Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Cancer Therapeutics, Targets, and Chemical Biology Research

Expression and Immunotherapeutic Targeting of the SSX Family of Cancer–Testis Antigens in Prostate Cancer

Heath A. Smith, Robert J. Cronk, Joshua M. Lang, and Douglas G. McNeel

Abstract Recent U.S. Food and Drug Administration approval of the first immunotherapy for prostate cancer encourages efforts to improve immune targeting of this disease. The X breakpoint (SSX) comprise a set of cancer–testis antigens that are upregulated in MHC class I–deficient germline cells and in various types of advanced cancers with a poor prognosis. Humoral and cell-mediated immune responses to the SSX family member SSX2 can arise spontaneously in prostate cancer patients. Thus, SSX2 and other proteins of the SSX family may offer useful targets for tumor immunotherapy. In this study, we evaluated the expression of SSX family members in prostate cancer cell lines and tumor biopsies to identify which members might be most appropriate for immune targeting. We found that SSX2 was expressed most frequently in prostate cell lines, but that SSX1 and SSX5 were also expressed after treatment with the DNA demethylating agent 5-aza-20-deoxycytidine. Immunohistochemical analysis of microarrayed tissue biopsies confirmed a differential level of SSX expression in human prostate cancers. Notably, SSX expression in patient tumor samples was restricted to metastatic lesions (5/22; 23%) and no expression was detected in primary prostate tumors examined (0/73; P < 0.001). We determined that cross-reactive immune responses to a dominant HLA-A2–specific SSX epitope (p103-111) could be elicited by immunization of A2/DR1 transgenic mice with SSX vaccines. Our findings suggest that multiple SSX family members are expressed in metastatic prostate cancers which are amenable to simultaneous targeting. Cancer Res; 71(21); 1–11. 2011 AACR.

Introduction Over the course of the last decade, several prostate cancer antigens have been evaluated in clinical trials as immuno- Prostate cancer is the most frequently diagnosed and therapeutic targets for prostate cancer therapy, including second leading cause of cancer-related death among Amer- prostate-specific antigen (5, 6), prostatic acid phosphatase ican men, and a significant health concern worldwide (1). (PAP; refs. 7, 8), PSMA (9), and PSCA (10, 11). Although these Organ-confined prostate cancer is initially treated by surgery protein targets are frequently expressed by prostate cancer or radiation therapy; however, approximately one-third of cells, they are also expressed by normal prostate tissue and patients relapse, and another one-third of these patients will may not be critical for the survival of the tumor; hence, their ultimately develop life-threatening, castrate-resistant tumors expression might be downregulated by cancer cells during the (1–3). Sipuleucel-T was recently U.S. Food and Drug Admin- course of immune targeting or disease progression (12–16). istration approved as the first immunotherapeutic vaccine Thus, there remains a need to identify immune targets of shown to improve overall survival in patients with castrate- prostate cancer that are highly expressed in metastatic disease resistant prostate cancer (4). The success of this approach and/or critical to the progression of the disease, as simulta- suggests that other simpler immunotherapies could be inves- neous antigen targeting may be important to prevent the tigated targeting additional antigens, potentially with the goal outgrowth of escape variants arising during the course of of preventing the development of castrate-resistant metastatic targeted therapy. disease. Cancer–testis antigens (CTA) are one class of tumor- associated antigen upregulated in tumors of different his- tologic origin, including prostate cancer, and especially Authors' Affiliation: Departments of Medicine and Oncology, University prevalent in advanced disease (17–19). Shown in some cases of Wisconsin-Madison, Madison, Wisconsin to be spontaneously immunogenic in cancer patients, these Note: Supplementary data for this article are available at Cancer Research proteins are normally only expressed in germ cells of the Online (http://cancerres.aacrjournals.org/). testis (20–25). Because of the blood–testis barrier, a paucity Corresponding Author: Douglas G. McNeel, Department of Medicine, 7007 Wisconsin Institutes for Medical Research, University of Wisconsin, of antigen-presenting cells, and a lack of MHC molecules on 1111 Highland Ave. Madison, WI 53705. Phone: 608-265-8131; Fax: 608- their surface, proteins exclusively expressed in germline 265-0614; E-mail: [email protected] tissue are considered immune privileged (26, 27). Thus, doi: 10.1158/0008-5472.CAN-11-2127 the ectopic expression of CTA in cancer tissue makes these 2011 American Association for Cancer Research. proteins ideal immunotherapeutic targets. NY-ESO-1 and

www.aacrjournals.org OF1

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Smith et al.

