12
Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis and Extracellular MatrixDirected Spheroid Formation Yuta Kasagi 1,2 , Yui Harada 1 , Yosuke Morodomi 1,2 , Toshiki Iwai 1,2,3 , Satoru Saito 1 , Kumi Yoshida 1 , Eiji Oki 2 , Hiroshi Saeki 2 , Kippei Ohgaki 2 , Masahiko Sugiyama 2 , Mitsuho Onimaru 4 , Yoshihiko Maehara 2 , and Yoshikazu Yonemitsu 1 Abstract Peritonitis carcinomatosa is an advanced and intractable state of gastrointestinal and ovarian cancer, where mechanistic elucidation might enable the development of more effective therapies. Peritoneal dissemination of this type of malignancy has been generally thought to initiate from "milky spots" of primitive lymphoid tissues in the peritoneal cavity. In this study, we offer evidence challenging this idea, based on the nding that tumor implantation and directional dissemination was not required for the presence of milky spots, but rather SCF/CXCL12expressing niche-like cells located at the border regions of perivascular adipose tissue. Interestingly, we found that peritoneal cavity lavage uid, which specically contains peritoneal collagen type IV and plasma bronectin, dramati- cally facilitated spheroid formation of murine and human colon cancer cells. Spheroid formation strongly induced the expression of CXCR4 in an Sp1-dependent manner to promote niche-directed metastasis. Notably, disrupting sphere forma- tion or inhibiting Sp1 activity was sufcient to suppress tumor dissemination and potentiated chemosensitivity to 5-uoro- uracil. Our ndings illuminate mechanisms of peritoneal can- cer dissemination and highlight the Sp1/CXCR4/CXCL12 sig- naling axis as a rational target for the development of thera- peutics to manage this intractable form of malignancy. Cancer Res; 76(2); 34757. Ó2016 AACR. Introduction Peritonitis carcinomatosis is an advanced and intractable state of gastrointestinal and ovarian cancers (1). Because this pathology remains highly resistant to the current standard therapeutics, including chemotherapeutic agents, and shows a high mortality ratio, its underlying mechanism requires elucidation. Once the cancer cells have detached from the primary tumor, they are thought to oat in the ascites as single cells or as clusters. At the initial stage of the peritoneal dissemination, cancer cells and their clusters have been shown to attach and adhere to the mesothelial layer, and then to form disseminated colonies via proteolytic activity, (1) and it has generally been thought that the peritoneal tumor dissemination occurs randomly in the perito- neal cavity (2) In support of this idea, an analysis of 102 involved regions in human subjects revealed random peritoneal dissemi- nation to the mesentery/small bowel (21.7%), pelvis (20.6%), omemtum/transverse colon (19.6%), ileocecal area (17.6%), and so on (3). On the other hand, some recent studies have indicated that the microdistribution of peritoneally disseminated cancer cells may not be completely random: so-called "milky spots," i.e., primitive lymphoid tissues in the peritoneal cavity of humans and animals, detected mainly in the greater omentum and the pelvic oor, have been suggested as the predominant sites for cancer dissemination (46). However, questions have arisen in regard to this milky spots hypothesis, as milky spots have been shown to be rare in the mesentery (5, 7), even though mesenteric dissemina- tion is not rare in clinical and experimental settings. To consider an alternative mechanism, therefore, knowledge of the underlying tumor dissemination in the peritoneal cavity irrespective of the location of milky spots is needed. A possible clue to such an alternative mechanism was rst provided by Yasumoto and colleagues (2), showing the essential role of the CXCR4/CXCL12 axis during peritoneal dissemination using human gastric cancer cells; their results indicated that the CXCR4 antagonist AMD3100 (8) was effective to prevent perito- neal carcinomatosis and accumulation of ascites. However, their ndings raised a number of further questions, including (i) the role of the alternative receptor, CXCR7, for CXCL12; (ii) whether the CXCR4/CXCL12 axis is truly not required for CXCR4-negative 1 R&D Laboratory for Innovative Biotherapeutics, Kyushu University Graduate School of Pharmaceutical Sciences, Fukuoka 812-8582, Japan. 2 Department of Surgery and Science, Kyushu University Grad- uate School of Medical Sciences, Fukuoka, Japan. 3 Chugai Pharma- ceutical Co., Ltd., Tokyo, Japan. 4 Department of Pathology, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan. Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Y. Kasagi and Y. Harada contributed equally to this article. Corresponding Author: Yoshikazu Yonemitsu, R&D Laboratory for Innovative Biotherapeutics, Graduate School of Pharmaceutical Sciences, Kyushu Univer- sity, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan. Phone: 819-2642- 6337; Fax: 819-2642-6342; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-15-1563 Ó2016 American Association for Cancer Research. Cancer Research www.aacrjournals.org 347 on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

  • Upload
    others

  • View
    2

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

Molecular and Cellular Pathobiology

Peritoneal Dissemination Requires anSp1-Dependent CXCR4/CXCL12 Signaling Axisand Extracellular Matrix–Directed SpheroidFormationYuta Kasagi1,2, Yui Harada1, Yosuke Morodomi1,2, Toshiki Iwai1,2,3, Satoru Saito1,Kumi Yoshida1, Eiji Oki2, Hiroshi Saeki2, Kippei Ohgaki2, Masahiko Sugiyama2,Mitsuho Onimaru4, Yoshihiko Maehara2, and Yoshikazu Yonemitsu1

Abstract

Peritonitis carcinomatosa is an advanced and intractablestate of gastrointestinal and ovarian cancer, where mechanisticelucidation might enable the development of more effectivetherapies. Peritoneal dissemination of this type of malignancyhas been generally thought to initiate from "milky spots" ofprimitive lymphoid tissues in the peritoneal cavity. In thisstudy, we offer evidence challenging this idea, based on thefinding that tumor implantation and directional disseminationwas not required for the presence of milky spots, but ratherSCF/CXCL12–expressing niche-like cells located at the borderregions of perivascular adipose tissue. Interestingly, we foundthat peritoneal cavity lavage fluid, which specifically contains

peritoneal collagen type IV and plasma fibronectin, dramati-cally facilitated spheroid formation of murine and humancolon cancer cells. Spheroid formation strongly induced theexpression of CXCR4 in an Sp1-dependent manner to promoteniche-directed metastasis. Notably, disrupting sphere forma-tion or inhibiting Sp1 activity was sufficient to suppress tumordissemination and potentiated chemosensitivity to 5-fluoro-uracil. Our findings illuminate mechanisms of peritoneal can-cer dissemination and highlight the Sp1/CXCR4/CXCL12 sig-naling axis as a rational target for the development of thera-peutics to manage this intractable form of malignancy. CancerRes; 76(2); 347–57. �2016 AACR.

IntroductionPeritonitis carcinomatosis is an advanced and intractable state

of gastrointestinal andovarian cancers (1). Because this pathologyremains highly resistant to the current standard therapeutics,including chemotherapeutic agents, and shows a high mortalityratio, its underlying mechanism requires elucidation.

Once the cancer cells have detached from the primary tumor,they are thought to float in the ascites as single cells or as clusters.At the initial stage of the peritoneal dissemination, cancer cellsand their clusters have been shown to attach and adhere to themesothelial layer, and then to form disseminated colonies via

proteolytic activity, (1) and it has generally been thought that theperitoneal tumor dissemination occurs randomly in the perito-neal cavity (2) In support of this idea, an analysis of 102 involvedregions in human subjects revealed random peritoneal dissemi-nation to the mesentery/small bowel (21.7%), pelvis (20.6%),omemtum/transverse colon (19.6%), ileocecal area (17.6%), andso on (3). On the other hand, some recent studies have indicatedthat the microdistribution of peritoneally disseminated cancercells may not be completely random: so-called "milky spots," i.e.,primitive lymphoid tissues in the peritoneal cavity of humans andanimals, detected mainly in the greater omentum and the pelvicfloor, have been suggested as the predominant sites for cancerdissemination (4–6). However, questions have arisen in regard tothismilky spots hypothesis, asmilky spots have been shown to berare in the mesentery (5, 7), even though mesenteric dissemina-tion is not rare in clinical and experimental settings. To consideran alternativemechanism, therefore, knowledge of theunderlyingtumor dissemination in the peritoneal cavity irrespective of thelocation of milky spots is needed.

