancer skin metastases to convert a difficulttherapeutic problem into an opportunity for in situ vacci-nation of the patients against the tumor.

Topically applied imiquimod inhibited TSA tumorgrowth. This effect was associated with increased tumorinfiltration by DC and T cells and was abolished bydepletion of CD8þ cells. Similar CD8 dependence of thetherapeutic effect of topical imiquimod was previouslydescribed by Lu and colleagues in the neu-transgenicmodel of breast cancer (30). In the latter, tumor-specificCD8þ T cells were increased in imiquimod-treated mice,and local tumor control was associated with decreased

Figure 5. Low-dosecyclophosphamide reduces Tregcells and IL-10 production andenhances tumor-specific IFN-gproduction by TDLNs. A and B, micewere injected s.c. with TSA cells onday 0. Cyclophosphamide wasadministered i.p. on day 9 in a singledose of 100 mg/kg, and spleen (A)and blood (B) were analyzed on day12 for the presence of Treg cells bystaining for CD4, CD25, and FoxP3.Data are the mean � SD of 3 mice/group and are representative of 2experiments. C, tumor growth delayin mice treated with PLA (opensquares), IMQ (closed triangles),cyclophosphamide þ placebo(PLA; open circles), andcyclophosphamide þIMQ(closed squares). PLA and IMQwere administered as in Fig. 1.Cyclophosphamide was given onceat day 9. Data are themean�SEMof7 mice/group. D, production of IFN-gby TDLN cells harvested at day 18and stimulated with the tumor-specificCTL epitopeAH1 (full circles)or control peptide (empty circles).Each symbol represents one mouse.Horizontal lines indicate the mean ofAH1 (solid line) or control (dotted line)peptide-stimulated cultures. E and F,IL-10 concentrationmeasured at day18 in supernatants of ex vivostimulated spleen cells (E) and serum(F). Each symbol represents onemouse. Data are representative of2 experiments. ���, P < 0.0005;��, P < 0.005; �, P < 0.05.

TLR7 Stimulation and Radiotherapy for Breast Cancer

www.aacrjournals.org Clin Cancer Res; 18(24) December 15, 2012 6675

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

spontaneous lung metastases, an effect that could resultfrom decreased seeding from the treated tumor and/orinhibition by antitumor T cells that are active systemi-cally. However, recent evidence by Drobits and colleaguesindicates that the antitumor effect of topical imiquimod,at least within the first 1 or 2 weeks, can be entirelymediated by pDCs that acquire CD8 and lytic function(29). In a mouse melanoma model, they showed thatimiquimod induced increased tumor infiltration byCD8þ T cells and natural killer cells but neither wasrequired for tumor inhibition. It is, therefore, possiblethat CD8þ pDCs play a role in inhibition of the TSAtumors. However, evidence that imiquimod appliedto the primary tumor leads to inhibition of a secondarytumor that is not topically treated (Fig. 3) is consistentwith development of an adaptive immune response.Importantly, the long tumor-free interval (up to 40 days)seen in mice with complete tumor regression after treat-ment with the combination of imiquimod þ RT, and thedevelopment of long-term protective memory in micethat received cyclophosphamide þ imiquimod þ RTindicate that imiquimod can be used as an adjuvant toimprove the therapeutic efficacy and pro-immunogeniceffects of RT and low-dose cyclophosphamide (31).

Improved tumor inhibition observed inmice treatedwithimiquimodandCD4þT-cell depletion suggests thatCD4þTcells have a regulatory or suppressive role (Fig. 1B). Induc-tion of IL-10 secretion by a CD4þ T-cell subset differentfrom Treg cells in mice treated with imiquimod was previ-ously shown to be a mechanism of suppression of theantitumor immune response (30). However, in our model,imiquimod treatment did not lead to increased levels of IL-10 in tumor or TDLNs. A significant increase was noted inthe serum and in supernatants of ex vivo stimulated spleencells, suggesting that some induction of IL-10 production by