the MAGE family of antigens havebeenthemostextensively PCR, RT-PCR, and quantitative RT-PCR analysis studied CTAs. Proteins of these families have been found to Gradient PCR was carried out to amplify DNA products be preferentially expressed in metastases over primary tu- from SSX image clones encoding SSX1, 2, 3, 4, 5, 6, 7, or 9 mor of various histologic origin and are currently being cDNAs (ATCC), without cross-amplification of other SSX evaluated as target antigens in clinical trials (28, 29). family members. Image clones exclusively encoding SSX8 Another superfamily of CTA thought to be attractive and SSX10 were not commercially available. Amplification targets for cancer immunotherapy are the synovial sarcoma of products with SSX8 and SSX10 primers in cell lines was breakpoint (SSX) proteins (30–32). Although consequently verified by direct DNA sequencing. SSX primers the precise function of these proteins remains unknown, SSX used for PCR analysis were designed directly, or from pub- expression has been associated with stem cell migration, lished sequences (30, 35), and were commercially synthesized suggesting a potential biologically important role to the (Integrated DNA Technologies). PCR conditions were as fol- metastatic phenotype (33). The most investigated member lows: 95C for 1 minute followed by 30 cycles of 95C for 1 SSX2 has been shown to be expressed in prostate cancer minute, the specific annealing temperature for 1 minute, and lesions at the mRNA level (34), whereas expression of other 72C for 3 minutes. A final extension time was 10 minutes at SSX family members has been shown to be inducible with 72C. Products were then separated and evaluated by agarose epigenetic modifying agents (EMA) in colon carcinoma cell gel electrophoresis. lines (35). We have shown that SSX2 expression can be Reverse-transcriptase PCR (RT-PCR) using the One-Step upregulated in prostate cancer cell lines upon treatment RT-PCR kit (Qiagen) was carried out using RNA isolated from with 5-aza-20-deoxycytidine (21). We have also shown that cell lines with the primers and annealing temperatures listed SSX2 antibodies and SSX2 peptide-specific T cells can be in Table 1, as well as b-actin control primers (actinA: 50-TCAT- found in the peripheral blood of some patients with prostate GAAGTGTGACGTTGACATCCGT-30, actinB: 50-CTTAGAAG- cancer, indicating that patients can have preexisting im- CATTTGCGGTGCACGATG-30) under the following PCR mune responses to SSX2 (21, 36). Because the SSX antigens conditions: 50C for 30 minutes, 95C for 15 minutes, 35 are highly immunogenic and essentially tumor specific, cycles at 95C for 1 minute, specific annealing temperature these proteins might be more ideal as targets for immuno- for 1 minute, and 72C for 1 minute; final extension for 10 therapy than other prostate-associated antigens. However, minutes at 72C. For quantitative RT-PCR (qRT-PCR), 1 mg the relative expression of specific SSX family members in RNA was collected and reverse transcribed using qScript prostate cancer cell lines and prostate tumor tissues remains cDNA SuperMix (Quanta Biosciences) according to the man- unknown. In this study, we sought to identify which SSX ufacturer's instructions. qRT-PCR was done using PerfeCTa proteins are expressed in prostate cancer cell lines and SYBR Green SuperMix for iQ (Quanta Biosciences, Inc.) tissues and, therefore, potentially relevant target antigens according to the manufacturer's instructions with 1 mLof for prostate cancer immunotherapy. We then next sought the cDNA synthesis reaction mixed with primers specific for to determine whether it is possible to simultaneously each family member and the SuperMix. Samples were ana- target these SSX family members using immunotherapeutic lyzed using a MyiQ2 Two-Color Real-Time PCR Detection vaccines. System (Bio-Rad) with annealing temperatures as shown in Ct Table 1. All results were analyzed by the 2 D method (37) relative to the ribosomal protein P0 as a control (38): Materials and Methods P0 forward: 50-ACAATGGCAGCATCTACAAC-30; P0 reverse: 50-GCAGACAGACACTGGCAAC-30). Fold induction over vehi- C Cell culture cle treatment was calculated using the 2 DD t method, as Prostate cancer cell lines (LNCaP, 22-RV1, PC3, and DU-145) previously published (37). and noncancer immortalized prostate epithelial cell lines (RWPE-1 and PREC-6) were grown in RPMI 1640 media Western blot supplemented with 10% fetal calf serum (FCS; Invitrogen), Recombinant SSX GST-tagged proteins (SSX1-5; Abnova) or 200 U/mL penicillin/streptomycin, 1 mmol/L sodium pyru- PAP protein (Fitzgerald) were resolved on SDS polyacrylamide vate, and 0.1 mmol/L b-mercaptoethanol. VCaP and LAPC4 gels and transferred to nitrocellulose membranes using stan- cells were grown in Dulbecco's modified Eagle's medium dard procedures. Membranes were probed with an SSX2 supplemented with 5% glucose, 10% FCS, 200 U/mL penicil- monoclonal antibody (mAb; clone 1A4; Abnova) or an SSX5 lin/streptomycin, 1 mmol/L sodium pyruvate, and 0.1 mmol/L polyclonal antibody (B01P; Abnova). b-mercaptoethanol. Where indicated, cells were additionally cultured with 10 mmol/L 5-Aza-20-deoxycytidine (5-aza-dc; Immunohistochemistry Sigma;), 100 nmol/L Trichostatin A (TSA; Sigma), or both Paraffin-embedded tissue sections were washed in xylene agents simultaneously for 72 hours. RNA was collected after and rehydrated in ethanol, followed by antigen retrieval using treatment using the RNeasy Mini kit (Qiagen). Cell lines were Tris-EDTA buffer supplemented with 5 mmol/L CaCl2, 0.5% obtained from American Type Culture Collection (ATCC), Triton X-100 (pH 8.0), and 20 mg/mL Proteinase K. After verified using polyphasic (genotypic and phenotypic) testing washing in PBS/0.1% Tween-20 and blocking in PBS/10% to confirm identity, and passaged in our laboratory for less BSA, slides were stained with primary antibody overnight than 6 months. at 4C. Samples were then stained with goat anti-mouse

OF2 Cancer Res; 71(21) November 1, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Expression of the SSX CTA in Prostate Cancer

Table 1. SSX family member sequence sources and primer designa

Family Accession 50-Primer sequence Product Annealing member number 30Primer sequence size (bp) temperature (C)

SSX1 NM_005635.2 50-CTAAAGCATCAGAGAAGAGAAGC-30 422 60 50-AGATCTCTTATTAATCTTCTCAGAAA-30 SSX2 NM_175698.1 50-GTGCTCAAATACCAGAGAAGATC-30 435 60 50-TTTTGGGTCCAGATCTCTCGTG-30 SSX3 NM_021014.2 50-GGAAGAGTGGGAAAAGATGAAAGT-30 378 65 50-CCCCTTTTGGGTCCAGATATCA-30 SSX4 NM_001034832.2 50-ATGCTCAAATATCAGAGAAGTTACGA-30 590 69 50-GCAAGAAAATACAGCGGAAACGCCA-30 SSX5 NM_175723.1 50-GTTCTCAAATACCAGAGAAGATG-30 437 60 50-CTCTGCTGGCTTCTCGGGCG-30 SSX6 NT_079573.4 50-CTAAAGCATCAGAGAAGAGAAGC-30 198 67 50-TCTGAGAGTCTGTGGCCCGTT-30 SSX7 NM_173358.2 50-TTTGCAAGGAGACCTAGGGC-30 419 65 50-CTTGGTTTTCCTGGAGGGCACAGG-30 SSX8 NR_027250 50-AAAGAGACCCAGGGATGATGA-30 515 65 50-CTCTTCATAAATCACCAGCTGG-30 SSX9 NT_079573.4 50-TTTGCAAGGAGACCTAGGGC-30 419 73 50-GTTGGTTTTCCCGGGGGGCACAGC-30 SSX10 NT_079573.4 50-TTTGCAAGGAGACCCAGGGT-30 438 60 50-TCAGTCCTGGAGCAGGATTCTGCAG-30

aShown are the GenBank accession numbers for each SSX family member evaluated, the primer sequences and annealing temperatures used to amplify specific SSX products in PCR and RT-PCR studies, and the expected product size of each amplified family member transcript.