A possible clue to such an alternative mechanism was firstprovided by Yasumoto and colleagues (2), showing the essentialrole of the CXCR4/CXCL12 axis during peritoneal disseminationusing human gastric cancer cells; their results indicated that theCXCR4 antagonist AMD3100 (8) was effective to prevent perito-neal carcinomatosis and accumulation of ascites. However, theirfindings raised a number of further questions, including (i) therole of the alternative receptor, CXCR7, for CXCL12; (ii) whetherthe CXCR4/CXCL12 axis is truly not required for CXCR4-negative

1R&D Laboratory for Innovative Biotherapeutics, Kyushu UniversityGraduate School of Pharmaceutical Sciences, Fukuoka 812-8582,Japan. 2Department of Surgery and Science, Kyushu University Grad-uate School of Medical Sciences, Fukuoka, Japan. 3Chugai Pharma-ceutical Co., Ltd., Tokyo, Japan. 4Department of Pathology, KyushuUniversity Graduate School of Medical Sciences, Fukuoka, Japan.

Note: Supplementary data for this article are available at Cancer ResearchOnline (http://cancerres.aacrjournals.org/).

Y. Kasagi and Y. Harada contributed equally to this article.

Corresponding Author: Yoshikazu Yonemitsu, R&D Laboratory for InnovativeBiotherapeutics, Graduate School of Pharmaceutical Sciences, Kyushu Univer-sity, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan. Phone: 819-2642-6337; Fax: 819-2642-6342; E-mail: [email protected]

doi: 10.1158/0008-5472.CAN-15-1563

�2016 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org 347

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 2: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

tumor dissemination; (iii) how to explain the paradox betweenthe milky spot hypothesis and the abundant expression ofCXCL12 in the peritoneal mesothelial cells observed in theirstudy.

In the current study, therefore, we extensively address theprecise molecular and cellular mechanisms of peritoneal dis-semination to understand the intractable nature of peritonitiscarcinomatosa.

Materials and MethodsMice

Male 7- to 8-week-old Balb/c and C57BL/6mice were obtainedfrom KBT Oriental Co., Ltd. (Charles River Grade). Male 8-week-old C57BL/6-Tg (CAG-EGFP) mice were obtained from the Shi-zuoka Laboratory Animal Center. The mice were kept underspecific pathogen-free and humane conditions. The animalexperiments were reviewed and approved by the InstitutionalAnimal Care andUseCommittee and by the Biosafety Committeefor Recombinant DNA Experiments of Kyushu University. Theseexperiments were also done in accordance with the recommenda-tions for the proper care and use of laboratory animals andaccording to The Law (No. 105) and Notification (No. 6) of theJapanese Government, and theNIHGuide for the Care andUse ofLaboratory Animals.

Flow cytometric analysisCells were stained with the following FITC-, PE-, PE-Cy5-, or

APC-conjugated monoclonal antibodies (mAb): CXCR4 andCXCR7 (Biolegend). The appropriate conjugated isotype-matched IgGs were used as controls. Cells were analyzed usinga FACSCalibur with CellQuest software (Becton Dickinson) andFlowJo 7.6 software (Tree Star Inc.).

ImmunohistochemistryEn face. The mesentery was cut on poly-L-lysine–coated slides(VWR) and rinsed with PBS. Samples were immunolabeled withthe following antibodies after incubation with Blocking OneHisto (Nakalai Tesque) to eliminate nonspecific binding: SCF(polyclonal rabbit anti-mouse SCF; Sigma-Aldrich Japan; 1:400)or CXCL12 (polyclonal goat anti-mouse SDF-1; Santa Cruz Bio-technology, Inc.; 1:100). Slides were stained for at least 30minutes with primary antibodies specific for the antigens.The slides were then washed thoroughly with PBS and incubatedwith the following secondary antibodies for 30 minutes: DyLight549-labeled donkey anti-rabbit IgG (Biolegend; 1:500) orCF488A-labeled donkey anti-goat IgG (Biotium Inc.; 1:250). Afterthoroughly washing with PBS, the slides were mounted in DAPI-containing Vectashield (Vector Laboratories Inc.) and examinedusing a BZ-9000 microscope (Keyence Corp.).

Fresh-frozen cross-section. SCF and CXCL12 protein expressionswithin the mesentery were immunohistochemically determinedusing the same antibodies as used for the en face method. CT26spheres were immunolabeled with the following antibodies afterincubation with Blocking One Histo (Nakalai Tesque) to elimi-nate nonspecific binding: CXCR4 (polyclonal rabbit anti-mouseCXCR4; Santa Cruz Biotechnology, Inc.; 1:100) or SP1 (polyclon-al rabbit anti-mouse SP1; Santa Cruz Biotechnology; 1:100). Inbrief, OCT compound–mounted frozen sections of the peritone-um and CT26 spheres were incubated with Blocking OneHisto to

eliminate nonspecific binding. Tissue sections were reacted over-night with primary antibody at 4�C. After washing, sections werereacted with the appropriate DyLight-549 or CF488A-labeledsecondary antibody at 4�C for 30 minutes. After thoroughlywashing with PBS, the slides were mounted in DAPI-containingVectashield and examined using a BZ-9000 microscope (KeyenceCorp.). Non-immune rabbit IgG or goat IgG (isotype-matched)was also used instead of the respective primary antibody as anegative control.

Cell lines and culture conditionsThemurine (CT26) andhuman (Lovo,HCT116,Colo-201, and

SW602) colon cancers were purchased from the ATCC. The HRAcells were provided by the Cell Resource Center for BiomedicalResearch at Tohoku University (Sendai, Japan; ref. 9). These celllines weremaintained in RPMI1640medium supplemented with10% FBS, penicillin, and streptomycin in a humidified atmo-sphere containing 5% CO2 at 37�C. The growth and morphologyof the lines were found to be consistent with the original descrip-tions of the lines, and PCR confirmed there was no mycoplasmacontamination. Parental CT26 cells were transfected with theenhanced GFP expression simian lentiviral vector-GFP (10) andselected by single-cell cloning.

Mouse model of tumor disseminationFor the murine peritoneal metastasis model, CT26GFP cells

(2 � 106) were injected intraperitoneally with or withoutAMD3100 (0.25 mg/head; Sigma-Aldrich Japan), neutralizinganti-mouse CXCR4 (clone 2B11, rat IgG2bk, 2 mg/head; Biole-gend), anti-mouse CXCR7 (clone 8F11-M16, mouse IgG2bk, 2mg/head; Biolegend), anti-mouse CXCL12/SDF-1 antibody(Clone #79014, mouse IgG1, R&D Systems) or the appropriateisotype-matched IgGs in 200 mL PBS. CT26 cells were treated withor without mitomycin-C (MMC; 10 mg/mL for 1 hour) or aphi-dicolin (0.1 mg/mL for 24 hours) and rinsed with PBS beforeinjection. Twenty-four hours after the injection, the peritoneaewere cut on poly-L-lysine–coated slides and rinsed with PBS. Theslides were mounted on DAPI-containing Vectashield (VectorLaboratories) and examined using a BZ-9000 microscope.

