+ All Categories
Home > Documents > T Lymphocytes Restrain Spontaneous Metastases in …Microenvironment and Immunology T Lymphocytes...

T Lymphocytes Restrain Spontaneous Metastases in …Microenvironment and Immunology T Lymphocytes...

Date post: 27-Jan-2021
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
12
Microenvironment and Immunology T Lymphocytes Restrain Spontaneous Metastases in Permanent Dormancy Irene Romero 1 , Cristina Garrido 3 , Ignacio Algarra 4 , Antonia Collado 2 , Federico Garrido 1,3 , and Angel M. Garcia-Lora 1 Abstract Tumor dormancy is a clinical phenomenon related to immune equilibrium during cancer immunoediting. The mechanisms involved in dormant metastases are poorly understood due to the lack of preclinical models. Here, we present a nontransgenic mouse model in which spontaneous metastases remain in permanent immuno- mediated dormancy with no additional antitumor treatment. After the injection of a GR9-B11 mouse brosarcoma clone into syngeneic BALB/c mice, all animals remained free of spontaneous metastases at the experimental endpoints (38 months) but also as long as 24 months after tumor cell injection. Strikingly, when tumor-bearing mice were immunodepleted of T lymphocytes or asialo GM1-positive cells, the restraint on dormant disseminated metastatic cells was relieved and lung metastases progressed. Immunostimulation was documented at both local and systemic levels, with results supporting the evidence that the immune system was able to restrain spontaneous metastases in permanent dormancy. Notably, the GR9-B11 tumor clone did not express MHC class I molecules on the cell surface, yet all metastases in immunodepleted mice were MHC class Ipositive. This model system may be valuable for more in-depth analyses of metastatic dormancy, offering new opportunities for immunotherapeutic management of metastatic disease. Cancer Res; 74(7); 195868. Ó2014 AACR. Introduction The initiation and progression of cancer in an immuno- competent host involve numerous interactions between tumor cells and the immune system. The immune response exercises selective pressure against tumor cells, eliminating the more immunogenic phenotypes. This constant interaction between the immune system and cancer cells may ultimately result in the selection of less immunogenic "cancer-escape" variants that are able to survive and progress in the host (1, 2). The diverse escape mechanisms developed by cancer cells to evade the immune response (36) include the loss of surface expres- sion of MHC class I molecules (79). This loss may make the tumor cells invisible to T lymphocytes, allowing them to enter an "immunoblindness" stage (10). It is feasible that some cancer cells neither progress nor are destroyed by immune system during this selective process, remaining in a dormant stage and reaching equilibrium with the host tumor microenvironment (1113). Cancer dormancy has been observed in humans (14, 15), and several experimental studies have reported an immunomediated control of primary tumor cells in dormancy (1620). There is considerable evi- dence of metastasis relapse in human cancer patients after long periods of remission, when disseminated metastatic cells can persist for years or even decades as minimal residual disease (21, 22). Other clinical examples related to cancer immune control include tumors that arise after immunosup- pressive treatments (23, 24) and cases of transplanted organs carrying an undetectable tumor that grows after immunosup- pressive treatment of the patient (2527). These clinical phe- nomena support the existence of a state of equilibrium between the host and the cancer cells. The fact that immu- nosuppression can disturb this equilibrium and activate dor- mant cancer cells strongly suggests the existence of an immu- nomediated state of dormancy in these cases. The mechanisms involved in cancer dormancy remain largely unknown, due to difculties in isolating dormant human metastatic cells and constructing preclinical models of dormant metastases. Here, we presented a novel nontrans- genic preclinical mouse model of permanent immunome- diated metastatic dormancy. We have used an extensively studied brosarcoma mouse model (GR9) developed in our laboratory composed of several tumor clones with different MHC class I expression patterns and spontaneous metastatic capacities (28). Thus, the metastatic capacity is elevated in the clones with high MHC class I expression and reduced in those with low MHC class I expression (29). We show that an MHC-Inegative GR9-B11 tumor cell clone did not generate sponta- neous lung metastasis in immunocompetent mice, which Authors' Afliations: 1 Dept. Analisis Clinicos e Inmunologia, UGC Labor- atorio Clínico; 2 Unidad de Investigaci on, Hospital Universitario Virgen de las Nieves, Granada; 3 Departamento de Bioquímica, Biología Molecular e Inmu- nología III, Universidad de Granada, Granada; and 4 Departamento de Cien- cias de la Salud, Universidad de Ja en, Ja en, Spain Corresponding Author: Angel M. Garcia-Lora, UGC Laboratorio Clinico, Hospital Universitario Virgen de las Nieves, Av. de las Fuerzas Armadas 2, 18014, Granada, Spain. Phone: 349-580-20269; Fax: 349-580-20069; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-13-2084 Ó2014 American Association for Cancer Research. Cancer Research Cancer Res; 74(7) April 1, 2014 1958 on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084
Transcript
  • Microenvironment and Immunology

    T Lymphocytes Restrain Spontaneous Metastases inPermanent Dormancy

    Irene Romero1, Cristina Garrido3, Ignacio Algarra4, Antonia Collado2, Federico Garrido1,3, andAngel M. Garcia-Lora1

    AbstractTumor dormancy is a clinical phenomenon related to immune equilibrium during cancer immunoediting.

    The mechanisms involved in dormant metastases are poorly understood due to the lack of preclinical models.Here, we present a nontransgenic mouse model in which spontaneous metastases remain in permanent immuno-mediated dormancy with no additional antitumor treatment. After the injection of a GR9-B11mouse fibrosarcomaclone into syngeneic BALB/c mice, all animals remained free of spontaneous metastases at the experimentalendpoints (3–8 months) but also as long as 24 months after tumor cell injection. Strikingly, when tumor-bearingmice were immunodepleted of T lymphocytes or asialo GM1-positive cells, the restraint on dormant disseminatedmetastatic cells was relieved and lung metastases progressed. Immunostimulation was documented at bothlocal and systemic levels, with results supporting the evidence that the immune system was able to restrainspontaneous metastases in permanent dormancy. Notably, the GR9-B11 tumor clone did not express MHCclass I molecules on the cell surface, yet all metastases in immunodepleted mice were MHC class I–positive. Thismodel system may be valuable for more in-depth analyses of metastatic dormancy, offering new opportunitiesfor immunotherapeutic management of metastatic disease. Cancer Res; 74(7); 1958–68. �2014 AACR.

    IntroductionThe initiation and progression of cancer in an immuno-

    competent host involve numerous interactions between tumorcells and the immune system. The immune response exercisesselective pressure against tumor cells, eliminating the moreimmunogenic phenotypes. This constant interaction betweenthe immune system and cancer cells may ultimately result inthe selection of less immunogenic "cancer-escape" variantsthat are able to survive and progress in the host (1, 2). Thediverse escape mechanisms developed by cancer cells to evadethe immune response (3–6) include the loss of surface expres-sion of MHC class I molecules (7–9). This loss may make thetumor cells invisible to T lymphocytes, allowing them to enteran "immunoblindness" stage (10).

    It is feasible that some cancer cells neither progress nor aredestroyed by immune system during this selective process,remaining in a dormant stage and reaching equilibrium withthe host tumor microenvironment (11–13). Cancer dormancy

    has been observed in humans (14, 15), and several experimentalstudies have reported an immunomediated control of primarytumor cells in dormancy (16–20). There is considerable evi-dence of metastasis relapse in human cancer patients afterlong periods of remission, when disseminated metastatic cellscan persist for years or even decades as minimal residualdisease (21, 22). Other clinical examples related to cancerimmune control include tumors that arise after immunosup-pressive treatments (23, 24) and cases of transplanted organscarrying an undetectable tumor that grows after immunosup-pressive treatment of the patient (25–27). These clinical phe-nomena support the existence of a state of equilibriumbetween the host and the cancer cells. The fact that immu-nosuppression can disturb this equilibrium and activate dor-mant cancer cells strongly suggests the existence of an immu-nomediated state of dormancy in these cases.