IgG secondary antibody (Sigma), washed, and stained using maintained in microisolater cages under aseptic conditions, the LSAB System HRP kit (Dako) and DAB metal concentrate and all experimental procedures were conducted under an (Pierce). Slides were then counterstained with hematoxylin, IACUC-approved protocol. mounted with coverslips, and imaged using an Olympus BX51 microscope (Olympus) and SPOT RT analysis software Immunization studies (Diagnostic Instruments). Four- to 6-week-old A2/DR1 mice were immunized subcu- taneously with 100 mg of an SSX peptide or vehicle control in HLA-A2 T2 binding affinity assays complete Freund's adjuvant (Sigma). For DNA vaccination SSX nonamer peptides, derived from the amino acid studies, mice were immunized every 14 days intradermally in sequences of SSX1-9 and corresponding to SSX2 epitope the ear pinna with 100 mg of either plasmid DNA vector control p103-111, were evaluated for their predicted HLA-A2 affinity (pTVG4) or a plasmid DNA vaccine encoding SSX2 (pTVG- using the SYFPEITHI and BIMAS prediction algorithms (39, SSX2; ref. 35). Mice were euthanized 7 (peptide immunization) 40). The purity and identity of synthesized peptides was or 14 (DNA immunization) days after the last immunization, confirmed by mass spectrometry and gas chromatography and splenocytes were isolated by centrifugation after red (United Biochemical Research). In vitro HLA-A2 affinity for blood cell osmotic lysis. each peptide was determined by stabilization of HLA-A2 on TAP-deficient T2 cells as previously described (35, 41). IFNg ELISPOT Peptide-HLA-A2 stabilization was measured as relative IFNg ELISPOT was done according to the manufacturer's mean fluorescent intensity (MFI), normalized to vehicle- instructions (R&D Systems) as previously described (7, 35, 43). pulsed T2 MFI, and all measurements were conducted in Cells were incubated for 48 hours in the presence of 2 mg/mL triplicate. of each individual SSX peptide, influenza matrix protein A peptide, 10 mg/mL concanavalin A (Calbiochem), or media Mice alone, prior to IFNg detection. Spots were counted with an HLA-A2.01/HLA-DR1 expressing, murine MHC class I/II automated plate reader (Autoimmun Diagnostika). The num- knock-out, transgenic (A2/DR1) mice on C57Bl/6 background ber of spots was corrected for the media alone negative were provided by Dr. Francois¸ Lemonnier (42). Mice were control, and reported as the mean number of antigen-specific

www.aacrjournals.org Cancer Res; 71(21) November 1, 2011 OF3

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Smith et al.

IFNg spot-forming units (SFU) per 106 splenocytes from triplicate samples.

Results

Identification of gene-specific primers for ten SSX family members To evaluate mRNA expression of SSX family members, primers and annealing temperatures were first determined that would result in specific product amplification for each known family member. Using the published SSX gene-specific primers and annealing temperatures for PCR (30, 35), we observed cross-amplification of products from image clone DNA plasmids encoding different SSX family member cDNAs, particularly for the SSX4, 6, 7, and 9 primer pairs (data not shown). On the basis of these results, we designed new primers and conducted gradient PCR to establish conditions that would result in complete primer specificity. The primers and anneal- ing temperatures used for these experiments are shown in Table 1, and the location of the published and redesigned primers within the aligned SSX sequences are shown in Sup- plementary Fig. S1. Specific amplification was observed for all SSX primers (Fig. 1). Image clones encoding SSX8 and SSX10 were not available; however, SSX8 and SSX10 primer sets were tested independently for cross-amplification from the available image clones, with no amplification observed (Fig. 1B). These primers were also tested for PCR amplification from a human testis cDNA library. Products of the appropriate size were found for SSX8 but not SSX10 (Fig. 1C). Amplified products Figure 1. PCR amplification of SSX gene products from plasmids of the predicted size for SSX8 were sequenced, confirming expressing specific SSX family members. A, primer sets specific for each primer specificity and SSX8 expression in the testis. SSX family member were tested for PCR amplification using available image clone plasmids encoding different SSX family members as template SSX family members 1, 2, 3, 5, and 8 are expressed or DNA. SSX8 and SSX10 image clones were unavailable. B, SSX8 and SSX10 primer sets were tested against the available SSX image clones. inducible in prostate cell lines C, SSX2, 8, and 10 primer sets were evaluated by PCR amplification from Previous work by our group and others has shown that human testis cDNA to verify specificity of SSX8 and SSX10 primers. Data certain CTA, including SSX2, can be induced in cancer cell lines in each panel is representative of 2 independent experiments. with EMA (21, 35). To determine which SSX members could be potential targets for prostate cancer immunotherapy, 3 human androgen-dependent prostate cancer cell lines (LAPC4, LNCaP, qRT-PCR was conducted with the same RNA samples in all and VCaP), 3 androgen-independent cell lines (22-RV1, PC3, cases in which amplification was observed by RT-PCR. The and DU-145), and 2 immortalized prostate epithelial cell lines expression patterns found by RT-PCR for SSX1, 2, 3, 5, and 8 (RWPE-1 and PREC-E6) were cultured in vitro and treated with were highly similar by qRT-PCR (Fig. 3; SSX3, SSX8 not shown). or without 2 different EMA. Specifically, cells were treated with TSA did not seem to significantly induce expression either a DNA methyltransferase inhibitor, 10 mmol/L 5-aza-dc, a alone or in combination with 5-aza-dc, suggesting that reg- histone deacetylase inhibitor, 100 nmol/L TSA, or both agents ulation of SSX gene expression is predominantly mediated, simultaneously, whereas untreated controls were treated with directly or indirectly, by changes in DNA methylation. Overall, vehicle alone. RNA was collected 72 hours after treatment for these results show that SSX2 is the most commonly expressed RT-PCR analysis (Fig. 2). SSX1 was expressed at baseline only in family member in prostate cancer cell lines, with higher (and untreated PC3 cells but was frequently inducible following more frequent) expression detected relative to other SSX 5-aza-dc treatment. SSX2 was expressed at baseline in most of family members. However, SSX1 and SSX5 expression were the cell lines and was inducible to varying degrees in the frequently induced following treatment with a methylation androgen-independent cells and the normal prostate epithelial inhibitor. Individual cell lines could also express multiple SSX cell lines. In addition, in some instances SSX3, 5, and 8 were family members. inducible with EMA treatment. In the case of SSX8, amplified products were gel purified and sequenced, confirming SSX8 SSX proteins are expressed in human prostate cancer amplification in DU-145, LNCaP, and PREC-E6 cells, which to tissues our knowledge is the first identification of SSX8 expression in We next sought to determine whether the SSX family any cancer cell line or tissue. member proteins are expressed in human prostate tumors,