CFSE assayCT26GFP cells were treated with or without MMC (10 mg/mL

for 1 hour) or aphidicolin (0.1 mg/mL for 24 hours) and rinsedwith RPMI medium. The cells were CFSE-labeled and cultured at37�C in a humidified 5% CO2 incubator for 24, 48, or 72 hoursbefore analysis. These cells were analyzed using a FACSCaliburwith CellQuest and FlowJo 7.6 software.

Cell adhesion and clustering assayCT26GFP cells were cultured on a 24-well culture plate with or

without PBS (control), AMD3100 (1.25 mg/mL), or anti-CXCR4for 24 hours to determine the frequencies of cell adhesion. Toassess the effects of AMD3100 and anti-CXCR4 for the clusteringof CT26GFP cells, the cells were cultured on an MPC treatmentplate (MD6 with Lid Low-Cell Binding; Nalge Nunc Internation-al) with or without each reagent for 24, 48, or 72 hours.

Quantitative reverse transcription-PCRTotal RNA was extracted and cDNA was synthesized using a

SuperscriptIII cells direct cDNA synthesis system (Invitrogen)

Kasagi et al.

Cancer Res; 76(2) January 15, 2016 Cancer Research348

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 3: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

according to the manufacturer's protocol. Real-time PCR wasperformed in a Step One Plus Real Time PCR System(Life Technologies) using TaqMan Fast Universal PCRMaster Mix(Life Technologies) according to the manufacturer's protocol.The specific primer and probe set used in this study was asfollows: NRF1 (Mm01135606_m1), YY1 (Mm00456392_m1),HIF1a (Mm00468869_m1), NF-kb (Mm01310378_m1),FoxC1 (Mm01962704_s1), FoxC2 (Mm00546194_s1),SP1 (Mm00489039_m1), Foxo1 (Mm00490672_m1), Bcl2(Mm00477631_m1), PAX3 (Mm00435491_m1), b-actin(Mm00607939_s1), a-tubulin (Mm02528102_g1). All experi-ments were carried out in triplicate, and the data were analyzedusing Step One Software ver. 2.1 (Life Technologies). Themessenger RNA expression levels were standardized usingb-actin messenger RNA expression levels in each sample.

ELISAThe culture medium and cell lysate were used as samples. The

protein amounts of type IV collagen, cFN, and FN were deter-mined using a mouse specific ELISA kit (type IV collagen,E90149Mu: Uscn Life Science Inc.; cFN, BG-MUS10681: Blue-gene, Shanghai, China; FN, ab108849: Abcam) according to themanufacturer's protocol. Three independent experiments wereperformed.

Sp1 gene silencing by siRNATransfection of CT26 cells with siRNA was done using Lipo-

fectamine RNAiMAX Transfection Reagent (Life Technologies)according to the manufacturer's protocol. Briefly, siRNAs, includ-ing an siRNA targeting Sp1 (Mm_Sp1_5223_s/_as; Sigma-Aldrich) and a scrambled control (sc-37007; Santa Cruz Biotech-nology), were used at a final concentration of 10 nmol/L, andtransfection reagent was used at the dilution of 1:500 (v/v).

Chemotherapeutic sensitization against CT26 malignantascites

Coll-L (75 U/mL) administered twice, at the time of tumor cellinjection and 6 hours later, significantly inhibited the spheroidalformation as well as mesenteric metastasis in vivo (see Fig. 5A). Allmice that received higher doses (i.e., 150 and 300 U/mL) diedwithin 24 hours due to lysis of the mesentery associated withmassive intestinal necrosis, and therefore, 75 U/mL was selectedas the optimal dose. The sensitivity of CT26 tumor cells to 5-fluorouracil (5-FU) and L-OHP under monolayer or sphere for-mation was examined in vitro. The design of the in vivo experimentis shown in Fig. 5B. Briefly, at the time of intraperitoneal inoc-ulation of CT26 tumor cells, each chemical at each concentrationor buffer was administered simultaneously. Six hours later, 5-FU(50 mg/kg) or buffer was administered in all groups except forGroup C, and thereafter, 5-FU or buffer was administered weekly.Animal death was monitored daily and body weight was mea-sured at 3-day intervals.

Sampling of the human mesenteryThe experiments using human surgical sections were reviewed

and approved by the Institutional Review Board of KyushuUniversity Graduate School of Medicine (Approval No. 25-341). Human mesentery samples from seven patients who pro-vided written informed consent and who underwent surgicalresection for primary cancers (2 females and 5 males; age range:

45–72 years; one case of gastrointestinal stromal tumor and sixcases of colon cancer) were subjected to the organ culture studyand to the identification of CAR niche-like cells.

Tissue processing of human mesentery and spheroid adhesionassay

Goat anti-human CXCL12 (sc-6193; Santa Cruz BiotechnologyInc.) and rabbit anti-human SCF (SAB3500292; Sigma-AldrichJapan) were used to identify CAR cells on the human mesenterysamples. LoVo-GFP (positive for CXCR4) and HCT116-CSFE(negative for CXCR4 in monolayer) at 104 cells were maintainedfor 24 hours to form spheroids on a 96-well plate that had beenpretreated with MPC. Forty to fifty spheroids were added ontoeachmediumof slicedhumanmesentery as organ culture thatwasplaced on the 6-well plate. Two days later, each mesentery wasplaced onto the slide grass, and en face immunohistochemicalanalysis for human SCF was performed.

Statistical analysisAll data were expressed as the means � SEM. The data were

examined statistically by Dunnett test or one-way ANOVA withScheffe adjustment. Aprobability value ofP<0.05was consideredstatistically significant. Statistical analyses were determined usingStatView software (SAS Institute).

ResultsMilky spots are not necessary for tumor dissemination

To confirm the localization of so-called milky spots and theirpotential role in the peritoneal dissemination of cancers, we firstidentified milky spots in the omentum and mesentery of Balb/c(Fig. 1A) and C57BL6j (data not shown) mice. As in the previousreports (4–6), we observed toluidine blue–stained cell aggregatesmainly on the surface of narrow fat tissue stripes of the omentum(Fig. 1A, arrowheads), but not in the mesentery (Fig. 1B, top) inboth animal strains. In addition, unlike in a previous study, (7)despite extensive observations we failed to identify fat-associatedlymphoid clusters (FALC), which have been considered to bedifferent from milky spots in the mesenteries of these animalstrains under developed specific pathogen-free conditions. Incontrast, intraperitoneal injection of OK-432, a streptococcalpreparation, could induce FALC-like structures (Fig. 1B, bottom),suggesting that a proinflammatory reaction may be a cause ofFALC formation.

Next, we assessed the distribution of disseminated tumornodules in the omentum andmesentery by using stably enhancedGFP-labeled CT26murine colon cancer cells that were establishedusing a simian lentiviral vector (10) after single-cell selection(CT26GFP; Supplementary Fig. S1). Twenty-four hours afterintraperitoneal administration of CT26GFP (2� 106 cells/head),the omentum and mesentery were examined under a dissectingfluorescentmicroscope (Fig. 1C andD).We also included a groupsimultaneously administered AMD3100, a specific inhibitor forCXCR4 (8), in these experiments. As expected, multiple GFP-positive tumor nodules were frequently observed in narrow fattissue stripes of the omentum (5), but not in not other fat tissues(Fig. 1C, top), and their occurrence was diminished by AMD3100(Fig. 1C, bottom), suggesting that milky spots may be the pre-dominant sites for tumor dissemination mediated by CXCR4.However, even though there were no milky spots, frequent GFP-positive nodules that were sensitive to AMD3100 were also

Molecular Targets of Tumor Dissemination

www.aacrjournals.org Cancer Res; 76(2) January 15, 2016 349

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 4: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

observed in the mesentery (Fig. 1D). These findings were alsoconfirmed bymeasurements of thewet weight of narrow fat tissuestripes and mesenteries at 1 weeks after CT26GFP tumor cellinoculation (Fig. 1E and F).

