smad7 promotes healing of radiotherapy- induced oral ......translational cancer mechanisms and...

12
Translational Cancer Mechanisms and Therapy Smad7 Promotes Healing of Radiotherapy- Induced Oral Mucositis without Compromising Oral Cancer Therapy in a Xenograft Mouse Model Jingjing Luo 1,2 , Li Bian 2,3,4 , Melanie A. Blevins 5 , Dongyan Wang 2,4 , Chao Liang 2 , Danfeng Du 2 , Fanglong Wu 1,2 , Barry Holwerda 6 , Rui Zhao 5 , David Raben 7 , Hongmei Zhou 1 , Christian D. Young 2,4 , and Xiao-Jing Wang 2,4 Abstract Purpose: We previously reported preventive and therapeu- tic effects of Smad7, a multifunctional protein, on radiother- apy (RT)-induced mucositis in mice without promoting human oral cancer cell survival or migration in vitro. The current study aims to determine whether a Smad7-based biologic can treat existing oral mucositis during radiotherapy for oral cancer and whether this treatment compromises RT-induced cancer cell killing in neighboring oral cancer. Experimental Design: We transplanted human oral cancer cells into the tongues of mice and applied craniofacial irradi- ation to simultaneously kill tumor cells and induce oral mucositis, thus modeling RT and mucositis in oral cancer patients. We topically applied a recombinant human Smad7 protein fused with the cell-penetrating Tat tag (Tat-Smad7) to the oral mucosa of tumor-bearing mice post RT when oral mucositis began to develop. Results: Topically applied Tat-Smad7 penetrated cells in both the oral mucosa and oral cancer, attenuating TGFb and NF-kB signaling as well as inammation at both sites. Tat-Smad7 treatment alleviated oral mucositis with reductions in DNA damage and apoptosis in keratinocytes, but increased keratinocyte proliferation compared with vehicle-treated mucositis lesions. In contrast, adjacent oral cancer exposed to Tat-Smad7 did not show alterations in proliferation or direct DNA damage, but showed increased oxidative stress damage and apoptosis compared with tumors treated with vehicle. Conclusions: Our results suggest that short-course Tat-Smad7 application to oral mucositis promotes its healing but does not compromise the cytotoxic effect of RT on oral cancer and has context-specic effects on oral mucosa versus oral cancer. Introduction Oral mucositis, painful ulceration of the oral mucosa, is a common toxic effect of radiotherapy (RT) for bone marrow transplant, craniofacial RT for head and neck cancer (HNC), and chemoradiotherapy for all cancer types (1). Approximately 70% of HNC patients develop oral mucositis during treatment, which can be severe enough to cause reduction in oral intake or prema- ture withdrawal from cancer treatment (1, 2). New RT technol- ogies such as intensity-modulated RT and stereotactic body RT (SBRT) more precisely target cancer lesions and spare more normal tissue. However, these treatments do not reduce acute toxicity in the oral mucosa of HNC patients (3), because neigh- boring mucosa is still exposed to high intensity RT, albeit at a lower dose than the cancer. Further, certain patients are at high risk for oral mucositis regardless of treatment regimen (4). Palifermin, recombinant human keratinocyte growth factor (KGF), is the only FDA-approved targeted therapy for preventing oral mucositis in bone marrow transplant patients (4% of the at-risk population), but it has no effect on existing mucositis (5). Palifermin clinical trials in oral cancer patients showed modest prevention of severe oral mucositis (6, 7). A major challenge in treating oral mucositis is to repair ulcerated mucosa without promoting cancer, as growth 1 State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, P.R. China. 2 Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado. 3 Department of Pathology, the First Afliated Hospital of Kunming Medical University, Kunming, P.R. China. 4 Allander Bio- technologies, LLC, Aurora, Colorado. 5 Department of Biochemistry and Molec- ular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Color- ado. 6 Mtibio, Las Vegas, Nevada. 7 Department of Radiation Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). J. Luo and L. Bian contributed equally to this article. Current address for C. Liang: School of Basic Medical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai, P.R. China; Current address for D. Du: Henan Key Laboratory for Esophageal Cancer Research and Department of Pathology of Basic Medical College, Zhengzhou University, Zhengzhou, Henan, P.R. China. Corresponding Authors: Xiao-Jing Wang, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Mailstop 8104, Aurora, CO 80045. Phone 303-724-3001; Fax: 303-724-4730; E-mail: [email protected]; Hongmei Zhou, State Key Laboratory of Oral Diseases, Department of Oral Medicine, West China Hospital of Stomatology, Sichuan University, No. 14, Sec. 3, Renmin Road South, Chengdu, Sichuan 610041, P.R. China. Phone: 86-028- 85503480; Fax: 86-028-85582167; E-mail: [email protected]; and Christian D. Young, Allander Biotechnologies, LLC, 12635 E. Montview Blvd #100, Aurora, CO 80045. Phone: 720-266-7063; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-18-1081 Ó2018 American Association for Cancer Research. Clinical Cancer Research Clin Cancer Res; 25(2) January 15, 2019 808 on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

Upload: others

Post on 29-Jan-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

  • Translational Cancer Mechanisms and Therapy

    Smad7 Promotes Healing of Radiotherapy-Induced Oral Mucositis without CompromisingOral Cancer Therapy in a Xenograft Mouse ModelJingjing Luo1,2, Li Bian2,3,4, Melanie A. Blevins5, Dongyan Wang2,4, Chao Liang2,DanfengDu2, FanglongWu1,2, Barry Holwerda6, Rui Zhao5, DavidRaben7, Hongmei Zhou1,Christian D. Young2,4, and Xiao-Jing Wang2,4

    Abstract

    Purpose: We previously reported preventive and therapeu-tic effects of Smad7, a multifunctional protein, on radiother-apy (RT)-induced mucositis in mice without promotinghuman oral cancer cell survival or migration in vitro. Thecurrent study aims to determine whether a Smad7-basedbiologic can treat existing oral mucositis during radiotherapyfor oral cancer and whether this treatment compromisesRT-induced cancer cell killing in neighboring oral cancer.

    Experimental Design: We transplanted human oral cancercells into the tongues of mice and applied craniofacial irradi-ation to simultaneously kill tumor cells and induce oralmucositis, thus modeling RT and mucositis in oral cancerpatients. We topically applied a recombinant human Smad7protein fused with the cell-penetrating Tat tag (Tat-Smad7) tothe oral mucosa of tumor-bearing mice post RT when oralmucositis began to develop.

    Results: Topically applied Tat-Smad7 penetrated cells inboth the oral mucosa and oral cancer, attenuatingTGFb and NF-kB signaling as well as inflammation at bothsites. Tat-Smad7 treatment alleviated oral mucositis withreductions in DNA damage and apoptosis in keratinocytes,but increased keratinocyte proliferation compared withvehicle-treated mucositis lesions. In contrast, adjacent oralcancer exposed to Tat-Smad7 did not show alterations inproliferation or direct DNA damage, but showed increasedoxidative stress damage and apoptosis compared with tumorstreated with vehicle.

    Conclusions: Our results suggest that short-courseTat-Smad7 application to oral mucositis promotes its healingbut does not compromise the cytotoxic effect of RT on oralcancer and has context-specific effects on oral mucosa versusoral cancer.