    The mechanisms involved in cancer dormancy remainlargely unknown, due to difficulties in isolating dormanthuman metastatic cells and constructing preclinical modelsof dormant metastases. Here, we presented a novel nontrans-genic preclinical mouse model of permanent immunome-diated metastatic dormancy. We have used an extensivelystudied fibrosarcoma mouse model (GR9) developed in ourlaboratory composed of several tumor clones with differentMHC class I expression patterns and spontaneous metastaticcapacities (28). Thus, the metastatic capacity is elevated in theclones with high MHC class I expression and reduced in thosewith lowMHC class I expression (29).We show that anMHC-I–negative GR9-B11 tumor cell clone did not generate sponta-neous lung metastasis in immunocompetent mice, which

    Authors' Affiliations: 1Dept. Analisis Clinicos e Inmunologia, UGC Labor-atorio Clínico; 2Unidad de Investigaci�on, Hospital Universitario Virgen de lasNieves, Granada; 3Departamento de Bioquímica, BiologíaMolecular e Inmu-nología III, Universidad de Granada, Granada; and 4Departamento de Cien-cias de la Salud, Universidad de Ja�en, Ja�en, Spain

    Corresponding Author: Angel M. Garcia-Lora, UGC Laboratorio Clinico,Hospital Universitario Virgen de las Nieves, Av. de las Fuerzas Armadas 2,18014, Granada, Spain. Phone: 349-580-20269; Fax: 349-580-20069;E-mail: [email protected]

    doi: 10.1158/0008-5472.CAN-13-2084

    �2014 American Association for Cancer Research.

    CancerResearch

    Cancer Res; 74(7) April 1, 20141958

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/

  • remained free of metastasis until their euthanasia at the end ofthe assays (3–24 months). Strikingly, immunodepletion of T orasialo GM1-positive cells in the mice awoke the disseminatedmetastatic cells from dormancy, generating lung metastases.

    Materials and MethodsCell lines and IFN-g treatmentGR9 cell line is derived from a mouse fibrosarcoma induced

    by methylcholanthrene in BALB/c mice and has been exten-sively characterized in our laboratory. It is composed of cellclones with distinct H-2 class I expression patterns and met-astatic capacities (29). Spontaneous metastasis assays wereperformed with different GR9 cell clones, and one of these, theGR9-B11 clone, was selected for this study. GR9-B11 and GR9-A7 are clones obtained by a limited dilution method from theGR9 cell line. GR9-B11 and GR9-A7 cell lines were recloned bypicking up individual cells under phase-contrast microscopy.All cell lines were characterized by PCR assay using shorttandem repeat and they were also regularly tested for MHC-Igenotype and surface expression. Cell lines were maintainedin Dulbecco's Modified Eagle Medium (Sigma-Aldrich) sup-plemented with 10% FBS (Life Technologies), 2 mmol/L glu-tamine (Sigma-Aldrich), and antibiotics. In some experiments,cell lines were treated with 100 U/mL IFN-g for 48 hours(Sigma-Aldrich).

    MiceEight-week-old male BALB/c and athymic nu/nu BALB/c

    (Charles River Laboratories) mice were used in the experi-ments. The breeding and care of animals were undertaken incompliance with European Community Directive 86/609/CEEand Spanish law (Real Decreto 1201/2005) for the use oflaboratory animals. Housing and all experimental proceduresinvolving animals were performed according to protocolsapproved by the hospital Animal Care Committee and incompliance with the animal welfare guidelines of the NationalCommittee for Animal Experiments.

    Spontaneous metastasis assayDifferent cell doses (50� 105, 25� 105, 12.5� 105, and 6.25�

    105) of GR9-B11 were subcutaneously injected into the footpadof groups of syngeneic immunocompetent and nude BALB/cmice. The growth of local tumors was recorded 3 times perweek in all animals, measuring the largest diameter of eachtumor with electronic calipers. Tumors were excised when thelargest diameter reached 10 mm. The model resembles met-astatic development in humans after surgical removal of theprimary tumor.Micewere anesthetizedwith 0.04mLdiazepam(Valium, Roche) and 0.1mL ketamine (Ketolar) before removalof the primary tumors with sterilized instruments, usingelectrocautery to minimize bleeding and closing the woundswith surgical clips and adhesive. After the surgery, each animalwas housed alone until recovery from anesthesia. At the end ofthe assays, animals were anesthetized and euthanized bycervical dislocation. A complete necropsy was performed, andthe number of spontaneous metastases was counted. Localtumors and macroscopically visible metastatic nodules wereexcised, disaggregated, and adapted to tissue culture. Then, the

    lungs were fixed in Bouin solution (Sigma-Aldrich) and themicrometastases were counted.

    Preparation of splenic and lung leukocytesSpleens and lungs were excised and gently homogenized in a

    stomacher in cold PBS (Sigma-Aldrich). A tissue fragment wasremoved and a sterile falcon cell strainer (BD Biosciences) wasused to create a single-cell suspension. Red blood cells werelysed with ACK lysing buffer (Gibco) for 5 minutes and thenwashed twice inPBS. Viable cells were counted andused for theantibody staining reaction.

    Flow cytometry analysis of immune cell subsetsFor direct immunofluorescence, the following labeled anti-

    bodies (Miltenyi-Biotech) were used: CD3e-APC, CD4-FITC,CD8-PE, CD25-PE, FoxP3-APC, CD19-FITC, CD49b-FITC,anti-MHC class II-APC, anti-CD11c-PE, and anti-CD11b-FITC.Isotype-matched nonimmune mouse IgGs conjugated withFITC, PE, or APC served as controls. FcR Blocking reagentwas used to block unwanted binding of antibodies to mousecells expressing Fc receptors. Immunofluorescence was doneaccording to themanufacturer's instructions (Miltenyi Biotec),using FoxP3 staining buffer to obtain optimal FoxP3 immu-nofluorescent staining. Cells were analyzed on a FACSCantocytometer (BD Biosciences). Each sample consisted of a min-imum of 5 � 104 cells and was analyzed with CellQuest-Prosoftware.

    MHC class I surface expressionMHC class I surface expression was analyzed by indirect

    immunofluorescence using FACS (FACScan; Becton Dickin-son) according to a standard protocol. In brief, 5 � 105 cellswere washed twice with PBS and incubated for 30 minutes at4�C with the primary antibodies anti-H-2 Kd (K9-18), anti-H-2Dd (34-5-8), and anti H-2 Ld (28.14.8 and 30.5.7), all obtainedfrom the American Type Culture Collection. The secondaryfluorescein isothiocyanate (FITC)-conjugated antibody (anti-mouse FITC IgG/Fab, Sigma-Aldrich) was used in 1:120 dilu-tion for 30 minutes at 4�C in the dark. Isotype-matchednonimmune mouse IgG and cells labeled with the fluoresce-in-conjugated antibody alone served as controls. A minimumof 1� 104 cells were analyzed with CellQuest-Pro software. Allcell lines were studied in baseline conditions and after IFN-gtreatment.

    Real-time RT-PCR analysisAn mRNA isolation kit (Miltenyi-Biotech) was used to

    extract mRNA from tumor cell lines. First-strand cDNA wassynthesized with 100 ng of mRNA using a High CapacityReverse Transcription Kit (Applied Biosystems) in a totalvolume of 20 mL. These cDNAs were diluted to a final volumeof 100mL. Real-time quantitative PCRanalyseswere carried outin the 7500 Fast System (Applied Biosystems), performing PCRreactions in quadruplicate and expressing the values obtainedas means � SD. Quantitative PCR was performed with thePower SYBR Green Master Mix (Applied Biosystems); theprimers and amplicon size for each gene were previouslyreported (30). GADPH and b-actin genes were used as

    Spontaneous Metastases in Immune-Mediated Dormancy

    www.aacrjournals.org Cancer Res; 74(7) April 1, 2014 1959

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/

  • housekeeping genes. PCR conditions were 40 cycles of 15seconds of denaturation at 95�C and 60 seconds at 60�C.