Together, these results indicate that narrow fat tissue stripes inthe omentum and mesentery are the predominant sites forCXCR4-dependent tumor dissemination; however, milky spotsare not always necessary for peritoneal dissemination.

CXCR4-dependent tumor metastasis to the mesentery isdirectional to CXCL12-expressing niche-like cells

A previous study suggested that peritoneal cells abundantlyexpress CXCL12 and that tumor dissemination might occur atrandom (2); however, our experiments shown in Fig. 1 unexpect-edly revealed early tumor nodules at the border area of perivas-cular adipose tissue (Fig. 2A, arrowheads). In fact, subsequentquantitative examination revealed that more than 90% of these

nodules were located at the border area of perivascular adiposetissue (Fig. 2A, graph) when nodules larger than 30 mm indiameter were counted. Unlike in the previous report (2), theexpression of CXCL12 in parietal peritoneum samples obtainedfrom the abdominal wall was much lower than those in theretroperitoneal fat, mesentery, and omentum narrow fat tissuestripes (Fig. 2B), suggesting that the parietal peritoneummay notbe important for tumor dissemination.

Next, we determined the localization of CXCL12-expressingcells by en face observation using mesenteric tissue, and here wealso examined the expression and localization of stem cell factor(SCF) simultaneously, because the CXCL12/CXCR4 axis is largelyknown for its niche-related proteins, which play roles in hema-topoiesis and cancer metastasis (11–15). Cells in the mesentery,possibly mesothelium cells and perivascular adipocytes, abun-dantly expressed SCF, but in some cases, these cells also coex-pressed CXCL12 (Fig. 2C). Importantly, the SCFþ/CXCL12þ cells

AC

D

B

E

F

100 µm

100 µm

100 µm

100 µm

100 µm

100 µm

Figure 1.Milky spots are not necessary for tumor dissemination. Each experiment was done more than three times and showed similar results. A, representative en facefindings of the toluidine blue-stained mouse omentum for detecting milky spot–like structures by a dissecting microscope. Note the cell aggregation on thenarrow fat tissue stripe (right, arrowheads). B, representative en face findings of the toluidine blue-stained mouse mesentery for detecting milky spot–like and/orFALC structures by dissecting microscopy. No blue-stained cell aggregate can be seen in the mesentery from na€�ve mice (top). Bottom, a FALC-like structure in themesentery from mice intraperitoneally treated with OK-432 (0.01K.E/head). Note the islands of cell aggregates (arrows). H&E, hematoxylin and eosin. C,representative en face findings of metastatic lesions on the mouse narrow fat tissue stripe 24 hours after intraperitoneal inoculation of CT26GFP (top).These disseminated lesions were completely diminished by simultaneous injection with AMD3100, a specific inhibitor for CXCR4. D, representative en facefindings ofmetastatic lesions on themousemesentery 24 hours after intraperitoneal inoculation of CT26GFP (top). These disseminated lesionswere also completelydiminished by simultaneous injection with AMD3100. E, representative findings of the mouse omentum (top) and mesentery (bottom) 7 days after CT26GFPinoculation with or without AMD3100. F, graphs indicate the net weight of mouse narrow fat tissue stripes of the omentum (left graph) andmesentery (right graph).Note that AMD3100 almost abolished the weight gain due to the tumor dissemination.

Kasagi et al.

Cancer Res; 76(2) January 15, 2016 Cancer Research350

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 5: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

were frequently located at the border regions of perivascularadipose tissue (Fig. 2D, inset C, arrows) but rather rarely seen inmembranous regions (Fig. 2D, insets A and B, arrow), althoughsingle SCFþ cells were abundant. We considered that these SCFþ/CXCL12þ cells might be so-called "CXCL12-abundant reticular(CAR) cells," (16) and indeed, these cells were confirmed to havethe reticular morphology typical of CAR cells (Fig. 2E, arrows).

We then examined the 10 mesenteric samples with CT26GFPtumor metastasis that were immunohistochemically determinedto contain CAR-like cells, and approximately 85% of the noduleslarger than 30 mm in diameter were located on the SCFþ cells (Fig.2F). These results indicated that CXCR4-dependent tumor metas-tasis to the mesentery is directional to CXCL12-expressing niche-like cells, but not random.

However, this raised two additional questions: (i) how was thevisible size ofmesenteric metastasis realized within 24 hours aftertumor cell inoculation, and (ii) whywas AMD3100 so effective, asshown in Fig. 1F, even though CT26GFP and its parent cellsexpress very low levels of CXCR4?

Dramatic facilitation of spheroid formation in PCLF ismediated by the cooperative action of collagen type IV andplasma fibronectin

We first assessed the detailed process of mesenteric dissem-ination. To do this, we determined the time courses of therecovery of PCLF and mesenteric metastasis after intraperito-neal inoculation of CT26GFP cells; representative time coursesare shown in Fig. 3A. Single cells as well as scattered cell

% Number of nodules (n = 10)

A

C D

E F

B

% Number of nodules (n = 10)

Mem Bo

(na

On Free

Figure 2.Site-directed peritoneal dissemination targeting SCFþ/CXCL12þ niche-like cells. Experiments were each done more than three times and showed similar results.CT26GFP cells (2 � 106) were injected intraperitoneally. A, grossly visible GFPþ peritoneal dissemination arises within 24 hours after intraperitoneal tumorcell injection. Note that amajority of disseminated tumor nodules were seen at the border regions of perivascular adipose tissue (inset A). Among the GFPþ nodulesthat were more than approximately 30 mm in diameter, 90.7% (mean) were located at the border area of perivascular adipose tissue (graph). B, proteinexpression level of CXCL12 at each site of the intraperitoneal tissue. The previously identified parietal peritoneum showed a lower level of CXCL12 than themesentery or the narrow fat tissue of the omentum. C, representative en face findings of the mouse mesentery immunohistochemically stained for SCF (red) withCXCL12 (green), suggesting that some SCFþ cells would be CAR–like cells (white arrows). D, representative findings of the mouse mesentery on cross sectionsidentifying the localization of CAR cells (CXCL12þ/SCFþ). The two top left panels show the tissue processing for perivascular adipose tissue (bracket, a branch of themesenteric artery is indicated by elastica van Gieson staining) associated with the membranous region. The left bottom panel is a low powered view of the mergedfluorescence (SCF, red; CXCL12, green; and nuclei, DAPI) and the hatched areas of A and B (in the membranous region), and that of C (at the border area ofperivascular adipose tissue) correspond to the respective inset panels. Membranous regionswere generally composedof SCFþ/CXCL12� (inset B) and rarely showedSCFþ/CXCL12þCAR cells (inset A, arrow). In contrast, such doubly positive CAR cellswere frequently seen at the border area of perivascular adipose tissue, as shownin inset C (white arrows). E, representative en face findings of the mouse mesentery immunohistochemically stained for SCF (red) as examined by a fluorescentmicroscope. Nuclear staining was done using DAPI (blue). Note the scattered solitary SCFþ cells showing typical reticular figures (white arrows). F, among theGFPþ nodules that were more than approximately 30 mm in diameter, 85.3% (mean) were on SCFþ cell sheets.