    IntroductionOral mucositis, painful ulceration of the oral mucosa, is a

    common toxic effect of radiotherapy (RT) for bone marrowtransplant, craniofacial RT for head and neck cancer (HNC), andchemoradiotherapy for all cancer types (1). Approximately 70%of HNC patients develop oral mucositis during treatment, whichcan be severe enough to cause reduction in oral intake or prema-ture withdrawal from cancer treatment (1, 2). New RT technol-ogies such as intensity-modulated RT and stereotactic body RT(SBRT) more precisely target cancer lesions and spare morenormal tissue. However, these treatments do not reduce acute

    toxicity in the oral mucosa of HNC patients (3), because neigh-boring mucosa is still exposed to high intensity RT, albeit at alower dose than the cancer. Further, certainpatients are at high riskfor oralmucositis regardless of treatment regimen (4). Palifermin,recombinant human keratinocyte growth factor (KGF), is the onlyFDA-approved targeted therapy for preventing oral mucositis inbone marrow transplant patients (4% of the at-risk population),but it has no effect on existing mucositis (5). Palifermin clinicaltrials in oral cancer patients showed modest prevention of severeoral mucositis (6, 7). A major challenge in treating oral mucositisis to repair ulceratedmucosawithout promoting cancer, as growth

    1State Key Laboratory of Oral Diseases, Department of Oral Medicine, WestChina Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, P.R.China. 2Department of Pathology, University of Colorado Anschutz MedicalCampus, Aurora, Colorado. 3Department of Pathology, the First AffiliatedHospital of Kunming Medical University, Kunming, P.R. China. 4Allander Bio-technologies, LLC, Aurora, Colorado. 5Department of Biochemistry and Molec-ular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Color-ado. 6Mtibio, LasVegas, Nevada. 7Department of RadiationOncology, Universityof Colorado Anschutz Medical Campus, Aurora, Colorado.

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

    J. Luo and L. Bian contributed equally to this article.

    Current address for C. Liang: School of Basic Medical Sciences, ShanghaiUniversity of Traditional ChineseMedicine, Shanghai, P.R. China; Current addressfor D. Du: Henan Key Laboratory for Esophageal Cancer Research and

    Department of Pathology of Basic Medical College, Zhengzhou University,Zhengzhou, Henan, P.R. China.

    Corresponding Authors: Xiao-Jing Wang, University of Colorado AnschutzMedical Campus, 12800 East 19th Avenue, Mailstop 8104, Aurora, CO 80045.Phone 303-724-3001; Fax: 303-724-4730; E-mail: [email protected];Hongmei Zhou, State Key Laboratory of Oral Diseases, Department of OralMedicine,West ChinaHospital of Stomatology, SichuanUniversity, No. 14, Sec. 3,Renmin Road South, Chengdu, Sichuan 610041, P.R. China. Phone: 86-028-85503480; Fax: 86-028-85582167; E-mail: [email protected]; and ChristianD. Young, Allander Biotechnologies, LLC, 12635 E. Montview Blvd #100, Aurora,CO 80045. Phone: 720-266-7063; E-mail:[email protected]

    doi: 10.1158/1078-0432.CCR-18-1081

    �2018 American Association for Cancer Research.

    ClinicalCancerResearch

    Clin Cancer Res; 25(2) January 15, 2019808

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://crossmark.crossref.org/dialog/?doi=10.1158/1078-0432.CCR-18-1081&domain=pdf&date_stamp=2018-12-27http://clincancerres.aacrjournals.org/

  • factors (e.g., KGF) and their receptors are often overexpressed incancer cells. To date, there is no FDA-approved drug to treat oralmucositis in cancer patients. Clinical trials for GC4419, a super-oxide dismutase mimetic, to treat oral mucositis in HNC patientsrevealed reductions in severe oral mucositis cases (8). GC4419requires i.v. infusions 1 hour prior to each of 14 doses of RT.Because it is difficult to predict which patients will develop severeoral mucositis, we sought to develop a therapeutic interventionthat is topically applied to existing oral mucositis and targetsmultiple pathogenic processes of oral mucositis.

    Oral mucositis develops as a consequence of complex molec-ular and cellular pathobiology processes, leading to apoptosis ofbasal epithelia cells, loss of epithelial renewal, atrophic damage,excessive inflammation, and ulceration (9). Our previous studydiscovered that in addition to NF-kB activation, activated TGFbsignaling contributes greatly to RT-induced oral mucositis (10).TGFb is a potent growth inhibitor and apoptosis inducer forepithelial cells and a proinflammatory cytokine in oral mucosa(11). To dampen both TGFb and NF-kB pathways to treat oralmucositis, we developed a recombinant Smad7 protein thatcontains human Smad7 fused to the HIV-1 Tat protein transduc-tion domain. Tat-Smad7 protein rapidly penetrates cells uponcontact (10). Local delivery of Tat-Smad7 to mouse oral mucosashows prophylactic and therapeutic effects on RT-induced oralmucositis (10). A remaining question is how to effectively utilizethis therapeutic intervention without compromising RT-directedcancer cell killing. Smad7 can be either tumor suppressive ortumor promotive in different cancer types (12). For cancersoutside the oral cavity, the concern for potential systemictumor-promoting effects of Tat-Smad7 is relatively minorbecause orally administered Tat-Smad7 protein will be degradedas it passes through thedigestive tract.However, whenRT-inducedoral mucositis in oral cancer patients is treated with Tat-Smad7,the oral cancer will also be exposed to Tat-Smad7. Althoughincreasing epithelial cell proliferation and reducing apoptosis inoral mucositis promote wound healing, these effects could com-promise RT-directed cancer cell death. We have previously shownthat Tat-Smad7 increases survival of humanoral keratinocytes butnot head and neck squamous cell carcinoma (HNSCC) cells afterRT. Similarly, Tat-Smad7 increases migration of normal kerati-nocytes but not HNSCC cells (10). These in vitro data indicate thenecessity of in vivo testing of Tat-Smad7 using a model systemmimicking RT in human oral cancer with an RT dose sufficient to

    induce oral mucositis. Although our previous study shows bothpreventive and therapeutic effect of Tat-Smad7 on oral mucositis(10), the current study focused on assessing the therapeuticeffect of Tat-Smad7 on promoting healing of existing oralmucositis in a model of oral cancer. We chose two HNSCCcell lines: FaDu, with a SMAD4 deletion representing alteredTGFb signaling found in tobacco-associated oral cancer (13),and UM-SCC-1, with wild-type SMAD4, representing intactTGFb signaling in tumor cells. We provide evidence that Tat-Smad7 application to RT-induced oral mucositis with neigh-boring oral cancer effectively promotes oral mucositis healingwhile paradoxically increasing tumor cell death in RT-treatedoral cancer.

    Materials and MethodsCell lines

    Two human oral cancer cell lines were used. FaDu was pur-chased from theATCC, andUM-SCC-1 (14)was provided byMTAthroughUniversity ofMichigan. Cells were authenticated by shorttandem repeat profiling (University of Colorado Cancer CenterProtein Production,Monoclonal Antibody, TissueCulture SharedResource) prior to our experiments. Cells were cultured in DMEMcontaining 10% FBS.

    Generation and application of Tat-Smad7As previously described, we produced recombinant Tat-

    tagged, human Smad7 protein as a GST fusion protein fromE. coli (10). Tat-Smad7 was cleaved from GST with PreScissionprotease followed by size exclusion chromatography to purifyTat-Smad7 protein as a monomer (Supplementary Fig. S1). Forin vivo treatment, 1 mg Tat-Smad7 (in 30 mL PBS containing 30%glycerol) was applied to mouse oral cavity; food/water waswithdrawn for 1 hour after treatment to minimize treatmentdisruption.