    Immunodepletion protocols in spontaneous metastasisassay

    A total of 12.5 � 105 GR9-B11 cells were injected into thefootpad of syngeneic BALB/c mice. The growth of the tumorswas measured 3 times per week. At around 20 to 22 days, thetumors reached 10mmandwere excised as described above. At151 days, one group of mice was euthanized and the remainingmice were randomly divided among six groups. Four of thesegroups received immunodepletion treatments with monoclo-nal antibodies. The following protocols were used: (i) 100 mganti-CD4 monoclonal antibody (mAb; clone YTS191)þ 100 mganti-CD8 mAb (clone YTS 169), (ii) 100 mg anti-CD4 mAb, (iii)100 mg anti-CD8 mAb, and (iv) 50 mg anti-asialo GM1 (Wakochemicals USA Inc.). A fifth group was treated with 100 mgcontrol immunoglobulin (Polyclonal rat IgG, Sigma-Aldrich),while the remaining group received no treatment (untreatedmice). The agents were administered twice a week for 90 daysfrom day 152 after the tumor cell injection. Depletion of thecorresponding immune subpopulations in the mice was con-firmed by flow cytometry. Mice from each group were eutha-nized at the end of the treatments (day 242 post-cell injection).A complete necropsy was performed, isolating and analyzingthe lungs as described above.

    Statistical analysisData were expressed as means� SD. The Student t test was

    used to compare mean values. A significance level of P < 0.05was assumed for all statistical tests. SPSS 16.0.2 (IBM)was usedfor the data analyses. All statistical tests were two-sided.

    ResultsH-2 class I phenotype ofGR9-B11fibrosarcoma clone cellline and local primary tumors

    GR9 is a methylcholanthrene-induced fibrosarcoma gener-ated in BALB/c mice and is composed of several tumor cloneswith different H-2 class I surface expressions (28). GR9-B11fibrosarcoma cell line was derived from the GR9 tumor bylimited dilution cloning. The GR9-B11 clone was recloned bypicking up individual cells under phase-contrast microscopy.Under baseline conditions, this tumor clone cell line has anegative expression of H-2 Kd, Dd, and Ld molecules (Fig. 1A).IFN-g treatment upregulated all the three molecules (Fig. 1A).Possible mechanisms underlying the loss of surface MHC-Iexpression of GR9-B11 were investigated by analyzing thetranscriptional gene expression of antigen processing machin-ery (APM), H-2 class I heavy chains, and b2-microglobulingenes. The GR9-B11 fibrosarcoma clone was compared with apositive MHC-I GR9-A7 clone cell line, normalizing the datato the expressions of GAPDH and b-actin housekeepinggenes. Figure 1B depicts the results, using the values forGR9-A7 cells as reference (assigned a relative value of 1).GR9-B11 showed a downregulation of H-2 Ld, calreticulin,LMP2, TAP-1, and tapasin (Fig. 1B). These results demonstratethat the molecular mechanism underlying the loss of MHC-Isurface expression involves the coordinated transcriptional

    downregulation of several APM components andMHC-I heavychains.

    In tumor-initiating capacity (TIC) assays, four cell doses (50.0� 105, 25.0 � 105, 12.5 � 105, and 6.25 � 105 cells) were locallyinjected into the footpad in groups of 10mice. The local tumorswere removed from the animals when the largest tumor diam-eter reached 10 mm. The tumors were then adapted to tissueculture to analyze their H-2 class I surface expression incomparisonwith that of the GR9-B11 clone. The surface expres-sion of H-2 class I molecules in vitro was higher in the localtumor cell lines than in the original clone. The tumors showedpositive surface expression of H-2 Kd and Dd molecules andnegative for H-2 Ld molecule in baseline conditions (Fig. 1C).TheH-2 Kd andDdmolecules were strongly upregulated after invitro treatment with IFN-g and showed an even higher expres-sion than what was observed on the original tumor clone.However, two populations were observed with different pat-terns of expressionofH-2 Ldmolecule: in onepopulation, H-2 Ld

    molecule was clearly upregulated, whereas in the other, H-2 Ld

    molecule was negative (Fig. 1C). These results were observed inall local tumors analyzed regardless of the cell dose injected.

    Spontaneous metastasis assays from GR9-B11 clone inimmunocompetent and nu/nu BALB/c mice

    To determine the in vivometastatic capacity of the GR9-B11tumor clone, themice weremonitored weekly for spontaneousmetastatic spread after removal of primary tumors. The miceshowed no signs of disease and were eventually euthanized at90 days after the tumor excision. The autopsy revealed that allmice were metastasis-free, regardless of the cell dose used. Wethen repeated these assays and found that the mice remainedmetastasis-free at 24 months after primary tumor removal,when the mice were euthanized.

    Two possible explanations for these findings were thenexplored: the disseminated metastatic cells might not becapable of migrating or invading, or GR9-B11 metastatic cellsmight be eliminated by the immune system. These possibilitieswere examined by performing spontaneous metastasis assayswith GR9-B11 cells in immunodeficient nu/nu BALB/c mice,injecting 6.25� 105 cells into two groups of 10 nude mice. Thelocal growth rate of tumors in these mice was similar to that inimmunocompetentmice, reaching a largest diameter of 10mmin 32 versus 29 days, respectively. The local tumors wereremoved when this diameter was reached and were adaptedto tissue culture. Analysis of their H-2 class I surface expressionrevealed the same H-2 class I phenotype as expressed by theprimary tumors generated in immunocompetent mice (Fig.1C). The mice were monitored for the appearance of sponta-neous metastases and were euthanized when signs of diseasewere observed. Unexpectedly, lung metastases (range, 1–8)were found in 80% of these mice (Fig. 2A). The immunocom-petent mice served as controls in this assay, and all remainedmetastasis-free (Fig. 2A). These results indicate that GR9-B11cells have intrinsic migratory and invasive capacities. Allmacroscopically visible metastatic nodules were adapted totissue culture, and H-2 class I phenotype analysis revealed thatall of the metastases had the same H-2 class I phenotype,characterized by the high baseline expression of H-2 Kd and Dd

    Romero et al.

    Cancer Res; 74(7) April 1, 2014 Cancer Research1960

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/

  • molecules, which were induced by IFN-g treatment, and theabsence of surface expression of H-2 Ld molecule underbaseline conditions and after IFN-g treatment (Fig. 2B). Thisphenotype represents a new tumor variant that is not presentin the original clone but is observed in the primary tumors.To summarize, the GR9-B11 fibrosarcoma clone generated

    spontaneous metastases in T-cell–immunodeficient nu/nuBALB/c mice, but not in immunocompetent BALB/c mice,and all metastases evidenced total loss of H-2 Ld moleculesurface expression.The immunogenicity of the metastases derived from nude

    mice was evaluated by injecting these metastatic cell lines intothe footpad of immunocompetent BALB/cmice, administering

    different cell doses (12.5 � 105 and 6.25 � 105 cells) in twogroups of 5 mice each. The metastatic cells demonstrated anelevated immunogenicity, being rejected in 67% of the miceinjected with the higher cell dose and in 40% of those injectedwith the lower dose. Moreover, these mice did not developspontaneous metastases.