T cells may represent a negative feedback reaction to theincreased immune activation caused by imiquimod. Itremains to be determined whether this increased polariza-tion of T cells toward IL-10 production is responsible fortumor recurrence. Elevated levels of IL-10 were present inTSA tumors regardless of treatment and may be an imped-iment to tumor rejection. Interestingly, administration ofcyclophosphamide 1 day before the start of imiquimodtreatment decreased significantly IL-10 produced by spleencells (Fig. 5E) and showed a trend to increase IFN-g pro-duced by tumor-specific CD8 T cells (Fig. 5D). Althoughcyclophosphamide treatment did not significantly enhancethe therapeutic effect of imiquimod (Fig. 5C), in combina-tion with RT and imiquimod þ RT, cyclophosphamidepromoted thedevelopment of a durable antitumor immuneresponse able to prevent tumor recurrence and reject atumor challenge over 2 months after the end of treatment(Fig. 6). The best characterized immunomodulating effectof cyclophosphamide at non-lymphodepleting doses is itsability to selectively deplete and/or disable Treg cells (32,33) and uncover the therapeutic effect of immunotherapy(19, 20, 34). In mice treated at day 9 with cyclophospha-mide, there was a significant decrease in Treg cells at day 12in the spleen and peripheral circulation (Fig. 5A and B). Theability of cyclophosphamide given before RT to inducecomplete tumor regression and protective tumor-specificmemory in a fraction of TSA tumor–bearing mice indicatesthat cyclophosphamide can also uncover the ability of RT togenerate an in situ tumor vaccine (35). The relative contri-bution of Treg cells reduction, induction of an immuno-genic cell death, or other pro-immunogenic activities ofcyclophosphamide to this effect remains to be determined(36, 37). Importantly, the addition of topical imiquimodled to further improvement in the local tumor controlachieved by cyclophosphamide þ RT and a trend to

Figure 6. Low-dose cyclophos-phamide improves tumorinhibition by IMQ þ RT. A,treatment schema. Tumor growthdelay (B) and survival (C) inmice treated with placebo(PLA; n ¼ 17), IMQ þ RT (n ¼ 9),cyclophosphamide þ PLA þ RT(n¼ 13), and cyclophosphamide þIMQ þ RT (n ¼ 14), as indicated.Only a subset of mice in the controlgroup was followed for survival.The arrow indicates the day ofrechallenge of mice that hadremained tumor-free. Data showthe mean �SEM of 9 to 17 mice/group and are from 2 independentexperiments combined.���, P < 0.0005; ��, P < 0.005.

Dewan et al.

Clin Cancer Res; 18(24) December 15, 2012 Clinical Cancer Research6676

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

improved survival (Fig. 6B and C). About 42% ofmice withcomplete tumor regression following treatment with cyclo-phosphamide þ imiquimod þ RT remained tumor-free,whereas mice with complete regression after imiquimod þRT in different experiments all developed recurrent tumors.As cyclophosphamide was given only once before imiqui-mod and RT treatments started, it is likely that the strengthof the initial antitumor response induced is what deter-mines the long-term tumor control, whereas the cyclophos-phamide-mediated reduction in IL-10–secreting T cellsinduced by imiquimod may contribute to the persistenceof the response (30).In different experiments, the percentage of mice with

complete tumor regression following treatment with imi-quimod þ RT varied from 11% to 66%, with the largestpercentage seen in mice receiving imiquimod up to day 35(Fig. 2). Therefore, prolonged treatment with imiquimodmay be required to achieve optimal effects.Whether imiquimod is needed to sustain the immune

response or to sensitize tumor cells to immune rejectionremains to be established. The enhanced expression ofMHC class I and ICAM-1molecules induced by imiquimodon tumor cells (Fig. 4) suggests that the improved responseof the secondary tumor to the abscopal antitumor responsetriggered by treatment of the primary tumor with imiqui-mod þ RT can be due to better recognition of tumor by Tcells. However, other effects of imiquimod could contributeto this sensitization, including angiogenesis inhibition(38), and increased T-cell recruitment and function (39).We are currently conducting a comprehensive analysis ofthe changes induced by imiquimod in TSA tumors aswell asin patients with breast cancer (40) to address thesequestions.Overall, the data presented indicate that topical imi-

quimod is a valuable adjuvant for use in the treatment ofbreast cancer metastases involving the skin. Imiquimodcan improve antitumor immune responses by itself and inconcert with RT. The addition of low-dose cyclophospha-mide further enhanced these effects. The complexity ofthe microenvironment of a given tumor and the preexist-