    Orthotopic human oral cancer xenotransplantationAnimal experiments were performed in accordance to an Insti-

    tutional Animal Care and Use Committee–approved protocol.We used female 8- to 10-week-old athymic nude mice (CharlesRiver Laboratories) as xenotransplantation recipients. Mice wereanesthetized with 80mg/kg ketamine and 12mg/kg xylazine (i.p.injection). Note that 105 oral cancer cells (FaDu or UM-SCC-1)were suspended in 20 mL of 50% PBS/50% Matrigel and injecteddirectly into the anterior/middle tongue using a syringe with30-gauge needle.

    Craniofacial irradiation to oral tumor–bearing miceMice with tongue tumors (7 days after injection of cancer

    cells) were cranially irradiated to induce oral mucositis using aRS2000 biological irradiator (Rad Source Technologies) aspreviously reported (10). Based on our previous observationthat a single 18 Gy dose has kinetics and severity of oralmucositis similar to 8 Gy x3 fractionated irradiation (10), wechose 18 Gy irradiation to reduce the death rate due to repeatedanesthesia for fractionated irradiation to oral tumor-bearingmice that had deteriorating health. Each mouse was anesthe-tized with 80 mg/kg ketamine and 12 mg/kg xylazine andplaced under a lead shield exposing only their head. The dayof irradiation was designated day 1. All animals were providedsoft food in addition to standard diet. On day 6, when micebegan to lose weight due to oral ulcer-associated reduced food

    Translational Relevance

    Oral mucositis, painful oral ulcerations, is one of the mostcommon toxic effects of radiotherapy (RT) and chemotherapyin cancer patients and can lead to therapy withdrawal or dosereduction. In head and neck cancer (HNC) patients, a majorchallenge in treating oral mucositis is to repair ulceratedmucosa without promoting neighboring cancer growth. Todate, there is no FDA-approved drug to treat oral mucositis inHNC patients. We studied a novel biological agent for treatingoral mucositis in a mouse model mimicking RT-inducedoral mucositis adjacent to oral cancer. Our preclinical datademonstrate the feasibility of a novel therapeutic approachfor treating existing oral mucositis without compromisingradiotherapy in neighboring cancer.

    Smad7 Promotes Oral Mucositis Healing without Aiding Cancer

    www.aacrjournals.org Clin Cancer Res; 25(2) January 15, 2019 809

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/

  • intake, mice were divided into two groups (of equal weight andtumor size) and treated daily with Tat-Smad7 or vehicle. BrdU(125 mg/kg) was administered i.p. 2 hours before euthanasia.Mice were sacrificed and tongue samples collected on day 10 forpathologic evaluation and immunostaining.

    Pathologic evaluation, immunostaining, and terminaldeoxynucleotidyl transferase dUTP nick end labeling assay

    Tongue tissues were fixed in 10% formalin, embedded inparaffin, and cut in 5 mm sections. Tongue epithelium and tumorhistology were evaluated using hematoxylin and eosin (H&E)–stained slides. Open ulcer size, defined as complete loss ofepithelium, was measured using NIS (Nikon Intensilight) Ele-ments software by two independent investigators, and the resultsaveraged to determine ulcer size (mm). We performed immuno-histochemical and immunofluorescent staining as previouslydescribed (10). The primary antibodies used were guineapig anti-Keratin14 (1:200, Fitzgerald, 20R-CP200), chickenanti-Keratin5 (1:200, Biolegend, #905901), FITC-labeled anti-body to BrdUþ (BD Bioscience, 347583), rat anti-mouse CD45(1:50, BD Bioscience, 550539), rabbit anti-mouse F4/80 (1:400,Cell Signaling Technology, 70076), rat anti-mouse Ly6G (1A8,1:3,200, Biolegend #127602), rabbit anti–NF-kB subunit p50(1:100, Santa Cruz Biotechnology, SC-7178), rabbit anti-pSmad2(1:200, Cell Signaling Technology, 3101), rabbit anti-pSmad3(1:400, Abcam, ab52903), rabbit anti-pH2AX (1:100, CellSignaling Technology, 9718), and mouse anti–8-OHdG (1:100,Alpha Diagnostic, 8OHG11-M). Smad7 antibody was producedusing human Smad7 recombinant protein to immunize rabbits.Specificity of Smad7 antiserum was confirmed by Western blot(Supplementary Fig. S2). Secondary antibodies conjugated toAlexa Flour 594 (red) or 488 (green) were used (1:200 for all,Invitrogen). Terminal deoxynucleotidyl transferase dUTP nickend labeling (TUNEL) staining was performed with a TUNEL kit(Promega) according to the manufacturer's directions to detectapoptotic cells. Slides were mounted with coverslips using Fluor-omount-G or DAPI Fluoromount-G (SouthernBiotech).

    Quantification of immunostainingIn oral mucosa, we quantified BrdUþ, pH2AX-, or 8-hydroxy-20-

    deoxyguanosine (8-OHdG)–positive cells as cells per mm base-ment membrane length (including all epithelial cells), TUNEL-positive cells as cells per mm basement membrane length (includ-ing all epithelial layers and stroma above themuscle layer), CD45-positve cells as DAPI-positive cells per mm2 epithelial and stromaarea above muscle layer, nuclear pSmad2- or NF-kB p50–positivecells as the percentage of positive cells per total epithelial cell count(excluding sloughed epithelial cells induced by irradiation).Sequential 20x images along the basement membrane were quan-tified and averaged per sample. In oral cancer regions, K14- or K5-positive cells were defined as tumor cells. All tumor cells weremeasured together as the area of tumor (mm2), excluding stromalcells. We quantified percentage of nuclear pSmad2- or NF-kBp50–positive cells/total tumor (epithelial and stromal) cells. Wequantified BrdUþ, pH2AXþ, 8-OHdGþ, CD45þ, and TUNELþ cellsas cells permm2 tumor area. Ly6Gþ or F4/80þ cellswere quantifiedas cells per mm2 tumor area based upon staining intensity andmorphology todistinguish them fromnonspecific stainingofothercell types. For pH2AX staining, cells with more than three nuclearfoci were defined as pH2AX-positive cells. Five random 20x tumorimages were quantified and averaged per sample.

    Statistical analysisStatistical analyses were performed using GraphPad Prism

    software. Normality was determined by D'Agostino and Pearsonnormality test (or Shapiro–Wilk normality test for smaller sam-ples size, n < 8). Every dataset passed these normality tests;differences between two treatment groups were determined usingthe Student t test, and differences between more than two groupswere determined by one-way ANOVA with Tukey multiple com-parison test. Data are presented as the mean � SEM.