OF4 Cancer Res; 71(21) November 1, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Expression of the SSX CTA in Prostate Cancer

Figure 2. RT-PCR analysis reveals SSX CTA expression in prostate cancer cell lines and inducible expression of SSX mRNA transcripts with epigenetic modifying agents. Three androgen-dependent and 3 androgen-independent prostate cancer cell lines and 2 immortalized prostate epithelial cell lines were evaluated for expression of SSX1-10 mRNA either in media alone without treatment (NT) or after treatment with 10 mmol/L 5-aza-dc (AZA), 100 nmol/L TSA (TSA), or treatment with both agents simultaneously (AþT). These results are representative of 2 independent studies.

and whether this expression is associated with stage of was used (Fig. 4C and Supplementary Fig. S2). Given the cross- disease. Because the SSX proteins have highly similar amino reactivity of these antibodies, the identification of specific SSX acid sequences, the cross-reactivity to SSX1-5 GST-tagged family members was not possible, but the presence of staining recombinant proteins was evaluated by Western blot. Recom- with 1A4 and not B01P in certain lesions, together with our binant proteins for SSX family members 6 to 10 were not findings in cell lines, suggested that some tissues may express available and were thus not evaluated. We found that a mAb, only SSX2 (among SSX1–SSX5). Staining with B01P, but not 1A4, recognized SSX2 and SSX3, whereas a murine polyclonal 1A4, suggested that other tissues may express one or more of antibody, B01P, recognized SSX1, 3, 4, and 5, but not SSX2 the other SSX family members. In addition, the nuclear SSX (Fig. 4A). These antibodies could also detect SSX protein staining patterns varied among tissues; in some cases SSX expression in paraffin-embedded human testis tissue expression was homogenous, whereas in other tissues the (Fig. 4B). Using a paraffin-embedded tissue microarray, which nuclear staining was punctate, with expression detected in included biopsy samples from 95 patients with varying stages some cells but not adjacent tumor cells. Interestingly, SSX of prostate cancer, 25 patients with high-grade prostatic protein expression among tumors was found exclusively in intraepithelial neoplasia (HGPIN), or benign cores from 72 metastatic lesions (5/22, 23%), and not in primary tumors control patients, we evaluated SSX expression in cancer (0/73, P < 0.001, c2; Table 2). A single benign tissue sample was tissues by immunohistochemistry (IHC). SSX expression scored positive; however, given predominantly cytoplasmic was detected in some of these tissue specimens with differ- staining, this might have been a staining artifact (Supplemen- ential staining observed depending on which SSX antibody tary Fig. S2).

www.aacrjournals.org Cancer Res; 71(21) November 1, 2011 OF5

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Smith et al.

Figure 3. Quantitative analysis of relative expression and fold induction of SSX 1, 2, and 5 mRNA in prostate cancer cell lines treated with epigenetic modifying agents. RNA isolated from prostate cancer cell lines showing SSX expression by RT-PCR was evaluated by qRT-PCR for expression relative to an internal control transcript (P0; A) and specific fold induction (B) of SSX transcripts following EMA treatment. qRT-PCR was done with primers specific for SSX1, 2, and 5, conducted in triplicate, and repeated in an independent experiment. Error bars represent the mean and SD of 6 wells from these 2 experiments. Comparison between groups was made with a 1-way ANOVA followed by post hoc analysis with the Tukey test. *, P < 0.05 compared with vehicle treatment (A) or with TSA treatment (B).

OF6 Cancer Res; 71(21) November 1, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Expression of the SSX CTA in Prostate Cancer

Figure 4. SSX proteins can be detected in tumor biopsy specimens from prostate cancer patients. A, 200 ng of GST-tagged recombinant SSX proteins (SSX1-5) or PAP protein (negative control) were resolved by SDS-PAGE. Cross-recognition of SSX proteins was determined by staining with mAb (1A4) or pAb (B01P) in Western blot. B, these antibodies were also evaluated for their recognition of SSX proteins in paraffin-embedded testis tissue by IHC. An IgG isotype control was used as a primary antibody negative control for staining specificity. C, tissue microarray slides were stained using mAb 1A4 mAb, pAb B01P, or an IgG isotype control. Specimens were considered positive for SSX expression if 2 blinded observers independently scored the cores positive. Metastatic biopsy samples from 3 patients with differential SSX expression are shown (Patient 1: bone metastasis; patient 2: brain metastasis; and patient 3: lymph node metastasis). Positive and negative staining are denoted by "þ" and "", respectively.