Molecular Targets of Tumor Dissemination

www.aacrjournals.org Cancer Res; 76(2) January 15, 2016 351

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 6: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

clusters were recovered without apparent mesenteric metasta-sis within 1 and 3 hours after tumor inoculation. Importantly,apparent and grossly visible spheroids (Fig. 3A, arrowheads) aswell as tumor nodules on the mesentery began to be observedfrom 6 hours after inoculation, and at 12 hours, the size ofspheroids was dramatically increased (Fig. 3A, arrows) andsingle cell recovery was very rare. The time course of theprocess of the spheroid formation in vivo seemed to be muchearlier than that seen in vitro, suggesting that the peritonealcavity might contain substances that facilitated sphere forma-tion of the CT26 cells. To confirm this, a floating cultivation ofCT26 cells was performed in fresh medium (fresh RPMI) aswell as in the medium that was used after intraperitonealirrigation (PCLF-RPMI). Surprisingly, the spheroid formationwas dramatically facilitated in the PCLF-RPMI (Fig. 3B, red

square) compared with that seen in fresh RPMI (Fig. 3B, bluesquare), indicating that soluble substances in the peritonealcavity may be involved in the accelerated formation ofspheroids.

Next, since such dissemination has been foundonly at 24hoursor later after inoculation of tumor cells, we examined whether ornot cell divisionmight be involved in this process. CT26 cells werepretreated with a cell-cycle inhibitor, either aphidicolin or mito-mycin-C (MMC), and confirmed to be near cell-cycle arrest byFACS analyses for CFSE using parent CT26 cells (5- or 6-(N-Succinimidyloxy-carbonyl)-fluorescein 3',6'-diacetate; data notshown), then intraperitoneally inoculated. However, the resultsshowed that therewere no significant differences among groups inthe frequency of sphere formation (Fig. 3C), indicating that celldivision was not important in this process.

Mesentery

Untreated

Untreated

treatedAphidicolin-

Aphidicoline(n = 23) (n = 11) (n = 9)

MMC

Protein content in PCLF

Fresh-RPMI

A

C

F

B

D

E

Nod

ules

/vas

cula

r loo

p

Undetectable

ng/mL/head

Collagenase: 5 mg/mL

Figure 3.Facilitated spheroid formation of cancer cells in the peritoneal cavity is mediated by preexisting collagen type IV (Coll-IV) and plasma fibronectin (pFN).Each experiment was done more than three times and showed similar results. A, the representative time courses of the recovery of PCLF and mesentericmetastasis after intraperitoneal inoculation of CT26GFP cells. Single cells as well as scattered cell clusters were recovered without apparent metastasis within 1 and3 hours after tumor inoculation. Grossly visible spheroids (arrowheads) as well as tumor nodules on the mesentery began to be observed from 6 hours afterinoculation, and at 12 hours after inoculation, the size of spheroids was increased (arrows) and single cell recovery was very rare. B, PCLF facilitated sphereformation in vitro. Floating cultivation of CT26 was done in the fresh medium (fresh RPMI) as well as the medium that was used after intraperitonealirrigation (PCLF-RPMI). C, cell cycle inhibitionby aphidicolin ormitomycinCdid not affect the formation of disseminated nodules. D, spontaneous spheroid formationand PCLF-mediated facilitation of spheroid formation were sensitive to collagenase. In both cases, CT26 cells could not form spheroids. E, Coll-IV and pFNcooperatively facilitated spheroid formation in vitro. Coll-IV, but neither Coll-I nor plasma/cellular fibronectins (pFN and cFN), facilitated the sphere formation(green squares), compared with baseline (blue squares). Importantly, the combination of Coll-IV and pFN showed the optimal effect (red squares). F, involvementof Coll-IV and pFN in the spheroid formation. E and F, measurement of Coll-IV and pFN in murine PCLF by ELISA (E), and immunohistochemical detection of Coll-IVand pFN in the CT26 spheroids (F) recovered from the peritoneal cavity.

Kasagi et al.

Cancer Res; 76(2) January 15, 2016 Cancer Research352

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 7: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

To identify the protein(s) essential to the accelerated sphereformation, an in vitro spheroid formation assay was performedusing two major ECM-sensitive proteases, collagenase andhyaluronidase (CT26 cells expressed hyaluronan receptorCD44 and collagen/laminin/RGD receptors integrin b1/a5;Supplementary Fig. S2A). Importantly, the sphere formationwas completely abolished by collagenase (Fig. 3D), but was notsensitive to hyaluronidase or anti-integrin–neutralizing anti-bodies (Supplementary Fig. S2B and C), and these findingswere common to all four human colon cancer cell lines tested(Supplementary Fig. S2D). An extensive spheroid formationassay also revealed that collagen type IV (Coll-IV), but neitherColl-I nor plasma/cellular fibronectins (pFN and cFN), facili-tated the sphere formation (Fig. 3E, green squares). Important-ly, the combination of Coll-IV and pFN showed the optimaleffect (Fig. 3D, red squares), indicating that Coll-IV is essentialand pFN supports the formation of spheroids. In fact, consid-erable amounts of Coll-IV and pFN were found in murine PCLF(Fig. 3F), and these ECM proteins were certainly involved inthe spheroids that were recovered from the peritoneal cavity

(Fig. 3F), results that explain very well the clinical findingspreviously reported by our laboratory (17).

Role of spheroid formation onCXCR4 expression, involvementof Sp1

Next, we investigated the role of the CXCR4/CXCL12 axisduring mesenteric tumor dissemination. For this purpose, wefirst assessed the expressions of CXCR4 andCXCR7, an alternativereceptor for CXCL12, in response to inoculation with CT26GFP.The results showed that the expression of CXCR4 was very low(Supplementary Fig. S1b); however, administration ofaCXCR4, aneutralizing antibody for CXCR4, either alone or together withaCXCR7 or AMD3100 or aCXCL12 antibody significantly inhib-ited the mesenteric microdissemination, while neither aCXCR7alone nor control IgG showed a significant inhibition of mesen-teric dissemination of tumor cells (Fig. 4A), indicating that theexpression of CXCR4 in tumor cells, but not that of CXCR7,significantly contributed to peritoneal dissemination.

These paradoxical data raise a question, namely, if CXCR4playsan essential role in niche-directed tumor dissemination, why was

In vitro

UntreatedControl

Untreated

UntreatedUntreated

Monolayer Sphere

Sphere

Mithramycin A (nmol/L)

(0 nmol/L) 0 nmol/L

500 nmol/L

(500 nmol/L) 0 4 6 82Mesenteric nodules/vascular loop

P < 0.01 n = 8, each1 h 3 h 12 h

aCXCL12aCXCR4/7aCXCR7aCXCR4

aCXCR4(n = 7)

aCXCR7(n = 6)

aCXCR4/7(n = 14)

aCXCL12(n = 8)

AMD3100(n = 14)

A

C

E F

D

BN

od

ule

s/va

scu

lar

loo

p

Cont. siRNA

Mesentery24 h

Isotype

Figure 4.Spheroid formation-induced CXCR4 expression is Sp1 transcription factor-dependent. Each experiment was done more than three times and showed similarresults. A, CXCR4 played a significant role during disseminated nodule formation. Just before intraperitoneal injection of tumor cells, 1 � 106 CT26GFP cellswere treated with neutralizing aCXCR4, aCXCR7, both, or aCXCL12 antibody. Pretreatment with AMD3100 was also included. B, method and detection of theupregulation of CXCR4 expression via spheroid formation in vitro. Twenty-four hours after floating culture, the spheroids were subjected to FACS analysisafter propagation with EDTA (5 mol/L) or to immunohistochemistry. C, neither AMD3100 nor aCXCR4 abrogated CT26GFP sphere formation. The right graphindicates the diameters of the sphere. D and E, Sp1 was essential for the sphere formation–induced CXCR4 expression. Spheroid-induced upregulation ofCXCR4 was abrogated by pretreatment with the Sp1-specific inhibitor mithramycin A in a dose-dependent manner (D) and with Sp1-specific siRNA (E). F, Sp1 wasessential for the mesenteric metastasis of GFPþCT26 cells. Mesenteric metastasis of GFPþCT26 cells was significantly inhibited by the pretreatment withmithramycin A without any effect on spheroid formation in vivo.