    ResultsTat-Smad7 treatment reduced oral mucositis severity withoutprotecting adjacent oral cancer

    To mimic RT-induced oral mucositis in oral cancer patients,we used a human oral cancer orthotopic tongue xenograftmodel in athymic nude mice. Bone marrow transplant patientsare immune suppressed and experience severe RT-induced oralmucositis (15), suggesting that T lymphocytes are not majorcontributors to mucositis. We transplanted two human oralcancer lines, FaDu or UM-SCC-1, into the tongues of athymicnude mice. The health of tumor-bearing mice rapidly deterio-rated due to cancer burden and compromised oral intake.Therefore, after tumors were established, we irradiated micewith 18 Gy cranial RT instead of fractionated RT to minimizedeath due to repeated doses of anesthesia required forfractionated RT. This irradiation dose induces oral mucositiswith the kinetics and severity similar to 3 � 8 Gy fractionatedRT in mice with obvious mucosal damage starting on day 5 afterRT (10). Six days after RT, when mice began to lose weight (10),we treated them with Tat-Smad7 produced as we previouslydescribed (ref. 10; Supplementary Fig. S1). We chose the doseand regimen of topical Tat-Smad7 application that effectivelytreated oral mucositis (1 mg/30 mL oral dose, daily). Vehicletreatment (30% glycerol in PBS) was used as control. Mice weresacrificed and tongues harvested 10 days after RT. We assessed ifTat-Smad7 protein was delivered to both oral mucosa and oralcancer with immunofluorescent staining using a human Smad7antibody that also cross-reacts with mouse Smad7 (Supple-mentary Fig. S2). Similar to our previous report (10), in the oralepithelium of Tat-Smad7–treated mice, Smad7 was present inboth the cytoplasm and the nucleus, whereas endogenousSmad7 levels in vehicle-treated mice were low (Fig. 1A). Intumors, Tat-Smad7 was detected in the cytoplasm of bothtumor epithelia and stroma (Fig. 1A), suggesting that TGFb inthese cells drives Smad7 cytoplasmic translocation to blockTGFb signaling (16). We measured tumor area microscopicallyand found that both FaDu and UM-SCC-1 tumor sizes werereduced in RT-treated mice and Tat-Smad7 treatment did notsignificantly affect tumor size compared with vehicle in thisshort-course treatment (Fig. 1B and C). Tongue mucosaadjacent to tumor showed RT damage and mucositis formationprimarily at the posterior dorsal surface which has fewercornified layers compared with tongue papillae at the tip ofthe tongue (Fig. 1F and G). Tat-Smad7–treated mice hadsignificantly smaller RT-induced tongue ulcers compared withvehicle-treated mice (Fig. 1D and E).

    Histopathologic analysis revealed that irradiated epitheliumexhibited atrophy, flattened tongue papillae, thinning cuticle, andulceration compared with untreated epithelium (Fig. 1F and G).Irradiated mucosa treated with Tat-Smad7 showed less epithelial

    Luo et al.

    Clin Cancer Res; 25(2) January 15, 2019 Clinical Cancer Research810

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/

  • atrophy, smaller ulcerated area, and fewer immune cells com-pared with vehicle-treated mucosa (Fig. 1F and G; ref. 10). Com-pared with nonirradiated tumors, irradiated tumors near muco-sitis showed obvious cell damage with vacuolar degeneration andpyknotic nuclei or dysplastic cell shape with enlarged, smudgedchromatin. Apoptotic cells, inflammation, and interstitial fibrosiswere obvious in irradiated tumors. FaDu tumors were moresensitive to irradiation as demonstrated by more damaged tumorcells than UM-SCC-1 tumors (Fig. 1F and G).

    Tat-Smad7 reduced NF-kB and TGFb signaling in both oralmucositis and oral cancer

    To examine on-target effects of Tat-Smad7 on oral mucosaand cancer, we performed immunostaining for pSmad2and pSmad3, markers of TGFb pathway activation, andnuclear NF-kB p50, a marker of NF-kB pathway activation.

    Oral mucosa after RT had more nuclear pSmad3þ cells com-pared with nonirradiated mucosa (Supplementary Fig. S3),similar to changes in pSmad2þ cells as previously reported(10). Fewer nuclear pSmad2þ and pSmad3þ cells were observedin irradiated oral mucosa and adjacent oral cancer in Tat-Smad7–treated mice compared with vehicle treatment (Fig. 2A, B, E, F, I,and J; Supplementary Fig. S3). In nonirradiated tongue tumors,both UM-SCC-1 and FaDu had more pSmad3þ cells than oralmucosa (Supplementary Fig. S3), indicating TGFb-dependentSmad3 activation (even in Smad4-deficient FaDu tumors) aspreviously reported (13). In addition, fewer nuclear NF-kBp50þ cells were observed in both irradiated oral mucosa andadjacent oral cancer in Tat-Smad7–treated mice compared withvehicle treatment (Fig. 2C, D, G, H, K, and L). These datademonstrate the on-target efficacy of Tat-Smad7 against knownSmad7 targets in tongue mucosa with oral tumors.

    Figure 1.

    Oral Tat-Smad7 treatment alleviated RT-induced oral mucositis but did not affect radiotherapy on neighboring oral cancer. A, Representativeimmunofluorescent images using Smad7 antibody to detect endogenous Smad7 or Tat-Smad7 in irradiated mouse tongue mucosa and neighboringxenografted FaDu tumor (no endogenous Smad7). Weak endogenous Smad7 was detected in mouse tongue mucosa and oral cancer stromal cells of mouse origin.K14 antibody was used to counterstain epithelial cells. B and C, Quantification of oral tumor size (mean � SEM) 10 days after 18 Gy RT in mice untreated(no RT), treated with RT and vehicle (RT þ Vehicle) or RT and Tat-Smad7 (RTþTat-Smad7). D and E, Quantification of oral ulcer diameter (mean � SEM)10 days after 18 Gy RT in mice treated as described in B and C. P values determined by the Student t test (two groups) or one-way ANOVA with Tukeymultiple comparison test (more than two groups). F and G, Representative H&E images of oral mucosa and oral cancer in tongue samples harvested from micebearing FaDu (F) or UM-SCC-1 (G) tumors 10 days after 18 Gy RT. Top plots present low power images of tongue epithelium and tumor. Yellow solid linesdefine the ulcer boundary. Black dotted lines delineate the boundary of tongue tumor. Scale bars, 200 mm. Mid plots present high power images of oralepithelium from nonirradiated posterior dorsal tongue mucosa and irradiated mucosa from the same region with ulcer. White dotted lines delineate epithelial cellsmigrated underneath the ulcer. Scale bars, 100 mm. Bottomplots present high power images of the oral cancer. Yellow arrows point to dying or apoptotic tumor cells.White arrows point to areas without obvious RT-induced damage. Scale bars, 100 mm.

    Smad7 Promotes Oral Mucositis Healing without Aiding Cancer

    www.aacrjournals.org Clin Cancer Res; 25(2) January 15, 2019 811

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/

  • Tat-Smad7 treatment alleviated inflammation and DNAdamage–associated cell death in oral mucositis lesions of oralcancer–bearing mice

    We quantified leukocytes with CD45 staining and foundTat-Smad7–treated mucositis contained fewer CD45þ leukocytes(Fig. 3A and B). We next performed F4/80 and Ly6G staining todetermine relative levels of F4/80þmacrophages and Ly6Gþ cells[a marker of polymorphonuclear (PMN) neutrophils or PMN

    myeloid-derived suppressor cells (MDSC)]. RT-induced oralmucositis harbors primarily neutrophils and macrophages (9)that were obvious in oralmucositis lesions of tumor-bearingmice(Fig. 3A). Tat-Smad7–treated mucositis had fewer PMNs andmacrophages than vehicle-treated lesions (Fig. 3A), consistentwith smaller ulcers and fewer inflammatory cells observedbyH&Eand CD45 staining. To examine epithelial slough due to DNAdamage–induced cell death and cessation of proliferation, we

    Figure 2.

    Oral Tat-Smad7 treatment attenuated nuclear pSmad2 and NF-kB p50 in RT-induced oral mucositis and adjacent cancer. A–D, Representativeimmunofluorescent staining of pSmad2 (A, B) and NF-kB subunit p50 (C, D) in RTþVehicle or RTþTat-Smad7–treated oral mucosa and oral cancer (FaDu:A, C; UM-SCC-1: B, D) with K14 or K5 epithelial cell counterstain. Scale bars, 100 mm for all sections. E–L, Quantifications of nuclear staining for pSmad2 or NF-kBp50 (mean� SEM). Nuclear pSmad2 andNF-kB p50–positive cells were quantified as a percentage of total epithelial cells in oral mucosa (FaDu: E,G; UM-SCC-1: I,K)or in tumor cells (FaDu: F, H; UM-SCC-1: J, L). P values determined by the Student t test.