    Changes in immune cell subpopulations promoted byGR9-B11 tumor cells

    We carried out assays to evaluate individual immune cellsubpopulations at local and systemic level in GR9-B11 tumor-bearing immunocompetent mice. Two groups of 30 mice wereeuthanized on days 25 and 50 after removal of the local tumors,

    A

    B

    C

    00

    2040

    6080

    100

    020

    4060

    8010

    0

    Cou

    nts

    Cou

    nts

    2040C

    ount

    s

    1,4

    1,2

    1

    0,8

    0,6

    0,4

    0,2

    0

    Rel

    ativ

    e m

    RN

    A le

    vel

    H-2

    KH-

    2 D

    H-2

    Lβ2

    mCa

    lnCa

    lr

    LMP2

    LMP7

    Tap1

    Tap2 Tp

    n

    100

    100 101 102 103 104 100 101 102 103 104

    101 102 103

    FL-1

    FL-1

    Negative H-2 K H-2 D H-2 L

    FL-1

    104

    6080

    100

    020

    40C

    ount

    s

    100 101 102 103

    FL-1104

    6080

    100

    120

    Basal

    Basal

    A7 B11

    IFN-γ

    IFN-γ

    Figure 1. H-2 class I surface expression onGR9-B11 clone and in local primarytumors. A, H-2 class I expression of theGR9-B11 fibrosarcoma cell line underbaseline conditions and after treatmentwith IFN-g (100U/mL) for 48 h: H-2 K (grayline), H-2 D (dotted line), and H-2 L (blackline). GR9-B11 is H-2–negative and allthree molecules are induced after IFN-gtreatment. B, transcription levels of H-2class I heavy chains, b2m, and severalAPM components detected by real-timeRT-PCR. Expression level of the genes ofinterest was determined with respect tolevels of b-actin and GAPDHhousekeeping genes. Data for GR9-A7are set to 1. Values, mean � SD of threeindependent experiments performed inquadruplicate; �,P

  • and spleen leukocyte populations were analyzed by flowcytometry (Fig. 3 and Table 1). Tumor-bearing mice showedstatistically significant changes in the lymphocyte subpopula-tions on days 25 (25 d) and 50 (50 d) in comparison with thenontumor-injected animals (NT; P < 0.05), with an increase inCD3þ (44.4 and 53.7 vs. 33.1%, respectively), CD3þCD4þ (33.3and 40.4 vs. 26.1%), and CD3þCD8þ (11.1 and 13.0 vs. 6.9%)lymphocytes, a slight increase inNKTcells (1.0 and 1.4 vs. 0.4%),and an increase in dendritic cells (8.7 and 8.0 vs. 3.0%) andmacrophages (8.6 and 7.1 vs. 3.8%; Fig. 3B and Table 1).

    We also analyzed the changes in lymphocyte subpopulationsin the lungs of mice on day 25 and 50 after removal of the localtumor (Fig. 4A and Table 1), finding a major rise in thepercentage of CD3þ lymphocytes, which had increased to64% on both days, versus 51.9% in nontumor-injected mice(Fig. 4B); this expansion corresponded to increases in T-helperlymphocytes (46.2% and 50.5% vs. 40.6%, respectively) and T-cytotoxic lymphocytes (17.4 and 12.1 vs. 9.7%, respectively; Fig.4B and Table 1). We highlight that the percentage of CD8þ Tlymphocytes had increased by 180% on day 25.

    T or asialo GM1-positive cells maintain dormantspontaneous metastases in a state of equilibrium

    According to the above experiments, the GR9-B11 cloneproduced spontaneous pulmonary metastatic nodules in nudemice but not in immunocompetent mice, which remainedmetastasis-free and developed an immune response.We there-

    fore hypothesized that disseminated metastatic cells would beeliminated by the immune system in immunocompetent miceor, alternatively, would be kept in a dormant state. Thesepossibilities were tested in a new experiment (Fig. 5A), inwhich 12.5 � 105 GR9-B11 cells were injected into the footpadin seven groups of immunocompetent BALB/c mice. The localtumors were removed at 20 to 22 days, and one of the groupswas euthanized at 151 days; the necropsy revealed that nometastases were present in any mice in this group. From day152, the mice in another four groups were treated weekly withanti-CD4 þ anti-CD8 mAbs, anti-CD4 mAb, anti-CD8 mAb, orimmunoglobulins (control Ig group; Fig. 5A). A sixth group ofmice was treated with the anti-asialo GM1 antibody (Fig. 5A).After 3 months of treatment, the mice were euthanized on day242 (Fig. 5A) and the depletion of each subpopulation wasconfirmed by flow cytometry. In the mice treated with controlimmunoglobulin, no metastatic nodules were detected in thenecropsy (Table 2). In the groups treated with anti-CD4þ anti-CD8 mAbs or with anti-CD8 mAb alone, 100% of the micedeveloped spontaneous pulmonary metastases. Similar resultswere found in the group treatedwith anti-asialo GM1 antibody,in which 87% of the mice developed metastases (Table 2).However, in the group in which CD4þ T cells alone weredepleted, only 23% of the mice developed metastases. Thenumber of metastases per mouse differed among the groups(Table 2): it ranged from 4 to 62 micrometastases in the micetreated with anti-CD4 þ anti-CD8 mAbs and from 3 to 17 in

    100 100 101 102 103 104101 102

    Basal

    FL-1

    Negative H-2 K H-2 D H-2 L

    FL-1

    IFN-γ

    103 104

    0 0

    20

    2040

    6080

    100

    120

    4060

    8010

    0

    Cou

    nts

    Cou

    nts

    10

    8

    6

    4

    2

    0BALB/c

    Num

    ber

    of lu

    ng m

    etas

    tase

    s

    A

    B

    nu/nu BALB/c

    Figure 2. Spontaneous metastasis assaysin immunocompetent and nu/nu BALB/cmice. A, the graph depicts the mice withmetastases and the number of lungmetastases per mouse. A dot representsthe number of lungmetastases identified ineach mouse. These results werereproducible in another two independentexperiments. B, H-2 class I phenotype ofspontaneous pulmonary metastases innudemice. All metastases showed a singlephenotype under baseline conditions andafter IFN-g treatment, with positiveexpression of K and Dmolecules alone. H-2K (gray line), H-2D (dotted line), andH-2 L(black line). A representative example isdepicted.

    Romero et al.

    Cancer Res; 74(7) April 1, 2014 Cancer Research1962

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/

  • B

    A

    CD3+ CD19+CD3+CD4+ MHC-II+CD11b+

    MHC-II+CD11c+CD3+CD8+

    MH

    C-l

    l

    80

    60

    40

    20

    0

    50

    40

    30

    20

    10

    0

    12

    10

    8

    6

    4

    2

    0

    Spl

    een

    lym

    phoc

    ytes

    (%

    )

    Spl

    een

    lym

    phoc

    ytes

    (%

    )

    Spl

    een

    popu

    latio

    ns (

    %)

    NT NT NT25d 25d 25d50d 50d 50d

    NT 25d 50d

    100 101 102CD3 APC-A

    103 104 100 101 102CD3 APC-A

    103 104 100 101 102CD3 APC-A

    103 104

    100 101 102 103 104100 101 102 103 104

    100 101 102

    CD11b FITC-A CD11b FITC-A CD11b FITC-A103 104

    100 101 102

    CD4 FITC-A CD4 FITC-A CD4 FITC-A103 104

    100 101 102 103 104 100 101 102 103 104

    100 101 102 103 104100 101 102 103 104100 101 102

    CD11c PE-A CD11c PE-A CD11c PE-A103 104

    100

    101

    102

    103

    104

    CD

    19F

    ITC

    -A

    100

    101

    102

    103

    104

    CD

    19F

    ITC

    -A

    100

    101

    102

    103

    104

    CD

    19F

    ITC

    -A

    100

    101

    102

    103

    104

    100

    101

    102

    103

    104

    100

    101

    102

    103

    104

    CD

    8 P

    E-A

    CD

    8 P

    E-A

    CD

    8 P

    E-A

    100

    101

    102

    103

    104

    MH

    C-ll

    AP

    C-A

    MH

    C-ll

    AP

    C-A

    MH

    C-ll

    AP

    C-A

    MH

    C-ll

    AP

    C-A

    MH

    C-ll

    AP

    C-A

    MH

    C-ll

    AP

    C-A

    100

    101

    102

    103

    104

    100

    101

    102

    103

    104

    100

    101

    102

    103

    104

    100

    101

    102

    103

    104

    100

    101

    102

    103

    104

    Figure 3. Changes in splenic leukocyte populations (lymphocytes,macrophage, anddendritic cells). Thedifferent leukocyte populationswere analyzed at days25 (25 d) and 50 (50 d) after local tumor removal in comparison with nontumor-injected mice (NT). A, a representative experiment showing B (CD3�CD19þ)and T (CD3þCD4þ and CD3þCD8þ) lymphocyte subpopulations, macrophages (MHC-IIþCD11bþ), and dendritic cells (MHC-IIþCD11cþ). B, thegraphs depict the percentages (mean � SD) of CD3þ/CD19þ, CD4þ/CD8þ, and CD11bþ/CD11cþ cells. �, P < 0.05.