Molecular Targets of Tumor Dissemination

www.aacrjournals.org Cancer Res; 76(2) January 15, 2016 353

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 8: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

the basal expression of CXCR4 on the CT26 monolayer so low?We hypothesized that sphere formation might accelerate theexpression of CXCR4, and as shown in Fig. 4B, this was indeedconfirmed by FACS and immunocytochemistry. NeitherAMD3100 nor aCXCR4 affected the formation of the spheresthemselves (Fig. 3c), indicating that spheroid formation itselfmaybe a key event in the upregulation of CXCR4.

However, the inhibitor for FAK, a major signal moleculedownstream of integrin signaling, did not affect the sphere-induced CXCR4 expression (Supplementary Fig. S3A), and FNs(Supplementary Fig. S3B, left) or Coll-I and/or Coll-IV (data notshown) also did not alter the expression of CXCR4. In addition,CXCR4 expression on the monolayer was not upregulated byeither cultivation under a hypoxic condition, the addition of aninhibitor for hypoxia-inducible factor (HIF; Supplementary Fig.S3B, right), or the addition of an inhibitor for NF-kB (data notshown), unlike in previous reports (18, 19). Only two stressconditions, i.e., the presence of CoCl2 (20) and serum starvation(21), were identified as inducers of CXCR4 on the CT26 mono-layer (Supplementary Fig. S3C and D). However, the antioxidantN-acetyl cysteine did not have any effect on spheroid-inducedCXCR4 expression (Supplementary Fig. S3C, right). The latter hadbeen coexpressed with insulin-like growth factor-1 receptor

(IGFIR; Supplementary Fig. S3D), an inducer and transactivatorof CXCR4 (21); however, picropodophyllin, an inhibitor ofIGFIR, did not affect the expression level of CXCR4 (data notshown).

Therefore, we returned to a review of the transcriptional reg-ulation of CXCR4, based on the structure of its promoter region(22). As no gene encoding a transcriptional factor that regulatesCXCR4 exhibited a change in expression (Supplementary Fig. S3Eand F), we focused on the Sp1 transcription factor, which targetsGC boxes irrespective of its own transcriptional levels (23). Asshown in Fig. 4DandE, spheroid-induced upregulation ofCXCR4was abrogated by the Sp1-specific inhibitor mithramycin A in adose-dependent manner and by Sp1-specific siRNA. Importantly,mesenteric metastasis of GFPþCT26 cells was significantly inhib-itedby thepretreatmentwithmithramycinAwithout any effect onspheroid formation in vivo (Fig. 4F), indicating that Sp1-mediatedoverexpression of CXCR4 plays an essential role in directionalmetastasis during tumor dissemination.

Chemotherapeutic sensitization against tumor disseminationvia inhibition of Sp1, CXCR4, or spheroid formation

To assess the effect of inhibiting the CXCR4 axis as well asspheroid formation itself on the therapeutic effect of chemotherapy

Mesentery

(75 U/mL)

Cel

l num

ber (

%)

Cel

l num

ber (

%)

5-FU (mmol/L) L-OHP (mmol/L)

Sphere Monolayer

(75 U/mL)

(500 nmol/L)

Days after tumor inoculation

A

C D

B

Buffer

(75U/mL)

Figure 5.Modulation of the CXCR4 axis achieves chemotherapeutic sensitization against a murine malignant ascites model. A, Coll-L inhibited spheroid formation of GFPþCT26cells in vivo. Coll-L (75 U/mL) administered twice, at the time of tumor cell injection and 6 hours later, significantly inhibited the spheroidal formation aswell as mesenteric metastasis in vivo. All mice that received higher doses (i.e., 150 and 300 U/mL) died due to lysis of the mesenteric tissue associated withmassive intestinal necrosiswithin 24 hours. Each experiment was performed three to five timeswith similar results. B, experimental design. At the time of intraperitonealinoculation of CT26 tumor cells, each chemical at each concentration or buffer was administered simultaneously. Six hours later, 5-FU (50 mg/kg) or buffer wasadministered, except inGroupC, and thereafter 5-FUorbufferwasadministeredweekly. C, sensitivity ofCT26 tumor cells in amonolayer or sphere formation to5-FUandL-OHP in vitro. Sphere formation made the cells more resistant to both antitumor agents compared with the cells in a monolayer. Each group contained three animals.D, time course of body weight (B.W.) and survival (þ, dead animals over the 100 days after tumor inoculation; *, surviving animals over the same period).

Kasagi et al.

Cancer Res; 76(2) January 15, 2016 Cancer Research354

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 9: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

in vivo, we first optimized the concentration of collagenase-L(Coll-L) as 75 U/mL required to inhibit spheroid formation ofGFPþCT26 cells in vivo (Fig. 5A).

Before determining the experimental design (Fig. 5B), thesensitivity of two independent chemotherapeutic agents that arethe commonly used agents for colon cancers, namely 5-FU and L-OHP, was determined. Sphere formation made the cells signifi-cantlymore resistant to both antitumor agents compared with thecells remaining in a monolayer (Fig. 5C, P < 0.05). As 5-FU wasmore effective than L-OHP when the drugs were used at lowerconcentrations, we selected 5-FU for use in the subsequent in vivoexperiments (Fig. 5D).

All animals administered buffer as a control (Group A) weredead within 32 days, and 5-FU treatment (Group B) could notprolong their survival time significantly (P ¼ 0.12). In contrast,early inhibition of sphere formation by Coll-L (Group C) as wellas use of AMD3100 (Group D) or of mithramycin A (Group E)resulted in over 100 days of long-term survival in 3 (Groups C andD) and 4 (Group E) of the 8 animals in each group.

These results indicated that the inhibition of sphere formationin the peritoneal cavity and of directional sphere seedingmight bea potential strategy to sensitize the disseminated tumor cellsagainst chemotherapeutic agents.

Identification of CAR-like cell aggregates, accelerated sphereformation, and directional tumor metastasis in human tissueand cells

Finally, we addressed the question of the relevance of thesefindings to human tissue and cells.

A portion of the surgically resected human mesentery (Sup-plementary Fig. S4A, left) was subjected to en bloc immunohis-tochemistry for SCF, and en face observation under a dissectingfluorescent microscope demonstrated scattered SCFþ cells (Sup-plementary Fig. S4A, right, red staining, white arrows) on andaround the perivascular adipose tissue (Supplementary Fig. S4A,right, yellow bipolar arrow). Double immunohistochemistry(Supplementary Fig. S4B) for SCF (red) and CXCL12 (green) wasperformed to identify CAR-like cells (white arrows) using cryosec-tions, and doubly positive cells were frequently seen at the borderarea of perivascular adipose tissue (white arrows).

Next, we assessed the representative accelerated sphere forma-tion and AMD3100- and mithramycin A–sensitive mesenterictumor dissemination of two independent human colon cancercell lines, LoVo and HCT116, in the peritoneal cavity of nudemice. As shown in Supplementary Fig. S4C, spontaneous expres-sion of CXCR4 was seen in the monolayer of LoVo cells, but notthat of HCT116 cells, while CXCR4 expression was seen afterspheroid formation (arrow). Murine PCLF facilitated the sphereformation of these human colon cell lines in vitro (SupplementaryFig. S4d, red squares), but the formations were abolished whenColl-L was used (Supplementary Fig. S4d, green squares). Thesecells were intraperitoneally inoculated into the nude mice. Sevendays after inoculation of GFP-labeled LoVo cells (LoVo-GFP) orCFSE-labeled HCT116 cells (HCT116-CFSE), the disseminationwas mainly seen at the margin of the perivascular adipose tissue(Supplementary Fig. S4e, left, inset A and B), which was similar tothe findings obtained by using CT26GFP cells. These dissemina-tionswere significantly abrogated byAMG3100ormithramycin A(Supplementary Fig. S4e, right).