    Luo et al.

    Clin Cancer Res; 25(2) January 15, 2019 Clinical Cancer Research812

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/

  • first performed pH2AX staining as a marker of DNA-damagedcells. Irradiated oral mucosa had numerous pH2AXþ cells, andTat-Smad7–treated oral mucosa had fewer pH2AXþ cells com-pared with vehicle-treated mucosa (Fig. 3A and C). Next, stainingof nuclear 8-OHdG was performed as a marker of oxidativeDNA damage (17). There were fewer 8-OHdGþ cells in oralmucosa of Tat-Smad7–treated mice compared with vehicle-trea-tedmice (Fig. 3A andD).We performed TUNEL assays to quantifyapoptosis and noted numerous apoptotic cells in the oral mucosaof irradiated mice. Tat-Smad7–treated mucosa contained fewerapoptotic cells than vehicle control (Fig. 3A and E). Together,these data demonstrate that Tat-Smad7 treatment reduces inflam-mation, DNA damage, and apoptosis associated with RT-inducedoral mucositis in tumor-bearing mice.

    Tat-Smad7 reduced inflammation in irradiated oral cancerUnlike nonirradiated oral mucosa that had no obvious inflam-

    mation (Fig. 1), leukocyte infiltration was apparent in nonirra-diated tumors that contained numerous F4/80þ macrophages

    and Ly6Gþ cells (Supplementary Fig. S4). Very few infiltrated Bcells were detected (data not shown). In irradiated FaDu tumors,F4/80þ cells were still prominent, but Ly6Gþ cells were largelydiminished (Fig. 4A andCvs. Supplementary Fig. S4) even thoughLy6Gþ cells were numerous in adjacent mucositis (Fig. 3). BothF4/80þ cells and Ly6Gþ cells were still pronounced in UM-SCC-1–irradiated tumors (Fig. 4D, F, and G). Intriguingly, Tat-Smad7reduced the number of CD45þ leukocytes and F4/80þ macro-phages in both types of irradiated oral tumors and reduced Ly6Gþ

    cells in irradiated UM-SCC-1 tumors (Fig. 4).

    Effects of Tat-Smad7 on irradiated oral cancer diverge from itseffects on oral mucositis

    Similar to oral mucositis, irradiated tumors had numerouspH2AXþ cells that were significantly more abundant than innonirradiated tumors (Fig. 5A; Supplementary Fig. S5). FaDutumors harbored more pH2AXþ cells than UM-SCC-1 tumors(Fig. 5D and E; Supplementary Fig. S5). However, unlike oralmucositis, there was no difference in the number of pH2AXþ cells

    Figure 3.

    Oral Tat-Smad7 treatment mitigated inflammation, DNA damage, and apoptosis in RT-induced oral mucositis. A, Representative immunostaining of markersfor immune cells (CD45, Ly6G, F4/80), DNA damage markers (pH2AX, 8-OHdG), and apoptosis (TUNEL) in RTþVehicle or RTþTat-Smad7–treated oralmucosa adjacent to irradiated FaDu tongue tumors. Similar staining patterns were also seen in irradiated mucosa adjacent to UM-SCC-1 tumors (not shown)and quantified in B to E. K5 or K14 antibody was used to counterstain epithelial cells. DAPI was used to counterstain nuclei for CD45 staining, and propidiumiodide (PI) was used to counterstain nuclei for TUNEL. Scale bars, 100 mm for all sections. B–E, Quantification (mean � SEM) of immunostaining markers inoral mucosa adjacent to irradiated FaDu and UM-SCC-1 shown in A. CD45þ cells (B) were quantified based on mm2 epithelial and stromal areas above themuscle layer. Quantifications for pH2AXþ cells (C), 8-OHdGþ cells (D), and TUNELþ cells (E) in keratinocytes per mm basement membrane length. P valueswere determined by the Student t test.

    Smad7 Promotes Oral Mucositis Healing without Aiding Cancer

    www.aacrjournals.org Clin Cancer Res; 25(2) January 15, 2019 813

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/

  • in oral cancers treated with Tat-Smad7 versus vehicle in bothtumor types (Fig. 5D and E). 8-OHdGþ cells were hardly detect-able in nonirradiated tumors (Supplementary Fig. S5) but wereinduced significantly by RT (Fig. 5B). In contrast to Tat-Smad7effects on oral mucositis, 8-OHdGþ cells in oral cancer weresignificantly increased in Tat-Smad7–treatedmice compared withvehicle (Fig. 5F and G). Basal levels of TUNELþ apoptotic cellswere present in nonirradiated tumors (Supplementary Fig. S5)and RT induced numerous apoptotic cells in both tumor types(Fig. 5C). Apoptotic cells in oral cancer were increased inTat-Smad7–treated mice compared with vehicle-treated(Fig. 5H and I), opposite of what was observed in Tat-Smad7–treated mucositis. We performed BrdU staining to quantitateproliferating cells. Adjacent to ulceration, Tat-Smad7–treated oralmucosa showed more proliferative BrdUþ cells along the epithe-lial basement membrane than vehicle-treated oral mucosa (Fig.6A–D). In contrast, Tat-Smad7–treated oral cancer had no differ-ence in proliferation compared with vehicle-treated oral cancer(Fig. 6A, B, E, and F). In summary, Tat-Smad7–treated mucositis

    lesions had reduced RT-induced DNA damage, inflammation,and apoptosis and increased keratinocyte proliferation, whereasadjacent irradiated tumors treated with Tat-Smad7 demonstratedno resolution to DNA damage, decreased inflammation, andunaltered proliferation.

    DiscussionOneof themajor challenges for treating oralmucositis in cancer

    patients is that potential therapies reviving normal mucosalregeneration could also pose risk by protecting cancer cells. Weshow here that Tat-Smad7 topical application reduced activationof NF-kB and TGFb signaling and reduced inflammation in bothoralmucosa and cancer, but differentially affectednormalmucosaand oral cancer with respect to DNA damage and associated celldeath, and cell proliferation.

    Our current study confirms that in oral mucositis-inducedacute inflammation associated with NF-kB and TGFb activation(10, 18), macrophages and neutrophils are predominant

    Figure 4.

    Oral Tat-Smad7 treatment reduced leukocyte infiltrate in irradiated tongue tumors. A and D, Representative images of CD45, F4/80 and Ly6G staining inFaDu tumors (A) and UM-SCC-1 tumors (D). K5 antibody was used to counterstain epithelial cells, and DAPI was used to counterstain nuclei for CD45 staining.Note that tumor cells after irradiation had more nonspecific staining in F4/80 than in nonirradiated tumors (Supplementary Fig. S4). Quantification of F4/80þ

    macrophages in C and F is based upon staining intensity and morphology (indicated by yellow arrows, A and D). Black arrows in Ly6G vehicle panel inA point to a few remaining Ly6Gþ cells which were absent in Tat-Smad7–treated tumor. Scale bars, 100 mm for all sections. B and C, Quantifications of CD45þ

    and F4/80þ cells in FaDu tumors. E–G,Quantifications of CD45þ, F4/80þ, and Ly6Gþ cells in UM-SCC-1 tumors. Immune cellswere quantified (mean� SEM) permm2tumor area. P values were determined by the Student t test.

    Luo et al.