T cells specific for epitope p103-111 from SSX1 and determine whether they could be immunologically targeted SSX2 differentially recognize peptide p103-111 from following immunization with 1 SSX family member. To test other SSX proteins this, we focused on SSX2 as the dominant SSX family member, Ultimately, these studies were conducted to identify which with highest expression among prostate cancer cell lines, and SSX family members might be attractive targets for prostate restricted our analysis to a single HLA-A2–restricted epitope, cancer immunotherapy. The results above suggested that SSX2 p103-111 (36). We previously identified that T cells SSX1, 2, and 5, and possibly SSX3 and 8, may be the most specific for this immunodominant epitope can be detected relevant members expressed in prostate tumors. Because in HLA-A2þ patients with advanced prostate cancer and can these proteins are highly homologous, we next wanted to lyse HLA-A2þ SSX2þ prostate cells (36). We therefore wanted

www.aacrjournals.org Cancer Res; 71(21) November 1, 2011 OF7

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Smith et al.

Table 2. SSX proteins are preferentially expressed in metastatic prostate cancer lesionsa

SSX expression

Disease stage 1A4þ B01Pþ 1A4þ B01P 1A4 B01Pþ Total SSX Total tissues

Benign prostate tissue ——1b 1b 48 Benign prostatic hyperplasia ————24 HGPIN ————25 1 Prostate tumor–stage 2 ————42 1 Prostate tumor–stage 3 ————12 1 Prostate tumor–stage 4 ————19 Metastasis 122522

aShown are the numbers of patients in each disease stage with biopsy cores staining positive with either of the 2 SSX antibodies, or both antibodies simultaneously (total SSX) by tissue microarray IHC. Data are representative of 2 independent experiments. bStaining shown in Supplementary Fig. S2).

to assess whether SSX2 p103-111–specific T cells could rec- (34). When SSX6-9 were subsequently identified, it was found ognize the corresponding peptide derived from other relevant that SSX1, 2, 4, 5, and 6 could be induced with EMA treatment SSX family members. Using peptide-binding algorithms to in colon cancer cell lines (35). Prior to our study, a compre- predict HLA-A2 affinity, we found that most of the corre- hensive study evaluating SSX family member mRNA and sponding p103-111 peptides from the different family mem- protein expression had not been conducted for prostate bers were predicted to have moderate HLA-A2 affinity cancer cell lines or tumor tissues. (Fig. 5A). These peptides were then synthesized and evaluated In this study, we have identified primers that specifically for their in vitro HLA-A2 affinity (Fig. 5B). We found that each amplify individual SSX family members. These primers were SSX p103-111 peptide had affinity for HLA-A2, and this binding used to evaluate SSX expression in prostate cancer cell lines by corresponded with the predicted binding algorithm scores. As RT-PCR and qRT-PCR. We found that SSX2 mRNA transcripts a negative control, peptide p103-111 from SSX2 was modified were commonly expressed in prostate cancer cell lines at (SSX2-IP) to abolish HLA-A2 binding. baseline, whereas SSX1 and SSX5 were frequently inducible To evaluate whether T cells specific for SSX2 p103-111 can with EMA. To a lesser extent, SSX3 and SSX8 were also recognize peptides derived from different SSX family mem- induced (4/8 and 3/8 cell lines, respectively). Using SSX bers, we immunized HLA-A2 transgenic (A2/DR1) mice with antibodies that recognize different SSX family members, we an SSX2-encoding DNA plasmid vaccine (36). T cells specific found that tumors can have differential SSX protein expres- for SSX2 p103-111 recognized p103-111 from SSX3/5/9, but no sion, and expression is essentially restricted to metastatic other family members (Fig. 5C), suggesting that an SSX2 lesions. We also found that T cells specific for an HLA-A2– vaccine that elicits SSX2 p103-111 peptide-specific T cells restricted SSX2 immunodominant epitope can recognize the could also be used to target SSX3, 5, or 9. Similarly, T cells corresponding p103-111 peptide derived from other SSX fam- from A2/DR1 mice immunized directly with peptide p103-111 ily members. Cross-reactivity of SSX2 p103-specific T cells from SSX1 or SSX2 were evaluated for cross-reactive immune was observed for the corresponding p103-111 peptide from responses by IFNg ELISPOT (Fig. 5D). Again we found that SSX3/5/9, whereas SSX1 p103-111–specific T cells seem to T cells specific for SSX2 p103-111 could recognize peptide recognize p103 from SSX6/8 and perhaps SSX4 and SSX7. p103-111 from SSX3/5/9, whereas T cells elicited by immuni- In contrast to previous reports, we found that published zation with SSX1 p103-111 cross-recognized peptides p103- primers for SSX4, 6, 7, and 9 led to amplification of other family 111 from SSX4, SSX6/8, and SSX7. Together, these results show member transcripts. In our hands, SSX4 primers previously that multiple SSX family members can be expressed in pros- reported amplified products from SSX2, 3, and 4 cDNA tem- tate cancer and could potentially be targeted simultaneously plates at the annealing temperatures published for SSX4, using immunotherapeutic vaccines. whereas SSX6, 7, and 9 primers cross-amplified products from the SSX7 and SSX9 cDNA templates (data not shown). We also Discussion observed cross-amplification of SSX7 and SSX9 transcripts using the published SSX5 primers. We report here a set of SSX2 mRNA expression has previously been reported in primers and annealing temperatures specific to each family human prostate tumor tissues (21, 34, 44). Initially, Tureci and member that eliminated cross-amplification (Table 1; Supple- colleagues observed SSX2 expression in 5 of 25 prostate cancer mentary Fig. S1). We believe that cross-amplification with the lesions by RT-PCR (44) and later found SSX1, 2, 4, and 5 original SSX4 primers is a significant finding due to the expression in a number of different cancer tissues, however, reported expression of this family member in cell lines and SSX2 was the only member found in prostate cancer lesions tumor tissues in numerous studies. Because we found these

OF8 Cancer Res; 71(21) November 1, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Expression of the SSX CTA in Prostate Cancer