    Spontaneous Metastases in Immune-Mediated Dormancy

    www.aacrjournals.org Cancer Res; 74(7) April 1, 2014 1963

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/

  • 50% of the group with CD8þ T lymphocyte–depleted mice butreached 100 micrometastases in the other 50% of this group; itranged from 2 to 35 micrometastases in 62% of asialo GM1-depleted mice but reached more than 100 micrometastases in

    25% of this group; it ranged from 1 to 2 micrometastases in thegroup treated with anti-CD4mAb. Macrometastases were onlydetected in the mice treated with anti-CD4 mAb (13%) or anti-asialo-GM1 (30%) in a range of 1 to 2 per mouse. Mice from a

    Table 1. Changes in splenic and lung leukocyte populations

    Splenic leukocyte populations

    CD3þCD4þ CD3� CD3� CD3þ MHCIIþc MHCIIþc

    CD3þc CD4þc CD8þc CD25þFoxP3þa CD19þc CD49bþc CD49bþ CD11bþb CD11cþb

    NT 33.1 � 4.3 26.1 � 3.1 6.9 � 1.8 4.4 � 0.8 61.0 � 4.2 4.6 � 1.0 0.4 � 0.3 3.8 � 0.1 3.0 � 0.225d 44.4 � 6.8 33.3 � 5.1 11.1 � 1.9 4.3 � 1.0 48.9 � 6.8 5.4 � 1.7 1.0 � 0.2 8.6 � 1.2 8.7 � 0.750d 53.7 � 5.5 40.4 � 4.1 13.0 � 2.9 3.6 � 1.7 41.0 � 4.7 3.7 � 1.0 1.4 � 0.6 7.1 � 1.4 8.0 � 1.6

    Lung lymphocyte populations

    CD3þCD4þ CD3� CD3� CD3þ

    CD3þc CD4þc CD8þc CD25þFoxP3þa CD19þc CD49bþ CD49bþ

    NT 51.9 � 6.3 40.6 � 5.5 9.7 � 1.5 1.9 � 0.7 35.2 � 5.9 12.2 � 0.9 1.8 � 0.825d 64.5 � 8.2 46.2 � 7.0 17.4 � 1.4 3.3 � 0.8 24.2 � 7.9 11.1 � 4.4 1.5 � 1.050d 64.1 � 5.2 50.5 � 6.9 12.1 � 2.2 3.7 � 0.6 26.2 � 2.1 9.6 � 4.0 1.6 � 0.3NOTE: Data are expressed as mean � SD of 30 mice of each group.aPercentage among CD4þ cells.bPercentage among MHC-IIþ cells.cP < 0.05 compared with NT group.

    CD3+ CD19+ CD3+CD4+ CD3+CD8+NT NT25

    d25

    d50

    d50

    d

    Lung

    lym

    phoc

    ytes

    (%

    )

    Lung

    lym

    phoc

    ytes

    (%

    )

    NTA

    B

    25d 50d

    CD3 APC-A

    CD4 FITC-A

    CD

    8 P

    E-A

    CD

    8 P

    E-A

    CD

    8 P

    E-A

    CD4 FITC-A CD4 FITC-A

    CD3 APC-A CD3 APC-A

    100

    100 101 102 103 104 100 101 102 103 104 100 101 102 103 104

    100 100 101 102 103 104101 102 103 104

    CD

    19F

    ITC

    -A

    CD

    19F

    ITC

    -A

    CD

    19F

    ITC

    -A

    100

    100

    101

    102

    103

    104

    100

    101

    102

    103

    104

    100

    101

    102

    103

    10

    100

    101

    102

    103

    10

    100

    101

    102

    103

    1010

    110

    210

    3

    101 102 103 104

    Figure 4. Lung lymphocytes werequantified at different time points. Atdays 25 (25d) and 50 (50d) after removingthe local tumor, the lymphocytesubpopulations were measured andcompared with nontumor-injected mice(NT). A, a representative experimentshows B (CD3�CD19þ) and T(CD3þCD4þ and CD3þCD8þ)lymphocyte subpopulations. B, thegraphs depict the (mean � SD)percentages of CD3þ/CD19þ andCD4þ/CD8þ lymphocytes. �, P < 0.05.

    Romero et al.

    Cancer Res; 74(7) April 1, 2014 Cancer Research1964

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/

  • seventh group that received no treatment were maintained upto 24 months after local tumor removal and remained metas-tasis-free (Table 2).To summarize, we found a higher number of mice with

    metastases and higher number of metastases in the lungs inCD8- or asialo GM1-depleted mice than in CD4-depleted mice.

    The metastases remained in dormancy for 5 months andawoke when T or asialo GM1-positive cells were depleted.

    After necropsy, all macroscopically visible spontaneouspulmonary metastases from immunodepleted mouse groupswere adapted to tissue culture and H-2 class I surface expres-sion was analyzed. All metastases were characterized by apositive expression of Kd and Ddmolecules and a total absenceof Ld molecule expression under baseline conditions, while allthree molecules were induced after IFN-g treatment (Fig. 5B).

    DiscussionThis study describes for the first time a novel nontransgenic

    mouse tumor model of permanent immunomediated meta-static dormancy. In this model, spontaneous metastases arecompletely controlled and maintained in a dormant state bythe wild-type mice immune system, with no application of anyanticancer treatment. The local primary tumors grew rapidly,and the mice remained metastasis-free after tumor removal atthe end of the assays (3–8 months) and for more than 24months. Interestingly, immunodepletion of host T or asialoGM1-positive cells promoted the awakening of dormant dis-seminated spontaneous metastatic cells, which invaded thelungs of the mice. We highlight that the spontaneous metas-tases in our model are dormant, remaining in latency through-out the life of the animals; furthermore, this dormant meta-static state is maintained by the murine immune systemthrough T and asialo GM1-positive cells, with no previousimmunization or treatment of the hosts. Thismetastatic tumor

    Table 2. Number of metastases generated incontrol and depletion groups

    Range

    GroupsMetastasisincidence

    Micro-PMs

    Macro-PMs

    Untreated 0/30 0 0Control Ig 0/30 0 0Anti-CD4þCD8 30/30 4–62 0Anti-CD8 30/30 3–17 (50%) 0

    >100 (50%) 0

    Anti–asialo-GM1 26/30 (87%) 2–35 (62%)�

    1 (30%)

    0 (32%)>100 (25%) 0

    Anti-CD4 7/30 (23%) 1–2 (23%) 2 (13%)0 (10%)

    Abbreviations: micro-PMs, pulmonary micrometastases;macro-PMs, pulmonary macrometastases.