    Clin Cancer Res; 25(2) January 15, 2019 Clinical Cancer Research814

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/

  • leukocyte types (9). Fewer macrophages and neutrophils inTat-Smad7–treated mucositis could reflect less damage (smallerulcers and fewer dead cells) or a quicker resolution of inflamma-tion. Oral cancer without RT showed chronic inflammation in thetumor microenvironment that harbored numerous F4/80þ puta-tive M2 macrophages and Ly6Gþ putative tumor-associated neu-trophils or PMN-MDSCs; both are known to be tumor-promotingleukocytes attracted by TGFb1 and also a major source ofTGFb1 (19). With RT, F4/80þ and Ly6Gþ cells remained plentifulin UM-SCC-1 tumors, and Tat-Smad7 reduced numbers of thesecells similar to TGFb inhibitors and NF-kB inhibitors used inclinical trials of metastatic cancer, which has an inflammatorytumor microenvironment (20, 21). The subtypes of theseleukocytes in irradiated tumors require further study, e.g., if theyhave shifted from PMN-MDSCs to neutrophils as part of acute

    inflammation induced by RT. Interestingly, Ly6Gþ leukocyteswere largely diminished in irradiated FaDu tumors despitetheir presence in the adjacent mucosa. Because FaDu cells lackSmad4 but retain nuclear pSmad2þ and pSmad3þ, it is unclearif this altered TGFb signaling affected RT response–associatedchanges in Ly6Gþ cell infiltration or depletion in tumors. Incontrast, Tat-Smad7 still reduced F4/80þ macrophages in FaDutumors, possibly by blocking RT-induced TGFb signalingthrough Smad2/Smad3 and by direct blocking of non–Smad-mediated NF-kB activation by TGFb (22). These datahighlight the complex biology and heterogeneity of tumors inresponse to RT. The anti-TGFb/NF-kB effects of Smad7 couldhave a cascade effect on cytokine production directly or indi-rectly due to reduced leukocyte numbers. Therefore, futurein-depth studies are needed to dissect subtypes of immune

    Figure 5.

    Tat-Smad7 did not reduce RT-induced DNA damage but increased oxidative stress and apoptosis in irradiated tongue cancers. A–C, Representativestaining of pH2AX, 8-OHdG, and TUNEL in RT þ vehicle and RT þ Tat-Smad7–treated tongue cancer. K14 antibody was used to counterstain epithelialcells or propidium iodide (PI) to counterstain nuclei. Scale bars, 100 mm for all sections. D–I, Quantification (mean � SEM) of pH2AXþ, 8-OHdGþ, and TUNELþtumor epithelial cells in cancer per mm2 tumor epithelial area (FaDu: D, F, H; UM-SCC 1: E, G, I). P values were determined by the Student t test.

    Smad7 Promotes Oral Mucositis Healing without Aiding Cancer

    www.aacrjournals.org Clin Cancer Res; 25(2) January 15, 2019 815

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/

  • cells influenced by Smad7 as well as the production of cyto-kines by these cells.

    Because reducing inflammation is insufficient to alleviate oralmucositis, other pathogenic processes must also be targeted foreffective treatment. Among these processes, RT-induced DNAdamage kills tumor cells but also initiates oral mucositis(1, 23). Therefore, reducing DNA damage or accelerating repairis key for oral mucositis healing, but such an effect is undesirablefor eradicating tumor cells. The selective reduction of RT-inducedDNA damage by Smad7 in oral mucosa (Fig. 3) might beexplained by its ability to facilitate DNA damage repair throughprotein–protein interactions with the DNA repair proteincomplex containing ATM and Mre11-Rad50-Nbs1 proteins(24). However, Tat-Smad7 did not reduce RT-induced DNAdamage in tumor cells (pH2AXþ cells). This may be part of thereason that intrinsic DNA repair defects in tumor cells (evidentin nonirradiated tumors) cannot be reversed by Tat-Smad7. Inaddition, TGFb-mediated DNA repair is an important mech-

    anism of radioresistance in cancer treatment (25–29). In thiscontext, blocking TGFb signaling by Tat-Smad7 wouldenhance DNA damage in cancer cells. Further, RT-inducedoxidative stress is required to amplify DNA damage, causingcollateral damage leading to cancer cell death (30, 31) and oralmucositis (1, 23). Reduced oxidative stress marker 8-OHdGwith Tat-Smad7 treatment in oral mucositis is consistentwith previous reports that both TGFb and NF-kB pathwayscan induce oxidative stress (32–35). Paradoxically, TGFb isreported to reduce oxidative stress in a skin SCC model (36).This could explain why Tat-Smad7 treatment increased8-OHdGþ cells in oral cancer. Given these effects, it is notsurprising that DNA damage–associated apoptosis wasreduced by Tat-Smad7 in oral mucositis but increased inadjacent oral cancer. The effect of Tat-Smad7 on apoptosis isalso consistent with previous reports showing Smad7 reducedapoptosis in normal epithelia (10, 37) but enhanced apoptosisin cancer (38).

    Figure 6.

    Tat-Smad7 increased proliferation in irradiated oral mucosa but not neighboring oral cancer and summary of Tat-Smad7 actions.A andB,Representative staining ofBrdU-positive cells in irradiated mucosa and neighboring oral cancer (FaDu: A, UM-SCC-1: B). K14 antibody was used as counterstain epithelial cells. Scale bars,100 mm. C–F, Quantification (mean � SEM) of BrdUþ cells in oral mucosal keratinocytes per mm basement membrane length (adjacent to FaDu: C; adjacent toUM-SCC-1: D) and in cancer epithelial cells per mm2 tumor area (FaDu: E; UM-SCC-1: F). P values were determined by the Student t test. G, Smad7 functionalmechanisms in oralmucositis with neighboring irradiated oral cancer. RT directly activates TGFb andNF-kB, and induces DNAdamage. Smad7 alleviates RT-inducedDNAdamage (includingDNA strand breaks and oxidative damage), attenuates TGFb-induced growth arrest, attenuates apoptosis, and blocks inflammation inducedby TGFb and NF-kB, further promoting oral mucositis healing. H, Smad7 functional mechanisms in irradiated oral cancer. RT directly activates TGFb andNF-kB. Tumor cells harbor intrinsically damaged DNA and are thus more sensitive to RT-induced apoptosis. Smad7 enhances RT-induced oxidative damage thatconsequently increases apoptosis. Smad7 also attenuates TGFb and NF-kB activation–induced inflammation.

    Luo et al.

    Clin Cancer Res; 25(2) January 15, 2019 Clinical Cancer Research816

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/

  • In a subset of HNSCCs, endogenous Smad7 is activated byTGFb and NF-kB, which transcriptionally suppress Smad targetgenes that could either be tumor suppressive or promotive(22). Therefore, pharmacologic dosage of a Smad7-based bio-logic needs to be carefully assessed. Our study revealed thatHNSCCs derived from SMAD4 mutant (FaDu) and SMAD4wild-type (UM-SCC-1) cells responded to Tat-Smad7 similarly.This can be explained by the fact that the majority of humancancers escape Smad-dependent TGFb growth inhibition,which is also supported by Tat-Smad7 not affecting cancercell proliferation in our model. In addition, we have shownSMAD4-mutant epithelial cells rely primarily on Smad3-depen-dent signaling to mediate TGFb1-induced inflammation (13).Therefore, the use of Tat-Smad7 in SMAD4-mutant cancercould follow the same principle as TGFb inhibitor in treatingSMAD4-mutant metastatic cancer including pancreatic cancerthat has a high rate of SMAD4 mutation (39).