Figure 5. SSX1 and SSX2 p103-111–specific T cells exhibit cross-recognition for other SSX proteins. A, shown are the homologous amino acid sequences for SSX1-9 corresponding to the p103-111 epitope described previously for SSX2 (36). Highlighted amino acids represent nonidentical residues compared with SSX2. Columns 3 and 4 show the predicted HLA-A2 affinity scores for each peptide using the SYFPEITHI (40) and BIMAS (39) prediction algorithms. B, in vitro HLA-A2 affinity of p103-111 peptide from each family member at titrated peptide concentrations by T2 binding assay (MFI). These results are representative of 2 independent experiments. C, splenocytes from individual A2/DR1 mice immunized with either DNA vaccine encoding SSX2 (pTVG-SSX2, n ¼ 9, &) or vector control (pTVG4, n ¼ 3, *) were analyzed to determine the frequency of peptide-specific T cells by IFNg ELISPOT. Splenocytes were stimulated with one of each of the six p103-111 peptides from the different SSX family members, SSX2-IP, influenza matrix A peptide, or a ConA positive control. Each dot represents the frequency of peptide-specific IFNg SFU from an individual mouse, measured in triplicate with background subtracted. D, splenocytes from individual A2/DR1 mice immunized with peptide p103-111 from either SSX1 (n ¼ 5, &) or SSX2 (n ¼ 6, *) or a vehicle control (n ¼ 4, D) were analyzed to determine the frequency of peptide-specific T cells by IFNg ELISPOT as described above.

primers to cross-amplify SSX2, which is the most frequently family member expression. From our studies, the largest reported family member in cancer cell lines and tissues, it induction of transcription was found after DNA methyltrans- could be that some reports of the frequency of SSX4 expres- ferase inhibition with 5-aza-dc treatment, which is not sur- sion in various tumors are inaccurate. prising in light of previous findings that hypomethylation The observed patterns of SSX family member expression events arise early in prostate cancer tumorigenesis (45–47). and induction from our studies suggest underlying epigenetic Although the methylation status of CpG islands have not been marks in different cell lines that mediate responsiveness to reported for the SSX gene family members, whole genome EMA agents. Differences in promoter methylation of CpG bisulfite sequencing data from cancer cell lines is available islands, methylation of intragene cysteines, or alterations in online through the UCSC Genome browser (48). There seem to chromatin folding because of acetylation or methylation of be no documented methylation sites or CpG islands in the histone residues could all contribute to variations in SSX promoter region of SSX2, 3, 4, 6, 7, 8, or 9, even far upstream of

www.aacrjournals.org Cancer Res; 71(21) November 1, 2011 OF9

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Smith et al.

the start site or within intragene regions. Methylated sites are p103 from SSX6/8, SSX4, and SSX7. Interestingly, whereas SSX found in the promoter region of SSX1, and in an intragene antibodies were unable to fully distinguish between family region of SSX5 that could account for differences in baseline members, our T-cell cross-reactivity results show that subtle expression or inducibility. Other regulation sites or mechan- differences in amino acids in the TCR-interacting portion of isms may be present to account for the increased expression of the epitopes conferred specificity for some members and SSX2, 3, and 8 in some cell lines after EMA treatment. In 2 of cross-reactivity to others. These studies do not rule out the the 3 cell lines showing SSX8 inducibility, expression is only possibility that other SSX epitopes could be recognized among present with combined 5-aza-dc and TSA treatment, suggest- family members. Future work with T-cell–directed therapies ing that chromatin remodeling may play a role in transcrip- needs to consider whether SSX1, 2, and 5, as the most tional regulation of this family member. Future studies will expressed SSX family members, will specifically be targeted assess methylation patterns found in SSX at the DNA for prostate cancer. Immune targeting could potentially be level, before and after EMA treatment. accomplished by immunizing with multiple plasmids encod- We also show for the first time SSX protein expression in ing these members, or polytope vaccines could be designed human prostate cancer tissues. We observed different patterns encoding cross-reactive epitopes that will elicit T cells that of SSX family member protein expression in prostate tumors in can specifically recognize peptides from all SSX family mem- terms of member and homogeneity of staining. SSX expression bers relevant to prostate cancer. was found almost exclusively in metastatic prostate cancer Although SSX proteins may be expressed in advanced lesions as compared with primary prostate cancer lesions prostate tumors, it should also be noted that this expression (P < 0.001, c2). Expression identified here in bone, lymph node, may be heterogenous, as has been described for other CTA in and brain metastases suggest that SSX protein expression is various tumor types. This could potentially allow for antigen- inherent to metastatic disease, independent of tissue site. loss variants during immune targeting with a vaccine. Antigen Interestingly, primary tumor biopsies from patients with me- heterogeneity might be overcome with EMA treatment to tastases (stage IV) also were SSX negative, whereas metastatic upregulate antigen expression in tumor tissue, however, it tissues from 5 of 22 patients were SSX positive (P ¼ 0.027, c2), should be noted that these EMA might have deleterious effects further associating SSX expression specifically with metastatic on immune function or may upregulate antigen expression in disease. These results correspond to our previous studies look- normal tissues. Future work will need to assess the feasibility ing at the frequencies of SSX2 p103-111 peptide-specific T cells of combining these two treatments simultaneously to opti- in the peripheral blood mononuclear cells of patients with mize tumor immunotherapy. prostate cancer by tetramer analysis (36). We found that patients with advanced disease had significantly higher fre- Disclosure of Potential Conflicts of Interest quencies of SSX-specific T cells compared with healthy donors and patients with early-stage disease, further suggesting that No potential conflicts of interest were disclosed. these antigens may be associated with advanced prostate Acknowledgments cancer. These findings may or may not suggest a biological importance of SSX expression in metastatic disease progression. The authors thank Dr. Wei Huang for preparation of paraffin-embedded The expression of multiple SSX family members in some tissue and microarray slides used for IHC. metastatic prostate cancer lesions suggests that SSX immu- notherapy may need to consider targeting several SSX proteins Grant Support simultaneously. We have previously identified an immunodo- This work was supported by the Department of Defense Prostate Cancer minant SSX2 epitope (p103-111) that can elicit epitope- Research Program W81XWH-10-1-0495 and W81XWH-08-1-0341. specific T cells in transgenic A2/DR1 mice immunized with The costs of publication of this article were defrayed in part by the payment a plasmid vaccine encoding SSX2 (36). Cross-reactivity of SSX2 of page charges. This article must therefore be hereby marked advertisement in p103-specific T cells from DNA or peptide-immunized mice accordance with 18 U.S.C. Section 1734 solely to indicate this fact. was observed for the corresponding p103-111 peptide from Received June 24, 2011; revised August 23, 2011; accepted August 24, SSX3/5/9, whereas SSX1 p103-111–specific T cells recognized 2011; published OnlineFirst August 31, 2011.