    Negative

    A

    B

    H-2 D H-2 LH-2 K

    Basal IFN-γ

    100

    0 060

    8040

    2010

    012

    0

    2040

    6080

    100

    FL1-Height FL1-Height

    Cou

    nts

    Cou

    nts

    101 102 103 104 100 101 102 103 104

    Figure 5. Spontaneous metastasis assays in immunocompetent and immunodepleted BALB/c mice. A, time schedules of the immunodepletion protocols inspontaneous metastasis assays. The mice were injected with GR9-B11 cells on day 0. The local tumors were removed between days 20 and 22. Oneof the groupswas euthanized at 151days. Five groups ofmicewere treated twice aweek for 90 daysby intraperitoneal injection ofmAbs (anti-CD4þ anti-CD8or anti-CD4 or anti-CD8 or anti-asialo GM1) or Igs (control Ig), beginning on day 152, whereas another group remained untreated. All micewere euthanized onday 242, except the group (vii) control. B, H-2 class I surface expression ofmetastases awoken fromdormancy. H-2K (gray line), H-2 D (dotted line), andH-2 L(black line). Allmacrometastases showedpositive expressionofH-2KandDmolecules alone under baseline conditions, and all threemoleculeswere inducedafter IFN-g treatment. A representative example of three independent experiments is depicted.

    Spontaneous Metastases in Immune-Mediated Dormancy

    www.aacrjournals.org Cancer Res; 74(7) April 1, 2014 1965

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/

  • murine model faithfully resembles the progression of humancancers in which a long latency period with minimal residualdisease can follow the primary tumor resection, with themetastatic cells clinically manifesting years or even decadeslater (31–33). TheGR9-B11metastatic tumormodel is a uniqueand reproducible experimental system for detailed analysis ofthe phenomenon of immunomediated metastatic dormancy.

    Various authors recently demonstrated that the immuneresponse can delay cancer progression. Koebel and colleaguesstudied aMCA-induced tumormousemodel and reported thatthe immune response kept occult primary tumors in a state ofequilibrium and that the cancer progressed after the jointdepletion of CD4/CD8 cells and the depletion of IFN-g or IL-12(34). Unlike the present study, they investigated dormantpreneoplastic or neoplastic tumor cells but not metastatictumor cells. Another group found that the IFN-g produced bylung NK cells played a major role against the development ofpulmonary metastases (35), although these were experimentaland not spontaneousmetastases and theywere not dormant. Itwas also reported that cancer progression was accelerated bydepletion of CD8þ T cells in a RET.ADD melanoma transgenicmice model, although the nondepleted mice also eventuallydeveloped cancer (36). In our assays, the mice remainedmetastasis-free until euthanized.

    In the GR9 fibrosarcoma mouse model, MHC-I expressionon different tumor clones has been indirectly correlated within vivo TIC (28). In the present study, tumor cells grewrapidly immediately after the injection of GR9-B11, possiblyescaping from the immune system due to their altered MHC-I phenotype with low antigen presentation capacity. Aninverse behavior was reported in spontaneous metastasisassays, which showed a higher metastatic capacity in theMHC-positive GR9 tumor clones than in the MHC-negativeclones. A very high spontaneous metastatic capacity waspreviously reported for an MHC-I–positive clone of the GR9tumor model, GR9-A7, whose metastases were also MHC-I–positive (37). However, unlike findings for the GR9-B11clone, GR9-A7 tumor growth and metastatic disseminationproduced an immunosuppressive effect in the hosts. Resto-ration of the host immune response by immunotherapytreatments completely eradicated the spontaneous metas-tases and the animals remained metastasis-free (37). In thepresent study, immune stimulation was observed during theperiod of GR9-B11 primary tumor growth and disseminationof spontaneous metastatic cells, which was characterized byan increase in immune cells, especially T lymphocytes. Inboth cases, the spread of metastases was dependent on theimmune effect promoted by the primary tumor cells. Ourgroup also previously reported that GR9-B9, another tumorclone of GR9 primary fibrosarcoma, produced a largernumber of spontaneous pulmonary metastases in nu/nuBALB/c mice than in immunocompetent BALB/c mice(38, 39). These results and the present findings suggest thatimmunosurveillance plays a key role against metastaticprogression and that GR9-B11 tumor cells promote animmune response able to completely control disseminatedmetastatic cells and maintain them in a state of dormancy.This proposition is further supported by the finding that

    spontaneous metastases were generated in nu/nu andimmunodepleted BALB/c mice injected with GR9-B11 tumorcells.

    Baseline MHC-I surface expression was higher on allspontaneous metastases originated from the GR9-B11 tumorclone in immunodeficient and immunodepleted mice incomparison with GR9-B11 tumor cells. A similar phenome-non was previously reported for the GR9-B9 tumor clone, onwhich MHC-I expression is absent, finding that all metastasesin immunodeficient mice were MHC-I–positive under base-line conditions (38, 39). Hence, the MHC-I alterations werereversible (i.e., "soft lesions") in both clones (39, 40). Theseresults strongly suggest that MHC-I loss is not a requirementfor escape in the absence of a T-cell–mediated immuneresponse. Recent investigations in chemoresistance humanmodels of hormone-refractory prostate cancer (HRPC) iden-tified an HLA-I–negative cell subpopulation of the bulkpopulation of primary and metastatic prostate cancer tissue(41). These HLA-I–negative tumor cells exhibited resistanceto chemotherapy and their number correlated with the stageof the disease and its recurrence. In agreement with thepresent data, these HLA-I–negative tumor cells displayed ahigher TIC in immunodeficient murine hosts in vivo, with theprimary tumors again reproducing the initial HLA-I pheno-typic heterogeneity of the original tumor (41). In this context,relapse after adoptive cell transfer therapies in patients withmelanoma has been related to the reversible downregulationof antigen expression (42). In brief, MHC-I–positive andMHC-I–negative cells may exist in a dynamic and inter-changeable state of equilibrium that adapts in response tosignals from the microenvironment.

    The depletion of T or asialo GM1-positive cells was sufficientto awaken disseminated spontaneous metastatic cells fromtheir dormant state. These results demonstrate the capacity ofT or asialo GM1-positive cells in wild-type mice to maintainspontaneousmicrometastases permanently occult in a state ofequilibrium. In BALB/cmice, NK cells and some otherminoritysubpopulations express asialo GM1. We hypothesize that NKcells did not exert a direct cytotoxic effect in our model,because no increase in NK cells was observed during thesystemic or local immune response generated by GR9-B11tumor cells. Furthermore, the depletion of CD8þ T lympho-cytes produced spontaneous metastases in 100% of immuno-competentmice, andGR9-B11–bearing nudemice developed aconsiderable number of spontaneous metastases, despite thelarge amount of NK cells in these hosts. It was previouslyreported that NK cells may facilitate the development of anantitumor protective CTL response independent of CD4þ Tlymphocytes (43–45). Another possibility is that a subpopula-tion of asialo GM1þ CD8þ T cells might be involved incontrolling dormant metastases in this model (46, 47). How-ever, other subpopulations of T cells should also be implicated,because the results found in CD8- versus asialo GM1-depletedtumor-bearing mice were different. In addition, we wouldhighlight that MHC-I molecules can act directly as tumorsuppressor genes, arresting cancer cell proliferation (48), andthe FHIT tumor suppressor gene is directly implicated inMHC-I cell surface expression (30). Taken together, these results

    Romero et al.

    Cancer Res; 74(7) April 1, 2014 Cancer Research1966

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/

  • suggest that the expression of MHC-I molecules on thesemetastatic cells may promote the dormant state via immuno-mediated and oncogenic suppression mechanisms. Futureinvestigations will be designed to clearly decipher the role ofMHC-I molecules in this dormant state and the molecularmechanisms that may be involved.In summary, we present a novel murine metastatic tumor

    model in which the host immune response per se can fullycontrol spontaneous pulmonary metastases, maintainingthem in a state of dormancy. Dormant micrometastases wereawoken after the immunodepletion of T or asialo GM1-positivecells, revealing the major role of these immune cells in main-taining the metastases in a dormant state. This tumor modelresembles the metastatic dormancy observed in some humancancer patients. This preclinicalmetastatic tumormodel offersthe possibility of in-depth investigation of the intrinsic char-acteristics of the premetastatic niche (49) and of the mechan-ismsunderlyingmetastatic dormancy (50), offering newoppor-tunities for immunotherapeutic management of metastaticdisease. These data may help us understand how cancer mightbecome a chronic disease that persists in nonfatal form in aclinically healthy individual.