ing regulatory and immunosuppressive networks are like-ly to be important determinants of the overall efficacy ofthe treatment. The combination of imiquimod with selec-tive chemotherapy agents, such as low-dose cyclophos-phamide, and RT regimens that have pro-immunogeniceffects (24), is a promising strategy that can be translatedin the clinic.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design:M.Z. Dewan, S.C. Formenti, S. Adams, S. DemariaDevelopment of methodology: M.Z. DewanAcquisitionofdata (provided animals, acquired andmanagedpatients,provided facilities, etc.): M.Z. Dewan, C. Vanpouille-Box, N. Kawashima,S. DiNapoliAnalysis and interpretation of data (e.g., statistical analysis, biosta-tistics, computational analysis): M.Z. Dewan, C. Vanpouille-Box, N.Kawashima, J.S. Babb, S.C. Formenti, S. Adams, S. DemariaWriting, review, and/or revision of the manuscript: M.Z. Dewan, C.Vanpouille-Box, J.S. Babb, S.C. Formenti, S. Adams, S. DemariaAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): M.Z. Dewan, N. Kawashima, S.DiNapoli, S.C. FormentiStudy supervision: S. Demaria

AcknowledgmentsThe authors thank the personnel of NYUCancer Institute FlowCytometry

and Experimental Pathology Histopathology core facilities and the Depart-ment of Radiation Oncology for expert assistance. They also thank GracewayPharmaceuticals, Inc., for providing imiquimod 5%and placebo creams andDr. Tze-Chiang Meng of Graceway Pharmaceutical for scientific suggestionsabout imiquimod.

Grant SupportThe study was supported by NIH R01 CA113851 and The Chemotherapy

Foundation (S. Demaria) and NCI K23 CA125205 (S. Adams). M.Z. Dewanwas supported byMolecular Oncology and Immunology TrainingGrant T32CA009161-33-34. NYU Cancer Institute Core facilities were supported byNIH 5P30CA016087-32.

The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

Received April 9, 2012; revised August 31, 2012; accepted September 22,2012; published OnlineFirst October 9, 2012.

References1. Lookingbill DP, Spangler N, Helm KF. Cutaneous metastases in

patients with metastatic carcinoma: a retrospective study of 4020patients. J Am Acad Dermatol 1993;29:228–36.

2. Krathen RA, Orengo IF, Rosen T. Cutaneous metastasis: a meta-analysis of data. South Med J 2003;96:164–7.

3. Cheever MA. Twelve immunotherapy drugs that could cure cancers.Immunol Rev 2008;222:357–68.

4. Adams S. Toll-like receptor agonists in cancer therapy. Immunother-apy 2009;1:949–64.

5. Geisse J, Caro I, Lindholm J, Golitz L, Stampone P, Owens M.Imiquimod 5% cream for the treatment of superficial basal cell carci-noma: results from two phase III, randomized, vehicle-controlledstudies. J Am Acad Dermatol 2004;50:722–33.

6. Panelli MC, Stashower ME, Slade HB, Smith K, Norwood C, Abati A,et al. Sequential gene profiling of basal cell carcinomas treated withimiquimod in a placebo-controlled study defines the requirements fortissue rejection. Genome Biol 2007;8:R8.

7. Gnjatic S, Sawhney NB, Bhardwaj N. Toll-like receptor agonists: arethey good adjuvants? Cancer J 2010;16:382–91.

8. Adams S, O'Neill DW, Nonaka D, Hardin E, Chiriboga L, Siu K, et al.Immunization of malignant melanoma patients with full-length NY-ESO-1 protein using TLR7 agonist imiquimod as vaccine adjuvant. JImmunol 2008;181:776–84.

9. Schon MP, Schon M. TLR7 and TLR8 as targets in cancer therapy.Oncogene 2008;27:190–9.

10. Sch€onM,BongAB,DrewniokC,Herz J,GeilenCC,Reifenberger J, et al.Tumor-selective induction of apoptosis and the small-molecule immuneresponse modifier imiquimod. J Natl Cancer Inst 2003;95:1138–49.