References 1. Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J 5. Kantoff PW, Schuetz TJ, Blumenstein BA, Glode LM, Bilhartz DL, Wyand Clin 2010;60:277–300. M, et al. Overall survival analysis of a phase II randomized controlled 2. Vieweg J. Immunotherapy for advanced prostate cancer. Rev Urol trial of a Poxviral-based PSA-targeted immunotherapy in metastatic 2007;9Suppl 1:S29–38. castration-resistant prostate cancer. J Clin Oncol 2010;28:1099–105. 3. Pound CR, Partin AW, Eisenberger MA, Chan DW, Pearson JD, Walsh 6. Pavlenko M, Roos AK, Lundqvist A, Palmborg A, Miller AM, Ozenci V, PC. Natural history of progression after PSA elevation following et al. A phase I trial of DNA vaccination with a plasmid expressing radical prostatectomy. JAMA 1999;281:1591–7. prostate-specific antigen in patients with hormone-refractory prostate 4. Small EJ, Schellhammer PF, Higano CS, Redfern CH, Nemunaitis JJ, cancer. Br J Cancer 2004;91:688–94. Valone FH, et al. Placebo-controlled phase III trial of immunologic 7. McNeel DG, Dunphy EJ, Davies JG, Frye TP, Johnson LE, Staab MJ, therapy with sipuleucel-T (APC8015) in patients with metastatic, et al. Safety and immunological efficacy of a DNA vaccine encoding asymptomatic hormone refractory prostate cancer. J Clin Oncol prostatic acid phosphatase in patients with stage D0 prostate cancer. 2006;24:3089–94. J Clin Oncol 2009;27:4047–54.

OF10 Cancer Res; 71(21) November 1, 2011 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Expression of the SSX CTA in Prostate Cancer

8. Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, and metastatic malignant melanoma (MM)–correlation with prognos- et al. Sipuleucel-T immunotherapy for castration-resistant prostate tic factors. Cancer Immun 2007;7:11. cancer. N Engl J Med 2010;363:411–22. 29. Roeder C, Schuler-Thurner B, Berchtold S, Vieth G, Driesch P, Schuler 9. Olson WC, Heston WD, Rajasekaran AK. Clinical trials of cancer G, et al. MAGE-A3 is a frequent tumor antigen of metastasized therapies targeting prostate-specific membrane antigen. Rev Recent melanoma. Arch Dermatol Res 2005;296:314–9. Clin Trials 2007;2:182–90. 30. Gure AO, Tureci O, Sahin U, Tsang S, Scanlan MJ, Jager E, et al. SSX: 10. Krupa M, Canamero M, Gomez CE, Najera JL, Gil J, Esteban M. a multigene family with several members transcribed in normal testis Immunization with recombinant DNA and modified vaccinia virus and human cancer. Int J Cancer 1997;72:965–71. Ankara (MVA) vectors delivering PSCA and STEAP1 antigens inhibits 31. Smith HA, McNeel DG. The SSX family of cancer-testis antigens as prostate cancer progression. Vaccine 2011;29:1504–13. target proteins for tumor therapy. Clin Dev Immunol 2010:150591. 11. Thomas-Kaskel AK, Zeiser R, Jochim R, Robbel C, Schultze-See- 32. Taylor BJ, Reiman T, Pittman JA, Keats JJ, de Bruijn DR, Mant MJ, mann W, Waller CF, et al. Vaccination of advanced prostate cancer et al. SSX cancer testis antigens are expressed in most multiple patients with PSCA and PSA peptide-loaded dendritic cells induces myeloma patients: co-expression of SSX1, 2, 4, and 5 correlates with DTH responses that correlate with superior overall survival. Int J adverse prognosis and high frequencies of SSX-positive PCs. J Cancer 2006;119:2428–34. Immunother 2005;28:564–75. 12. Henshall SM, Quinn DI, Lee CS, Head DR, Golovsky D, Brenner PC, 33. Cronwright G, Le Blanc K, Gotherstrom C, Darcy P, Ehnman M, et al. Altered expression of androgen receptor in the malignant Brodin B. Cancer/testis antigen expression in human mesenchymal epithelium and adjacent stroma is associated with early relapse in stem cells: down-regulation of SSX impairs cell migration and prostate cancer. Cancer Res 2001;61:423–7. matrix metalloproteinase 2 expression. Cancer Res 2005;65: 13. Blades RA, Keating PJ, McWilliam LJ, George NJ, Stern PL. Loss of 2207–15. HLA class I expression in prostate cancer: implications for immuno- 34. Tureci O, Chen YT, Sahin U, Gure AO, Zwick C, Villena C, et al. therapy. Urology 1995;46:681–6; discussion 686–7. Expression of SSX genes in human tumors. Int J Cancer 1998;77: 14. Grob BM, Schellhammer PF, Brassil DN, Wright GL Jr. Changes in 19–23. immunohistochemical staining of PSA, PAP, and TURP-27 following 35. Gure AO, Wei IJ, Old LJ, Chen YT. The SSX gene family: character- irradiation therapy for clinically localized prostate cancer. Urology ization of 9 complete genes. Int J Cancer 2002;101:448–53. 1994;44:525–9. 36. Smith HA, McNeel DG. Vaccines targeting the cancer-testis antigen 15. Mhawech-Fauceglia P, Smiraglia DJ, Bshara W, Andrews C, Schwal- SSX-2 elicit HLA-A2 epitope-specific cytolytic T cells. J Immunother ler J, South S, et al. Prostate-specific membrane antigen expression is 2011;34:569–80. a potential prognostic marker in endometrial adenocarcinoma. Cancer 37. Pfaffl MW. A new mathematical model for relative quantification in Epidemiol Biomarkers Prev 2008;17:571–7. real-time RT-PCR. Nucleic Acids Res 2001;29:e45. 16. Reiman JM, Kmieciak M, Manjili MH, Knutson KL. Tumor immunoe- 38. Daud II, Scott ME. Validation of reference genes in cervical cell diting and immunosculpting pathways to cancer progression. Semin samples from human papillomavirus-infected and -uninfected women Cancer Biol 2007;17:275–87. for quantitative reverse transcription-PCR assays. Clin Vaccine Immu- 17. Caballero OL, Chen YT. Cancer/testis (CT) antigens: potential targets nol 2008;15:1369–73. for immunotherapy. Cancer Sci 2009;100:2014–21. 39. Parker KC, Bednarek MA, Coligan JE. Scheme for ranking potential 18. Ghafouri-Fard S, Modarressi MH. Cancer-testis antigens: potential HLA-A2 binding peptides based on independent binding of individual targets for cancer immunotherapy. Arch Iran Med 2009;12:395–404. peptide side-chains. J Immunol 1994;152:163–75. 19. Zendman AJ, Ruiter DJ, Van Muijen GN. Cancer/testis-associated 40. Rammensee H, Bachmann J, Emmerich NP, Bachor OA, Stevanovic genes: identification, expression profile, and putative function. J Cell S. SYFPEITHI: database for MHC ligands and peptide motifs. Immu- Physiol 2003;194:272–88. nogenetics 1999;50:213–9. 20. Bricard G, Bouzourene H, Martinet O, Rimoldi D, Halkic N, Gillet M, 41. Olson BM, McNeel DG. CD8þ T cells specific for the androgen et al. Naturally acquired MAGE-A10- and SSX-2-specific CD8þ T cell receptor are common in patients with prostate cancer and are able responses in patients with hepatocellular carcinoma. J Immunol to lyse prostate tumor cells. Cancer Immunol Immunother 2011;60: 2005;174:1709–16. 781–92. 21. Dubovsky JA, McNeel DG. Inducible expression of a prostate cancer- 42. Pajot A, Michel ML, Fazilleau N, Pancre V, Auriault C, Ojcius DM, et al. testis antigen, SSX-2, following treatment with a DNA methylation A mouse model of human adaptive immune functions: HLA-A2.1-/ inhibitor. Prostate 2007;67:1781–90. HLA-DR1-transgenic H-2 class I-/class II-knockout mice. Eur J Immu- 22. Gnjatic S, Wheeler C, Ebner M, Ritter E, Murray A, Altorki NK, et al. nol 2004;34:3060–9. Seromic analysis of antibody responses in non-small cell lung cancer 43. Olson BM, Frye TP, Johnson LE, Fong L, Knutson KL, Disis ML, et al. patients and healthy donors using conformational protein arrays. J HLA-A2-restricted T-cell epitopes specific for prostatic acid phos- Immunol Methods 2009;341:50–8. phatase. Cancer Immunol Immunother 2010;59:943–53. 23. Niemeyer P, Tureci O, Eberle T, Graf N, Pfreundschuh M, Sahin U. 44. Tureci O, Sahin U, Schobert I, Koslowski M, Scmitt H, Schild HJ, et al. Expression of serologically identified tumor antigens in acute leuke- The SSX-2 gene, which is involved in the t(X;18) translocation of mias. Leuk Res 2003;27:655–60. synovial sarcomas, codes for the human tumor antigen HOM-MEL- 24. Rentzsch C, Kayser S, Stumm S, Watermann I, Walter S, Stevanovic 40. Cancer Res 1996;56:4766–72. S, et al. Evaluation of pre-existent immunity in patients with primary 45. Li LC, Carroll PR, Dahiya R. Epigenetic changes in prostate cancer: breast cancer: molecular and cellular assays to quantify antigen- implication for diagnosis and treatment. J Natl Cancer Inst 2005; specific T lymphocytes in peripheral blood mononuclear cells. Clin 97:103–15. Cancer Res 2003;9:4376–86. 46. Weiss G, Cottrell S, Distler J, Schatz P, Kristiansen G, Ittmann M, et al. 25. Valmori D, Dutoit V, Lienard D, Rimoldi D, Pittet MJ, Champagne P, DNA methylation of the PITX2 gene promoter region is a strong et al. Naturally occurring human lymphocyte antigen-A2 restricted independent prognostic marker of biochemical recurrence in patients CD8þ T-cell response to the cancer testis antigen NY-ESO-1 in with prostate cancer after radical prostatectomy. J Urol 2009;181: melanoma patients. Cancer Res 2000;60:4499–506. 1678–85. 26. Simpson AJ, Caballero OL, Jungbluth A, Chen YT, Old LJ. Cancer/ 47. Dobosy JR, Roberts JL, Fu VX, Jarrard DF. The expanding role testis antigens, gametogenesis and cancer. Nat Rev Cancer of epigenetics in the development, diagnosis and treatment of 2005;5:615–25. prostate cancer and benign prostatic hyperplasia. J Urol 2007;177: 27. Fijak M, Meinhardt A. The testis in immune privilege. Immunol Rev 822–31. 2006;213:66–81. 48. Kent WJ, Sugnet CW, Furey TS, Roskin KM, Pringle TH, Zahler AM, 28. Velazquez EF, Jungbluth AA, Yancovitz M, Gnjatic S, Adams S, O’Neill et al. The browser at UCSC. Genome Res 2002; D, et al. Expression of the cancer/testis antigen NY-ESO-1 in primary 12:996–1006.

www.aacrjournals.org Cancer Res; 71(21) November 1, 2011 OF11

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research. Published OnlineFirst August 31, 2011; DOI: 10.1158/0008-5472.CAN-11-2127

Expression and Immunotherapeutic Targeting of the SSX Family of Cancer−Testis Antigens in Prostate Cancer

Heath A. Smith, Robert J. Cronk, Joshua M. Lang, et al.

Cancer Res Published OnlineFirst August 31, 2011.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-11-2127

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2011/08/31/0008-5472.CAN-11-2127.DC2

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2011/10/14/0008-5472.CAN-11-2127. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2011 American Association for Cancer Research.