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

    Authors' ContributionsConception and design: I. Romero, F. Garrido, A.M. Garcia-LoraDevelopment of methodology: I. Romero, C. Garrido, I. Algarra, A. Collado,A.M. Garcia-LoraAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): I. Romero, C. Garrido, I. Algarra, A. ColladoAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): I. Romero, I. Algarra, F. Garrido, A.M. Garcia-LoraWriting, review, and/or revision of the manuscript: I. Romero, F. Garrido,A.M. Garcia-LoraStudy supervision: F. Garrido, A.M. Garcia-Lora

    AcknowledgmentsThe authors thank I. Linares, A.B. Rodriguez, and E. Arias for technical advice

    and R. Davies for editorial assistance.

    Grant SupportThis work was supported by grants cofinanced by FEDER funds (EU) from

    the Instituto de Salud Carlos III (CP03/0111, PI12/02031, PI 08/1265, PI11/01022, RETIC RD 06/020 and RD09/0076/00165), Junta de Andalucía(Group CTS-143, and CTS-695, CTS-3952, CVI-4740 grants), and EuropeanCommunity (LSHC-CT-2004-503306, OJ 2004/c158, 18234). A.M. Garcia Lora.was supported by Miguel Servet Contract CP03/0111 and Contract I3 fromFPS and ISCIII, I. Romero by Rio-Hortega Contract CM12/00033 from ISCIII,and C. Garrido by FPU–MEC 1631.

    The costs of publication of this article were defrayed in part by the paymentof page charges. This article must therefore be hereby marked advertisementin accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

    Received July 24, 2013; revised January 7, 2014; accepted January 28, 2014;published OnlineFirst February 14, 2014.

    References1. Kerkar SP, Restifo NP. Cellular constituents of immune escape within

    the tumor microenvironment. Cancer Res 2012;72:3125–30.2. Shankaran V, Ikeda H, Bruce AT, White JM, Swanson PE, Old LJ, et al.

    IFNgamma and lymphocytes prevent primary tumour developmentand shape tumour immunogenicity. Nature 2001;410:1107–11.

    3. Nagaraj S, SchrumAG,ChoHI,Celis E,GabrilovichDI.Mechanismof Tcell tolerance inducedbymyeloid-derived suppressor cells. J Immunol2010;184:3106–16.

    4. Garrido F, Algarra I. MHC antigens and tumor escape from immunesurveillance. Adv Cancer Res 2001;83:117–58.

    5. Uyttenhove C, Pilotte L, Theate I, Stroobant V, Colau D, Parmentier N,et al. Evidence for a tumoral immune resistance mechanism based ontryptophan degradation by indoleamine 2,3-dioxygenase. Nat Med2003;9:1269–74.

    6. GallinaG,Dolcetti L,SerafiniP,DeSantoC,Marigo I,ColomboMP, etal.Tumors induce a subset of inflammatory monocytes with immunosup-pressive activity on CD8þ T cells. J Clin Invest 2006;116:2777–90.

    7. Seliger B, Cabrera T, Garrido F, Ferrone S. HLA class I antigenabnormalities and immune escape by malignant cells. Semin CancerBiol 2002;12:3–13.

    8. Garcia-Lora A, Algarra I, Garrido F. MHC class I antigens, immunesurveillance, and tumor immune escape. J Cell Physiol 2003;195:346–55.

    9. CarreteroR,WangE,RodriguezAI, Reinboth J, AsciertoML, EngleAM,et al. Regression of melanoma metastases after immunotherapy isassociated with activation of antigen presentation and interferon-mediated rejection genes. Int J Cancer 2012;131:387–95.

    10. Garrido C, Algarra I, Maleno I, Stefanski J, Collado A, Garrido F, et al.AlterationsofHLAclass I expression in humanmelanomaxenografts inimmunodeficient mice occur frequently and are associated with highertumorigenicity. Cancer Immunol Immunother 2010;59:13–26.

    11. Aguirre Ghiso JA. Inhibition of FAK signaling activated by urokinasereceptor induces dormancy in human carcinoma cells in vivo. Onco-gene 2002;21:2513–24.

    12. Muller-Hermelink N, Braumuller H, Pichler B,Wieder T, Mailhammer R,Schaak K, et al. TNFR1 signaling and IFN-gamma signaling determine

    whether T cells induce tumor dormancy or promote multistage carci-nogenesis. Cancer Cell 2008;13:507–18.

    13. KhazaieK, Prifti S, BeckhoveP,GriesbachA,Russell S,CollinsM, et al.Persistence of dormant tumor cells in the bone marrow of tumor cell-vaccinated mice correlates with long-term immunological protection.Proc Natl Acad Sci U S A 1994;91:7430–4.

    14. Weckermann D, Muller P, Wawroschek F, Harzmann R, Riethmuller G,Schlimok G. Disseminated cytokeratin positive tumor cells in the bonemarrow of patients with prostate cancer: detection and prognosticvalue. J Urol 2001;166:699–703.

    15. Meng S, Tripathy D, Frenkel EP, Shete S, Naftalis EZ, Huth JF, et al.Circulating tumor cells in patients with breast cancer dormancy. ClinCancer Res 2004;10:8152–62.

    16. Chen LP, Suzuki Y, LiuCM,Wheelock EF.Maintenance and cure of theL5178Y murine tumor-dormant state by interleukin 2: dependence ofinterleukin 2 on induced interferon-gamma and on tumor necrosisfactor for its antitumor effects. Cancer Res 1990;50:1368–74.

    17. Saudemont A, Quesnel B. In a model of tumor dormancy, long-termpersistent leukemic cells have increased B7-H1 and B7.1 expressionand resist CTL-mediated lysis. Blood 2004;104:2124–33.

    18. Farrar JD, Katz KH, Windsor J, Thrush G, Scheuermann RH, Uhr JW,et al. Cancer dormancy. VII. A regulatory role for CD8þ T cells and IFN-gamma in establishing and maintaining the tumor-dormant state.J Immunol 1999;162:2842–9.

    19. Muller M, Gounari F, Prifti S, Hacker HJ, Schirrmacher V, Khazaie K.EblacZ tumor dormancy in bone marrow and lymph nodes: activecontrol of proliferating tumor cells by CD8þ immune T cells. CancerRes 1998;58:5439–46.

    20. Suzuki Y, Chen LP, Liu CM, Morita T, Wheelock EF. Immuneregulation of the L5178Y murine tumor-dormant state: inductionof cell- and soluble factor-mediated inhibition of tumor cell growthby tumor necrosis factor and gamma-interferon. Cancer Res 1989;49:2028–33.

    21. Sagalowsky AI, Molberg K. Solitary metastasis of renal cell carcinomato the contralateral adrenal gland 22 years after nephrectomy. Urology1999;54:162.

    Spontaneous Metastases in Immune-Mediated Dormancy

    www.aacrjournals.org Cancer Res; 74(7) April 1, 2014 1967

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/

  • 22. Louis DN, Hochberg FH. Cerebral primitive neuroectodermal tumorin an adult, with spinal cord metastasis after 18-year dormancy.J Neurooncol 1990;9:77–80.

    23. Cozar JM, Aptsiauri N, Tallada M, Garrido F, Ruiz-Cabello F. Latepulmonary metastases of renal cell carcinoma immediately after post-transplantation immunosuppressive treatment: a case report. J MedCase Rep 2008;2:111.