Surgically obtained mesenteric tissues were placed on a 6-wellplate, and medium containing spheres of HCT116-CFSE was

added. Two days layer, these tissues were placed on a glass plate,and en bloc immunohistochemistry for SCF was done. En facefindings demonstrated that LoVo-GFP (Supplementary Fig. S4F,top bottom) as well as HCT116-CFSE (Supplementary Fig. S4F,left bottom) spheres (green) were frequently present on the SCFþ

cell sheet (red). Among the 39 and 35 green nodules counted, 26(66.7%) and 23 tumors (65.7%) were located on the SCFþ cellsheet, respectively.

Subsequently, we askedwhether themajorfindings obtained inthis study using colon cancer cells were also representative to aHRA human ovarian cancer, an alternative malignant neoplasmthat frequently induces peritoneal dissemination. As expected,sphere formation–induced spontaneous expression of CXCR4(Supplementary Fig. S5a), PCLF-mediated acceleration of thesphere-forming process (Supplementary Fig. S5b), and inhibitionof peritoneal dissemination of HRA cells in nude mice byAMD3100 as well as mythramycin (Supplementary Fig. S5C)were also representative.

Together with these findings, intraperitoneal acceleration ofsphere formation of cancer cells and CAR-like cell-directed dis-semination of spheres could be common mechanisms in humantissue and cancers, at least in case of gastrointestinal and ovariancancers.

DiscussionMainly using CT26 murine colon cancer, that has been shown

to share molecular features with aggressive, undifferentiated,refractory human colorectal carcinoma cells (24), we heredemonstrated the whole molecular and cellular mechanismsof peritoneal dissemination. The key findings of the currentstudy were as follows: (i) intraperitoneally inoculated tumorcells can be implanted to narrow fat tissue stripes of theomentum with milky spots as well as to border regions ofperivascular adipose tissue of the mesentery without milkyspots, and both are sensitive to a CXCR4 inhibitor, AMD3100,suggesting that milky spots are not required to form dissemi-nated cancer metastasis in the peritoneal cavity; (ii) SCFþ/CXCL12þ CAR-like cells located at the border area of perivas-cular adipose tissue of the mesentery are a predominant dis-semination site of tumor cells; (iii) peritoneal Coll-IV and pFNcooperatively facilitated the sphere formation of cancer cellswithin 3 to 6 hours; (iv) spheroid formation strongly promotedthe expression of CXCR4 in an Sp1 transcription factor-depen-dent fashion (a schematic representation of a possible mecha-nism of tumor dissemination is provided in Supplementary Fig.S6). Also, (v) CAR-niche–directed and CXCR4/CXCL12 axis–dependent metastasis were representative in human cancer cellsand mesentery.

Although a previous study simply demonstrated the essentialrole of the CXCR4/CXCL12 axis during tumor dissemination(2, 11–15), the current study made a related but apparentlydistinct contribution, i.e., the first demonstration of the globalmolecular and cellular mechanism of tumor dissemination. Spe-cifically, this study was the first to demonstrate that Coll-IV andpFN-dependent facilitation of spheroid formation within theinitial hours after dissemination was essential to induceCXCR4/CXCL12-dependent tumor dissemination, irrespective ofthe basal expression of CXCR4. First, it has been demonstratedthat spheroids form tight junctions containing both E-cadherinand ZO-1 to protect themselves against anticancer agents (25). In

Molecular Targets of Tumor Dissemination

www.aacrjournals.org Cancer Res; 76(2) January 15, 2016 355

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 10: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

fact, cancer spheroids are much more resistant to irinotecan thancancer cells in a monolayer (26). Second, spheroid formationpromptly induces antiapoptotic genes that might be involved inthe mechanism of drug resistance (25). Other metabolic changesvia the formation of spheroids, including hypoxia (27), starvation(28), anaerobic metabolism (27), proinflammatory potential(29, 30), as well as cancer stem cell–like potency (31, 32) mayalso contribute to the intractable potentials of disseminatedcancer foci (Supplementary Fig. S7).

One importantfinding obtained in this studywas that early andtransient inhibition of sphere formation as well as of the CXCR4axis rendered tumor cells in the peritoneal cavity significantlymore sensitive to chemotherapeutic agents. In any case, these dataindicated that the inhibition of the process of sphere formation aswell as CXCR4-dependent directed dissemination might be effec-tive to render tumor cells more sensitive to chemotherapeuticagents, and therefore these strategies need to be addressed infuture clinical studies.

In conclusion, we here demonstrated the globalmechanisms ofperitoneal cancer dissemination at the molecular and cellularlevels. Our results showed that the CXCR4-dependent interactionof tumor cells with the mesenteric CAR cell–like niche as well asthe spheroid formation occurring before tumor dissemination isthe critical mechanism underlying peritoneal carcinomatosis. Thedevelopment of technologies that disrupt this process would behighly desirable for the treatment of patients with peritonealcarcinomatosis.

Disclosure of Potential Conflicts of InterestT. Iwai is a researcher from Chugai Pharmaceuticals Inc. Y. Maehara and Y.

Yonemitsu report receiving a commercial research grant. No potential conflictsof interest were disclosed by the other authors.

Authors' ContributionsConception and design: Y. Kasagi, Y. Harada, Y. Morodomi, Y. YonemitsuDevelopment of methodology: Y. Kasagi, Y. Harada, Y. Morodomi, K. YoshidaAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): Y. Kasagi, Y. Harada, Y. Morodomi, S. Saito, T. Iwai,K. Ohgaki, M. Sugiyama, Y. Maehara, Y. YonemitsuAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): Y. Kasagi, Y. Harada, Y. Maehara, Y. YonemitsuWriting, review, and/or revision of the manuscript: Y. Kasagi, E. Oki,Y. YonemitsuAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): Y. Kasagi, S. Saito, K. Yoshida, Y. Harada, E. Oki,M. OnimaruStudy supervision: Y. Maehara, Y. Yonemitsu

AcknowledgmentsThe authors thank Ryoko Nakamura, Yukiko Nagatomo, Risa Tanaka, and

Natsumi Maeda for their invaluable help with the animal experiments and cellculture, and Mr. Hiroshi Fujii for tissue processing. The authors also thank KNInternational, Ltd. for their assistance in the revision of the English usage in thisarticle.

Grant SupportThis work was supported in part by a Grant-in-Aid (No. 15H05792 to

Y. Maehara; No. 13376659 to Y. Yonemitsu) from the Japanese Ministryof Education, Culture, Sports, Science, and Technology, and by financialsupport for collaboration research with Chugai Pharmaceutical Co., Ltd.(Y. Yonemitsu).

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received June 8, 2015; revised September 6, 2015; accepted October 7, 2015;published OnlineFirst January 7, 2016.

References1. Chung M, Kozuch P. Treatment of malignant ascites. Curr Treat Options

Oncol 2008;9:215–33.2. Yasumoto K, Koizumi K, KawashimaA, Saitoh Y, Arita Y, Shinohara K, et al.

Role of the CXCL12/CXCR4 axis in peritoneal carcinomatosis of gastriccancer. Cancer Res 2006;66:2181–7.

3. de Bree E, Koops W, Kr€oger R, van Ruth S, Witkamp AJ, Zoetmulder FA.Peritoneal carcinomatosis from colorectal or appendiceal origin: correla-tion of preoperative CT with intraoperative findings and evaluation ofinterobserver agreement. J Surg Oncol 2004;86:64–73.

4. Mebius RE. Lymphoid organs for peritoneal cavity immune response:milky spots. Immunity 2009;30:670–2.

5. Clark R, Krishnan V, Schoof M, Rodriguez I, Theriault B, Chekmareva M,et al. Milky spots promote ovarian cancer metastatic colonizationof peritoneal adipose in experimental models. Am J Pathol 2013;183:576–91.