11. Smyth EC, Flavin M, Pulitzer MP, Gardner GJ, Costantino PD, Chi DS,et al. Treatment of locally recurrent mucosal melanoma with topicalimiquimod. J Clin Oncol 2011;29:e809–11.

12. Ellis LZ, Cohen JL, High W, Stewart L. Melanoma in situ treatedsuccessfully using imiquimod after nonclearance with surgery: reviewof the literature. Dermatol Surg 2012;38:937–46.

TLR7 Stimulation and Radiotherapy for Breast Cancer

www.aacrjournals.org Clin Cancer Res; 18(24) December 15, 2012 6677

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984

13. Narayan R, Nguyen H, Bentow JJ, Moy L, Lee DK, Greger S, et al.Immunomodulation by imiquimod in patients with high-risk primarymelanoma. J Invest Dermatol 2012;132:163–9.

14. Milas L,MasonKA, ArigaH,Hunter N, Neal R, ValdecanasD, et al. CpGoligodeoxynucleotide enhances tumor response to radiation. CancerRes 2004;64:5074–7.

15. Mason KA, Ariga H, Neal R, Valdecanas D, Hunter N, Krieg AM, et al.Targeting toll-like receptor 9 with CpG oligodeoxynucleotidesenhances tumor response to fractionated radiotherapy. Clin CancerRes 2005;11:361–9.

16. Brody JD, Ai WZ, Czerwinski DK, Torchia JA, LevyM, Advani RH, et al.In situ vaccination with a TLR9 agonist induces systemic lymphomaregression: a phase I/II study. J Clin Oncol 2010;28:4324–32.

17. Weigel BJ, Rodeberg DA, Krieg AM, Blazar BR. CpG oligodeoxynu-cleotides potentiate the antitumor effects of chemotherapy or tumorresection in an orthotopic murine model of rhabdomyosarcoma. ClinCancer Res 2003;9:3105–14.

18. Dumitru CD, Antonysamy MA, Tomai MA, Lipson KE. Potentiation ofthe anti-tumor effects of imidazoquinoline immune responsemodifiersby cyclophosphamide. Cancer Biol Ther 2010;10:155–65.

19. Machiels JP, Reilly RT, Emens LA, Ercolini AM, Lei RY, Weintraub D,et al. Cyclophosphamide, doxorubicin, and paclitaxel enhance theantitumor immune response of granulocyte/macrophage-colony stim-ulating factor-secreting whole-cell vaccines in HER-2/neu tolerizedmice. Cancer Res 2001;61:3689–97.

20. Wada S, Yoshimura K, Hipkiss EL, Harris TJ, Yen HR, Goldberg MV,et al. Cyclophosphamide augments antitumor immunity: studies inan autochthonous prostate cancer model. Cancer Res 2009;69:4309–18.

21. Emens LA, Armstrong D, Biedrzycki B, Davidson N, Davis-Sproul J,Fetting J, et al. A phase I vaccine safety and chemotherapy dose-finding trial of an allogeneic GM-CSF-secreting breast cancer vaccinegiven in a specifically timed sequence with immunomodulatory dosesof cyclophosphamide and doxorubicin. Hum Gene Ther 2004;15:313–37.

22. RosatoA,SantaSD, ZosoA,Giacomelli S,MilanG,MacinoB, et al. Thecytotoxic T-lymphocyte response against a poorly immunogenicmammary adenocarcinoma is focused on a single immunodominantclass I epitope derived from the gp70 Env product of an endogenousretrovirus. Cancer Res 2003;63:2158–63.

23. Waibler Z, Detje CN, Bell JC, Kalinke U. Matrix protein mediatedshutdown of host cell metabolism limits vesicular stomatitis virus-induced interferon-alpha responses to plasmacytoid dendritic cells.Immunobiology 2007;212:887–94.

24. Dewan MZ, Galloway AE, Kawashima N, Dewyngaert JK, Babb JS,Formenti SC, et al. Fractionated but not single dose radiotherapyinduces an immune-mediated abscopal effect when combined withanti-CTLA-4 antibody. Clin Cancer Res 2009;15:5379–88.

25. Demaria S, Kawashima N, Yang AM, Devitt M-L, Babb JS, Allison JP,et al. Immune-mediated inhibition of metastases following treatmentwith local radiation and CTLA-4 blockade in a mouse model of breastcancer. Clin Cancer Res 2005;11:728–34.