    In summary, we provide evidence that local short-termTat-Smad7 protein delivery alleviated RT-induced oral muco-sitis without compromising RT-induced killing of neighboringoral cancer. Potential mechanisms for the context-specificeffects of Smad7 are as follows: In oral mucositis, Tat-Smad7attenuates TGFb-mediated growth arrest and apoptosis as wellas TGFb/NF-kB–mediated inflammation. Smad7 could alsodirectly reduce DNA damage or accelerate repair (Fig. 6G). Inneighboring oral cancer, Smad7 still attenuates TGFb/NF-kB–mediated inflammation, but is insufficient to reduce DNAdamage (Fig. 6H). In addition, Smad7 could block TGFb-mediated suppression of oxidative damage and consequentlyinduce apoptosis in tumor cells (Fig. 6H). Because high inten-sity/hypofractionated SBRT is increasingly used to treat oralcancers resistant to traditional low-dose fractionated RT, futurestudies that more closely mimic SBRT will be needed to assesstherapeutic efficacy of Tat-Smad7 in oral mucositis treatment.In addition, HNSCC mouse models with an intact immunesystem (13, 40) will further determine if effects of Tat-Smad7are influenced by T-cell–mediated tumor immunity. Futurestudies should carefully assess Tat-Smad7 dose-dependentefficacy to effectively treat oral mucositis but avoid potentialoncogenic effects on neighboring oral cancer, and assess the

    effect of long-term Tat-Smad7 use on oral cancer progressionwith an intact immune microenvironment.

    Disclosure of Potential Conflicts of InterestD.Raben is a consultant/advisory boardmember for EMDSerono. X.-J.Wang

    is an employee of Veterans Affairs, reports receiving commercial research grantsfrom NIH to Allander, and holds ownership interest (including patents) inAllander Biotechnologies. No potential conflicts of interest were disclosed bythe other authors.

    Authors' ContributionsConception and design: J. Luo, D. Raben, X.-J. WangDevelopment of methodology: J. Luo, L. Bian, C. Liang, D. Du, H. Zhou,C.D. Young, X.-J. WangAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): J. Luo, L. Bian, M.A. Blevins, D. Wang, C. Liang, D.Du, F. Wu, B. Holwerda, R. ZhaoAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): J. Luo, L. Bian, C. Liang, D. Du, F. Wu, H. Zhou,C.D. Young, X.-J. WangWriting, review, and/or revision of the manuscript: J. Luo, L. Bian,M.A. Blevins, D. Raben, H. Zhou, C.D. Young, X.-J. WangAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): F. WuStudy supervision: R. Zhao, H. Zhou, X.-J. Wang

    AcknowledgmentsWe thankDrs. Yosef Refaeli, Brian Turner, Steve Sonis, and Thomas Carey for

    advice and an anonymous donation for support of this work. D. Raben issupported by the Marsico Endowment fund for research.

    This work was supported by NIH grant DE024659 to C.D. Young andX.-J. Wang, and DE015953 to X.-J. Wang. X.-J. Wang is also a Research Biologistin Department of Veterans Affairs. J. Luo and F. Wu were supported by theNational Nature Science Foundation of China No. 81772898, and theState Scholarship Fund of China Scholarship Council No. 201506240122 andNo. 201606240201. M.A. Blevins was supported by T32 CA174648.

    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 7, 2018; revised August 2, 2018; accepted August 31, 2018;published first September 5, 2018.

    References1. Sonis ST. Oral mucositis in head and neck cancer: risk, biology, and

    management. Am Soc Clin Oncol Educ Book 2013;2013:236–40.2. Villa A, Sonis ST. Mucositis: pathobiology and management. Curr Opin

    Oncol 2015;27:159–64.3. Khuntia D, Harris J, Bentzen SM, Kies MS, Meyers JN, Foote RL, et al.

    Increased oralmucositis after IMRT versus non-IMRTwhen combinedwithcetuximab and cisplatin or docetaxel for head and neck cancer: preliminaryresults of RTOG 0234. Int J Radiat Oncol 2008;72.

    4. Al-Dasooqi N, Sonis ST, Bowen JM, Bateman E, Blijlevens N, Gibson RJ,et al. Emerging evidence on the pathobiology of mucositis. Support CareCancer 2013;21:2075–83.

    5. Sonis ST. Efficacy of palifermin (keratinocyte growth factor-1) in theamelioration of oral mucositis. Core Evid 2010;4:199–205.

    6. Henke M, Alfonsi M, Foa P, Giralt J, Bardet E, Cerezo L, et al. Palifermindecreases severe oralmucositis of patients undergoing postoperative radio-chemotherapy for head andneck cancer: a randomized, placebo-controlledtrial. J Clin Oncol 2011;29:2815–20.

    7. Le QT, KimHE, Schneider CJ, Murakozy G, Skladowski K, Reinisch S, et al.Palifermin reduces severe mucositis in definitive chemoradiotherapy oflocally advanced head and neck cancer: a randomized, placebo-controlledstudy. J Clin Oncol 2011;29:2808–14.

    8. Anderson CM, Sonis ST, Lee CM, Adkins D, Allen BG, Sun W, et al. Phase1b/2a trial of the superoxide dismutase mimetic GC4419 to reducechemoradiotherapy-induced oral mucositis in patients with oral cavity ororopharyngeal carcinoma. Int J Radiat Oncol Biol Phys 2018;100:427–35.

    9. Sonis ST. The pathobiology of mucositis. Nat Rev Cancer 2004;4:277–84.

    10. HanG,Bian L, Li F, CotrimA,WangD, Lu J, et al. Preventive and therapeuticeffects of Smad7 on radiation-induced oral mucositis. Nat Med2013;19:421–8.

    11. Lu SL, Reh D, Li AG, Woods J, Corless CL, Kulesz-Martin M, et al. Over-expression of transforming growth factor beta1 in head and neck epitheliaresults in inflammation, angiogenesis, and epithelial hyperproliferation.Cancer Res 2004;64:4405–10.

    12. Bian L, Han G, Zhao CW, Garl PJ, Wang XJ. The role of Smad7 in oralmucositis. Protein Cell 2015;6:160–9.

    13. Bornstein S, White R, Malkoski S, Oka M, Han G, Cleaver T, et al. Smad4loss in mice causes spontaneous head and neck cancer with increasedgenomic instability and inflammation. J Clin Invest 2009;119:3408–19.

    14. Brenner JC, GrahamMP, Kumar B, Saunders LM, Kupfer R, Lyons RH, et al.Genotyping of 73 UM-SCC head and neck squamous cell carcinoma celllines. Head Neck 2010;32:417–26.

    Smad7 Promotes Oral Mucositis Healing without Aiding Cancer

    www.aacrjournals.org Clin Cancer Res; 25(2) January 15, 2019 817

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/

  • 15. Gabriel DA, Shea T, Olajida O, Serody JS, Comeau T. The effect of oralmucositis on morbidity and mortality in bone marrow transplant. SeminOncol 2003;30 (6 Suppl 18):76–83.

    16. Itoh S, Landstrom M, Hermansson A, Itoh F, Heldin CH, Heldin NE, et al.Transforming growth factor beta1 induces nuclear export of inhibitorySmad7. J Biol Chem 1998;273:29195–201.

    17. Valavanidis A, Vlachogianni T, Fiotakis C. 8-hydroxy-2'-deoxyguano-sine (8-OHdG): A critical biomarker of oxidative stress and carcino-genesis. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev2009;27:120–39.