6. Oosterling SJ, van der Bij GJ, B€ogels M, van der Sijp JR, Beelen RH,Meijer S,et al. Insufficient ability of omentalmilky spots to prevent peritoneal tumoroutgrowth supports omentectomy in minimal residual disease. CancerImmunol Immunother 2006;55:1043–51.

7. Moro K, Yamada T, Tanabe M, Takeuchi T, Ikawa T, Kawamoto H, et al.Innate productionof T(H)2 cytokines by adipose tissue-associated c-Kit(þ)Sca-1(þ) lymphoid cells. Nature 2010;463:540–4.

8. Donzella GA, Schols D, Lin SW, Est�e JA, Nagashima KA, Maddon PJ, et al.AMD3100, a small molecule inhibitor of HIV-1 entry via the CXCR4 co-receptor. Nat Med 1998;4:72–7.

9. Kikuchi Y, Kizawa I, Oomori K, Miyauchi M, Kita T, Sugita M, et al.Establishment of a human ovarian cancer cell line capable of formingascites in nudemice and effects of tranexamic acid on cell proliferation andascites formation. Cancer Res 1987;47:592–6.

10. Ikeda Y, Goto Y, Yonemitsu Y, Miyazaki M, Sakamoto T, Ishibashi T, et al.Simian immunodeficiency virus-based lentivirus vector for retinal genetransfer: a preclinical safety study in adult rats. Gene Ther 2003;10:1161–1169.

11. Sugiyama T, Kohara H, NodaM, Nagasawa T. Maintenance of the hemato-poietic stem cell pool by CXCL12-CXCR4 chemokine signaling in bonemarrow stromal cell niches. Immunity 2006;25:977–88.

12. M€uller A, Homey B, Soto H, Ge N, Catron D, Buchanan ME, et al.Involvement of chemokine receptors in breast cancer metastasis. Nature2001;410:50–6.

13. Taichman RS, Cooper C, Keller ET, Pienta KJ, Taichman NS, McCauley LK.Use of the stromal cell-derived factor-1/CXCR4 pathway in prostate cancermetastasis to bone. Cancer Res 2002;62:1832–7.

14. Zeelenberg IS, Ruuls-Van Stalle L, Roos E. The chemokine receptor CXCR4is required for outgrowth of colon carcinomamicrometastases. Cancer Res2003;63:3833–9.

15. Shiozawa Y, Pedersen EA, Havens AM, Jung Y, Mishra A, Joseph J, et al.Humanprostate cancermetastases target the hematopoietic stem cell nicheto establish footholds in mouse bone marrow. J Clin Invest 2011;121:1298–312.

16. Nagasawa T. Microenvironmental niches in the bone marrow required forB-cell development. Nat Rev Immunol 2006;6:107–16.

17. Korenaga D, Funahashi S, Yano K, Maekawa S, Ikeda T, Sugimachi K.Relationship between peritoneal collagen type IV concentrations and thepresence of disseminated metastases in gastric cancer. Arch Surg 1995;130:769–73.

18. Staller P, Sulitkova J, Lisztwan J, Moch H, Oakeley EJ, Krek W. Chemokinereceptor CXCR4 downregulated by von Hippel-Lindau tumor suppressorpVHL. Nature 2003;425:307–11.

Kasagi et al.

Cancer Res; 76(2) January 15, 2016 Cancer Research356

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 11: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

19. Helbig G, Christopherson KW II, Bhat-Nakshatri P, Kumar S, Kishimoto H,Miller KD, et al. NF-kB promotes breast cancer cell migration and metas-tasis by inducing the expression of the chemokine receptor CXCR4. J BiolChem 2003;278:21631–8.

20. Schutyser E, Su Y, Yu Y, Gouwy M, Zaja-Milatovic S, Van Damme J, et al.Hypoxia enhances CXCR4 expression in human microvascular endo-thelial cells and human melanoma cells. Eur Cytokine Netw 2007;18:59–70.

21. Akekawatchai C, Holland JD, Kochetkova M, Wallace JC, McColl SR.Transactivation of CXCR4 by the insulin-like growth factor-1 receptor(IGF-1R) in human MDA-MB-231 breast cancer epithelial cells. J BiolChem 2005;280:39701–8.

22. Tarnowski M, Grymula K, Reca R, Jankowski K, Maksym R, Tarnowska J,et al. Regulation of expression of stromal-derived factor-1 receptors:CXCR4 and CXCR7 in human rhabdomyosarcomas. Mol Cancer Res2010;8:1–14.

23. Wierstra I. Sp1: Emerging roles- Beyond constitutive activationof TATA-lesshousekeeping genes. Biochem Biophys Res Commun 2008;372:1–13.

24. Castle JC, Loewer M, Boegel S, de Graaf J, Bender C, Tadmor AD, et al.Immunomic, genomic and transcriptomic characterization of CT26 colo-rectal carcinoma. BMC Genomics 2014;15:190.

25. Xiang X, Phung Y, Feng M, Nagashima K, Zhang J, Broaddus VC, et al. Thedevelopment and characterization of a human mesothelioma in vitro 3Dmodel to investigate immunotoxin therapy. PLoS ONE 2011;6:e14640.

26. Friedrich J, Seidel C, Ebner R, Kunz-Schughart LA. Spheroid-based drugscreen: considerations and practical approach. Nat Protoc 2009;4:309–24.

27. Franko AJ, Chapman JD. Binding of 14C-misonidazole to hypoxic cells inV79 spheroids. Br J Cancer 1982;45:694–9.

28. Shweiki D, Neeman M, Itin A, Keshet E. Induction of VEGF expression byhypoxia and by glucose deficiency in multicell spheroids: implications fortumor angiogenesis. Proc Natl Acad Sci USA 1995;92:768–72.

29. Gallardo-P�erez JC, Espinosa M, Ceballos-Cancino G, Daniel A, Rodríguez-Enríquez S, Aviles A, et al. NF-kB is required for the development of tumorspheroids. J Cell Biochem 2009;108:169–80.

30. Wada Y, Shimada K, Kimura T, Ushiyama S. Novel p38 MAP kinaseinhibitor R-130823 suppresses IL-6, IL-8 and MMP-13 production inspheroid culture of human synovial sarcoma cell line SW 982. ImmunolLett 2005;101:50–9.

31. Yeung TM, Gandhi SC, Bodmer WF. Hypoxia and lineage specification ofcell line-derived colorectal cancer stem cells. Proc Natl Acad Sci USA2011;108:4382–7.

32. Li L, Bhatia R. Stem cell quiescence. Clin Cancer Res 2011;17:4936–41.

www.aacrjournals.org Cancer Res; 76(2) January 15, 2016 357

Molecular Targets of Tumor Dissemination

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563

Page 12: Peritoneal Dissemination Requires an Sp1-Dependent CXCR4 ... · Molecular and Cellular Pathobiology Peritoneal Dissemination Requires an Sp1-Dependent CXCR4/CXCL12 Signaling Axis

2016;76:347-357. Published OnlineFirst January 7, 2016.Cancer Res   Yuta Kasagi, Yui Harada, Yosuke Morodomi, et al.   Spheroid Formation

Directed−CXCR4/CXCL12 Signaling Axis and Extracellular Matrix Peritoneal Dissemination Requires an Sp1-Dependent

  Updated version

  10.1158/0008-5472.CAN-15-1563doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2016/01/12/0008-5472.CAN-15-1563.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/76/2/347.full#ref-list-1

This article cites 32 articles, 10 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/76/2/347.full#related-urls

This article has been cited by 3 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]

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://cancerres.aacrjournals.org/content/76/2/347To request permission to re-use all or part of this article, use this link

on January 17, 2020. © 2016 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst January 7, 2016; DOI: 10.1158/0008-5472.CAN-15-1563