26. Matsumura S, Wang B, Kawashima N, Braunstein S, Badura M,Cameron TO, et al. Radiation-induced CXCL16 release by breastcancer cells attracts effector T cells. J Immunol 2008;181:3099–107.

27. Yu L, Chen S. Toll-like receptors expressed in tumor cells: targets fortherapy. Cancer Immunol Immunother 2008;57:1271–8.

28. Uehara A, Fujimoto Y, Fukase K, Takada H. Various human epithelialcells express functional Toll-like receptors, NOD1 and NOD2 to pro-duce anti-microbial peptides, but not proinflammatory cytokines. MolImmunol 2007;44:3100–11.

29. Drobits B, Holcmann M, Amberg N, Swiecki M, Grundtner R, HammerM, et al. Imiquimod clears tumors in mice independent of adaptiveimmunity by converting pDCs into tumor-killing effector cells. J ClinInvest 2012;122:575–85.

30. Lu H,Wagner WM, Gad E, Yang Y, Duan H, Amon LM, et al. Treatmentfailure of a TLR-7 agonist occurs due to self-regulation of acuteinflammation and can be overcome by IL-10 blockade. J Immunol2010;184:5360–7.

31. Ma Y, Kepp O, Ghiringhelli F, Apetoh L, Aymeric L, Locher C, et al.Chemotherapy and radiotherapy: cryptic anticancer vaccines. SeminImmunol 2010;22:113–24.

32. Lutsiak MEC, Semnani RT, De Pascalis R, Kashmiri SVS, Schlom J,Sabzevari H. Inhibition of CD4(þ)25 þT regulatory cell function impli-cated in enhanced immune response by low-dose cyclophosphamide.Blood 2005;105:2862–8.

33. Zhao J, Cao Y, Lei Z, Yang Z, Zhang B, Huang B. Selective depletion ofCD4þCD25þFoxp3 þregulatory T cells by low-dose cyclophospha-mide is explained by reduced intracellular ATP levels. Cancer Res2010;70:4850–8.

34. Ghiringhelli F, Larmonier N, Schmitt E, Parcellier A, Cathelin D,Garrido C, et al. CD4þCD25þ regulatory T cells suppress tumorimmunity but are sensitive to cyclophosphamide which allowsimmunotherapy of established tumors to be curative. Eur J Immunol2004;34:336–44.

35. Formenti SC, Demaria S. Systemic effects of local radiotherapy.Lancet Oncol 2009;10:718–26.

36. Schiavoni G, Sistigu A, Valentini M,Mattei F, Sestili P, Spadaro F, et al.Cyclophosphamide synergizes with type I interferons through system-ic dendritic cell reactivation and induction of immunogenic tumorapoptosis. Cancer Res 2011;71:768–78.

37. Sistigu A, Viaud S, Chaput N, Bracci L, Proietti E, Zitvogel L. Immu-nomodulatory effects of cyclophosphamide and implementations forvaccine design. Semin Immunopathol 2011;33:369–83.

38. Li VW, Li WW, Talcott KE, Zhai AW. Imiquimod as an antiangiogenicagent. J Drugs Dermatol 2005;4:708–17.

39. Huang SJ, Hijnen D, Murphy GF, Kupper TS, Calarese AW, Mollet IG,et al. Imiquimod enhances IFN-gamma production and effector func-tion of T cells infiltrating human squamous cell carcinomas of the skin.J Invest Dermatol 2009;129:2676–85.

40. Adams S, Kozhaya L, Martiniuk F, Meng TC, Chiriboga L, Liebes L,et al. Topical TLR7 agonist imiquimod can induce immune-mediatedrejection of skinmetastases in patientswith breast cancer. Clin CancerRes 2012;18:6748–57.

Dewan et al.

Clin Cancer Res; 18(24) December 15, 2012 Clinical Cancer Research6678

American Association for Cancer Research Copyright © 2012 on December 17, 2012clincancerres.aacrjournals.orgDownloaded from

Published OnlineFirst October 9, 2012; DOI:10.1158/1078-0432.CCR-12-0984


Recommended