    24. Stewart TH, Hollinshead AC, Raman S. Tumour dormancy: initiation,maintenance and termination in animals and humans. Can J Surg1991;34:321–5.

    25. Kauffman HM, McBride MA, Delmonico FL. First report of the UnitedNetwork for Organ Sharing Transplant Tumor Registry: donors with ahistory of cancer. Transplantation 2000;70:1747–51.

    26. Valente M, Furian L, Rigotti P. Organ donors with small renal cancer:report of 3 cases. Transplant Proc 2012;44:1846–7.

    27. MacKie RM, Reid R, Junor B. Fatal melanoma transferred in a donatedkidney 16 years after melanoma surgery. N Engl J Med 2003;348:567–8.

    28. Garrido A, Perez M, Delgado C, Garrido ML, Rojano J, Algarra I, et al.Influence of class I H-2 gene expression on local tumor growth.Description of a model obtained from clones derived from a solidBALB/c tumor. Exp Clin Immunogenet 1986;3:98–110.

    29. Perez M, Algarra I, Ljunggren HG, Caballero A, Mialdea MJ, GaforioJJ, et al. A weakly tumorigenic phenotype with high MHC class-Iexpression is associated with high metastatic potential after sur-gical removal of the primary murine fibrosarcoma. Int J Cancer1990;46:258–61.

    30. Romero I, Martinez M, Garrido C, Collado A, Algarra I, Garrido F, et al.The tumour suppressor Fhit positively regulates MHC class I expres-sion on cancer cells. J Pathol 2012;227:367–79.

    31. Ellis LM, Fidler IJ. Finding the tumor copycat. Therapy fails, patientsdon't. Nat Med 2010;16:974–5.

    32. Talmadge JE, Singh RK, Fidler IJ, Raz A. Murine models to evaluatenovel and conventional therapeutic strategies for cancer. Am J Pathol2007;170:793–804.

    33. Francia G, Cruz-Munoz W, Man S, Xu P, Kerbel RS. Mouse modelsof advanced spontaneous metastasis for experimental therapeutics.Nat Rev Cancer 2011;11:135–41.

    34. Koebel CM, Vermi W, Swann JB, Zerafa N, Rodig SJ, Old LJ, et al.Adaptive immunity maintains occult cancer in an equilibrium state.Nature 2007;450:903–7.

    35. Takeda K, Nakayama M, Sakaki M, Hayakawa Y, Imawari M, Ogasa-wara K, et al. IFN-gamma production by lung NK cells is critical for thenatural resistance to pulmonary metastasis of B16 melanoma in mice.J Leukoc Biol 2011;90:777–85.

    36. Eyles J, Puaux AL, Wang X, Toh B, Prakash C, Hong M, et al. Tumorcells disseminate early, but immunosurveillance limits metastaticoutgrowth, in a mouse model of melanoma. J Clin Invest 2010;120:2030–9.

    37. GarridoC,Romero I, Berruguilla E,CancelaB,Algarra I, ColladoA, et al.Immunotherapy eradicatesmetastaseswith reversible defects inMHCclass I expression. Cancer Immunol Immunother 2011;60:1257–68.

    38. Garcia-Lora A, Algarra I, Gaforio JJ, Ruiz-Cabello F, Garrido F. Immu-noselection by T lymphocytes generates repeated MHC class I-defi-cient metastatic tumor variants. Int J Cancer 2001;91:109–19.

    39. Garcia-Lora A, Martinez M, Algarra I, Gaforio JJ, Garrido F. MHC classI-deficient metastatic tumor variants immunoselected by T lympho-cytes originate from the coordinated downregulation of APM compo-nents. Int J Cancer 2003;106:521–7.

    40. Garrido F, Algarra I, Garcia-Lora AM. The escape of cancer from Tlymphocytes: immunoselection of MHC class I loss variants harboringstructural-irreversible "hard" lesions. Cancer Immunol Immunother2010;59:1601–6.

    41. Domingo-Domenech J, Vidal SJ, Rodriguez-Bravo V, Castillo-MartinM, Quinn SA, Rodriguez-Barrueco R, et al. Suppression of acquireddocetaxel resistance in prostate cancer through depletion of notch-and hedgehog-dependent tumor-initiating cells. Cancer Cell 2012;22:373–88.

    42. Landsberg J, Kohlmeyer J, Renn M, Bald T, Rogava M, Cron M, et al.Melanomas resist T-cell therapy through inflammation-inducedreversible dedifferentiation. Nature 2012;490:412–6.

    43. Xu D, Gu P, Pan PY, Li Q, Sato AI, Chen SH. NK and CD8þ T cell-mediated eradication of poorly immunogenic B16-F10 melanoma bythe combined action of IL-12 gene therapy and 4–1BB costimulation.Int J Cancer 2004;109:499–506.

    44. Mocikat R, Braumuller H, GumyA, Egeter O, Ziegler H, ReuschU, et al.Natural killer cells activated byMHCclass I(low) targets primedendriticcells to induce protective CD8 T cell responses. Immunity 2003;19:561–9.

    45. Adam C, King S, Allgeier T, Braumuller H, Luking C, Mysliwietz J, et al.DC-NKcell cross talk as a novel CD4þT-cell-independent pathway forantitumor CTL induction. Blood 2005;106:338–44.

    46. Trambley J, Bingaman AW, Lin A, Elwood ET, Waitze SY, Ha J, et al.Asialo GM1(þ) CD8(þ) T cells play a critical role in costimulationblockade-resistant allograft rejection. J Clin Invest 1999;104:1715–22.

    47. Moore ML, Chi MH, Goleniewska K, Durbin JE, Peebles RS Jr.Differential regulation of GM1 and asialo-GM1 expression by T cellsand natural killer (NK) cells in respiratory syncytial virus infection. ViralImmunol 2008;21:327–39.

    48. Garrido C, Paco L, Romero I, Berruguilla E, Stefansky J, Collado A,et al. MHC class I molecules act as tumor suppressor genes regulatingthe cell cycle gene expression, invasion and intrinsic tumorigenicity ofmelanoma cells. Carcinogenesis 2012;33:687–93.

    49. Sleeman JP. The metastatic niche and stromal progression. CancerMetastasis Rev 2012;31:429–40.

    50. Kang Y, Pantel K. Tumor cell dissemination: emerging biologicalinsights from animal models and cancer patients. Cancer Cell 2013;23:573–81.

    Romero et al.

    Cancer Res; 74(7) April 1, 2014 Cancer Research1968

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/

  • 2014;74:1958-1968. Published OnlineFirst February 14, 2014.Cancer Res Irene Romero, Cristina Garrido, Ignacio Algarra, et al. DormancyT Lymphocytes Restrain Spontaneous Metastases in Permanent

    Updated version

    10.1158/0008-5472.CAN-13-2084doi:

    Access the most recent version of this article at:

    Cited articles

    http://cancerres.aacrjournals.org/content/74/7/1958.full#ref-list-1

    This article cites 50 articles, 9 of which you can access for free at:

    Citing articles

    http://cancerres.aacrjournals.org/content/74/7/1958.full#related-urls

    This article has been cited by 5 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/74/7/1958To request permission to re-use all or part of this article, use this link

    on June 5, 2021. © 2014 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

    Published OnlineFirst February 14, 2014; DOI: 10.1158/0008-5472.CAN-13-2084

    http://cancerres.aacrjournals.org/lookup/doi/10.1158/0008-5472.CAN-13-2084http://cancerres.aacrjournals.org/content/74/7/1958.full#ref-list-1http://cancerres.aacrjournals.org/content/74/7/1958.full#related-urlshttp://cancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://cancerres.aacrjournals.org/content/74/7/1958http://cancerres.aacrjournals.org/

Recommended