    18. Sonis ST. Pathobiology of oralmucositis: novel insights and opportunities.J Support Oncol 2007;5 (9 Suppl 4):3–11.

    19. Lan Y, ZhangD, XuC,Hance KW,Marelli B, Qi J, et al. Enhanced preclinicalantitumor activity ofM7824, a bifunctional fusion protein simultaneouslytargeting PD-L1 and TGF-beta. Sci Translat Med 2018;10.

    20. Yingling JM, Blanchard KL, Sawyer JS. Development of TGF-betasignalling inhibitors for cancer therapy. Nat Rev Drug Discov 2004;3:1011–22.

    21. Yamamoto Y, Gaynor RB. Therapeutic potential of inhibition of the NF-kappaB pathway in the treatment of inflammation and cancer. J Clin Invest2001;107:135–42.

    22. Freudlsperger C, Bian Y, Contag Wise S, Burnett J, Coupar J, Yang X, et al.TGF-beta and NF-kappaB signal pathway cross-talk is mediated throughTAK1 and SMAD7 in a subset of head and neck cancers. Oncogene2013;32:1549–59.

    23. Treister N, Sonis S. Mucositis: biology and management. Curr OpinOtolaryngol Head Neck Surg 2007;15:123–9.

    24. Park S, Kang JM, Kim SJ, KimH,Hong S, Lee YJ, et al. Smad7 enhances ATMactivity by facilitating the interaction between ATM and Mre11-Rad50-Nbs1 complex in DNA double-strand break repair. Cell Mol Life Sci2015;72:583–96.

    25. Kirshner J, Jobling MF, Pajares MJ, Ravani SA, Glick AB, Lavin MJ, et al.Inhibition of transforming growth factor-beta1 signaling attenuates ataxiatelangiectasia mutated activity in response to genotoxic stress. Cancer Res2006;66:10861–9.

    26. Ewan KB, Henshall-Powell RL, Ravani SA, PajaresMJ, Arteaga C,Warters R,et al. Transforming growth factor-beta1mediates cellular response to DNAdamage in situ. Cancer Res 2002;62:5627–31.

    27. Glick AB, WeinbergWC,Wu IH, QuanW, Yuspa SH. Transforming growthfactor beta 1 suppresses genomic instability independent of a G1 arrest,p53, and Rb. Cancer Res 1996;56:3645–50.

    28. Wang J, Jacob NK, Ladner KJ, Beg A, Perko JD, Tanner SM, et al. RelA/p65functions to maintain cellular senescence by regulating genomic stabilityand DNA repair. EMBO Rep 2009;10:1272–8.

    29. Hardee ME, Marciscano AE, Medina-Ramirez CM, Zagzag D, Narayana A,Lonning SM, et al. Resistance of glioblastoma-initiating cells to radiationmediated by the tumor microenvironment can be abolished by inhibitingtransforming growth factor-beta. Cancer Res 2012;72:4119–29.

    30. Dayal R, SinghA, PandeyA,Mishra KP. Reactive oxygen species asmediatorof tumor radiosensitivity. J Cancer Res Ther 2014;10:811–18.

    31. Trachootham D, Alexandre J, Huang P. Targeting cancer cells by ROS-mediated mechanisms: a radical therapeutic approach? Nat Rev DrugDiscov 2009;8:579–91.

    32. Barcellos-HoffMH,Dix TA. Redox-mediated activationof latent transform-ing growth factor-beta 1. Mol Endocrinol 1996;10:1077–83.

    33. Liu RM, Gaston Pravia KA. Oxidative stress and glutathione in TGF-beta-mediated fibrogenesis. Free Radic Biol Med 2010;48:1–15.

    34. Li N, Karin M. Ionizing radiation and short wavelength UV activate NF-kappaB through two distinct mechanisms. Proc Natl Acad Sci U S A1998;95:13012–7.

    35. Morgan MJ, Liu ZG. Crosstalk of reactive oxygen species and NF-kappaBsignaling. Cell research 2011;21:103–15.

    36. Oshimori N, Oristian D, Fuchs E. TGF-beta promotes heterogeneity anddrug resistance in squamous cell carcinoma. Cell 2015;160:963–76.

    37. HeW, Li AG,Wang D, Han S, Zheng B, GoumansMJ, et al. Overexpressionof Smad7 results in severe pathological alterations in multiple epithelialtissues. EMBO J 2002;21:2580–90.

    38. Landstrom M, Heldin NE, Bu S, Hermansson A, Itoh S, ten Dijke P, et al.Smad7 mediates apoptosis induced by transforming growth factor beta inprostatic carcinoma cells. Curr Biol 2000;10:535–8.

    39. Malkoski SP, Wang XJ. Two sides of the story? Smad4 loss in pancreaticcancer versus head-and-neck cancer. FEBS Lett 2012;586:1984–92.

    40. Mishra AK, Kadoishi T,Wang X,Driver E, Chen Z,Wang XJ, et al. Squamouscell carcinomas escape immune surveillance via inducing chronic activa-tion and exhaustion of CD8þ T cells co-expressing PD-1 and LAG-3inhibitory receptors. Oncotarget 2016;7:81341–56.

    Clin Cancer Res; 25(2) January 15, 2019 Clinical Cancer Research818

    Luo et al.

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/

  • 2019;25:808-818. Published OnlineFirst September 5, 2018.Clin Cancer Res Jingjing Luo, Li Bian, Melanie A. Blevins, et al. Modelwithout Compromising Oral Cancer Therapy in a Xenograft Mouse Smad7 Promotes Healing of Radiotherapy-Induced Oral Mucositis

    Updated version

    10.1158/1078-0432.CCR-18-1081doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2018/09/05/1078-0432.CCR-18-1081.DC1

    Access the most recent supplemental material at:

    Cited articles

    http://clincancerres.aacrjournals.org/content/25/2/808.full#ref-list-1

    This article cites 38 articles, 10 of which you can access for free at:

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

    Subscriptions

    Reprints and

    [email protected]

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

    Permissions

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

    .http://clincancerres.aacrjournals.org/content/25/2/808To request permission to re-use all or part of this article, use this link

    on June 10, 2021. © 2019 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst September 5, 2018; DOI: 10.1158/1078-0432.CCR-18-1081

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-18-1081http://clincancerres.aacrjournals.org/content/suppl/2018/09/05/1078-0432.CCR-18-1081.DC1http://clincancerres.aacrjournals.org/content/25/2/808.full#ref-list-1http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/25/2/808http://clincancerres.aacrjournals.org/

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages false /GrayImageMinResolution 200 /GrayImageMinResolutionPolicy /Warning /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 300 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages false /MonoImageMinResolution 600 /MonoImageMinResolutionPolicy /Warning /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 900 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (None) /PDFXOutputConditionIdentifier () /PDFXOutputCondition () /PDFXRegistryName () /PDFXTrapped /False

    /CreateJDFFile false /Description > /Namespace [ (Adobe) (Common) (1.0) ] /OtherNamespaces [ > /FormElements false /GenerateStructure false /IncludeBookmarks false /IncludeHyperlinks false /IncludeInteractive false /IncludeLayers false /IncludeProfiles false /MarksOffset 18 /MarksWeight 0.250000 /MultimediaHandling /UseObjectSettings /Namespace [ (Adobe) (CreativeSuite) (2.0) ] /PDFXOutputIntentProfileSelector /NA /PageMarksFile /RomanDefault /PreserveEditing true /UntaggedCMYKHandling /LeaveUntagged /UntaggedRGBHandling /LeaveUntagged /UseDocumentBleed false >> > ]>> setdistillerparams> setpagedevice