Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

Cancer Therapy: Preclinical Clinical Cancer Research PF-03732010: A Fully Human Monoclonal Antibody against P- with Antitumor and Antimetastatic Activity

Cathy C. Zhang1, Zhengming Yan1, Qin Zhang1, Kyle Kuszpit4, Kenneth Zasadny4, Ming Qiu1, Cory L. Painter1, Anthony Wong2, Eugenia Kraynov3, Maria E. Arango1, Pramod P. Mehta1, Ian Popoff1, Gerald F. Casperson5, Gerrit Los1, Steve Bender1, Kenna Anderes1, James G. Christensen1, and Todd VanArsdale1

Abstract Purpose: P-cadherin is a membrane glycoprotein that functionally mediates tumor , proliferation, and invasiveness. We characterized the biological properties of PF-03732010, a human monoclonal antibody against P-cadherin, in cell-based assays and tumor models. Experimental Design: The affinity, selectivity, and cellular inhibitory activity of PF-03732010 were tested in vitro. Multiple orthotopic and metastatic tumor models were used for assessing the antitumor and antimetastatic activities of PF-03732010. Treatment-associated pharmacodynamic changes were also investigated. Results: PF-03732010 selectively inhibits P-cadherin–mediated cell adhesion and aggregation in vitro. In the P-cadherin–overexpressing tumor models, including MDA-MB-231-CDH3, 4T1-CDH3, MDA-MB- 435HAL-CDH3, HCT116, H1650, PC3M-CDH3, and DU145, PF-03732010 inhibited the growth of primary tumors and metastatic progression, as determined by bioluminescence imaging. Computed tomography imaging, H&E stain, and quantitative PCR analysis confirmed the antimetastatic activity of PF-03732010. In contrast, PF-03732010 did not show antitumor and antimetastatic efficacy in the coun- terpart tumor models exhibiting low P-cadherin expression. Mechanistic studies via immunofluorescence, immunohistochemical analyses, and 3′-[18F]fluoro-3′-deoxythymidine–positron emission tomography imaging revealed that PF-03732010 suppressed P-cadherin levels, caused degradation of membrane β-catenin, and concurrently suppressed cytoplasmic vimentin, resulting in diminished metastatic capac- ity. Changes in the levels of Ki67, caspase-3, and 3′-[18F]fluoro-3′-deoxythymidine tracer uptake also indicated antiproliferative activity and increased apoptosis in the tested xenografts. Conclusions: These findings suggest that interrupting the P-cadherin signaling pathway may be a novel therapeutic approach for cancer therapy. PF-03732010 is presently undergoing evaluation in Phase 1 clin- ical trials. Clin Cancer Res; 16(21); 5177–88. ©2010 AACR.

The classic constitute a superfamily of mole- major regulatory mechanism for oncogenic signaling cules that mediate calcium-dependent cell-cell adhesions. pathways (2, 3) that guide cell fate decisions through The intracellular domains of cadherins directly bind to β- the regulation of cell growth, differentiation, motility, catenin and link with cytoskeletal components, providing and survival (4). Several cadherins, such as E-cadherin, the molecular basis for stable cell-cell adhesion (1). The N-cadherin, VE-cadherin, P-cadherin, and cadherin 11, cadherin/catenin signaling pathway also represents a have been implicated in cancer disease progression (5). Among the cadherin families, E-cadherin and N-cadher- Authors' Affiliations: 1Oncology Research Unit, 2Drug Safety Research in are the most highly characterized subgroup of adhesion and Development, and 3Pharmacokinetics, Dynamics and Metabolism, . E-cadherin is ubiquitously expressed throughout Pfizer Global Research and Development, La Jolla Laboratories, San most epithelial tissues and serves as a negative regulator to Diego, California; 4Bio-Imaging Unit, Groton Laboratories, Pfizer Global Research and Development, Groton, Connecticut; and 5Protein functionally block the β-catenin signaling pathway and Therapeutics CoE, Pfizer Global Research and Development, St. Louis suppress tumor cell growth (6, 7) and invasion (8, 9). Nu- Laboratories, St. Louis, Missouri merous preclinical and clinical studies have shown that the Note: Supplementary data for this article are available at Clinical Cancer loss of E-cadherin occurs concurrently with the upregula- Research Online (http://clincancerres.aacrjournals.org/). tion of N-cadherin (10) or other cadherin family members Corresponding Author: Cathy Zhang, Translational Research Group in Oncology Research Unit, Pfizer Global Research and Development, implicated in invasive growth (5). This process, known as 10724 Science Center Rd., San Diego, CA 92121. Phone: 858-622- cadherin switching, has been reported to promote epithe- 3125; Fax: 858-622-5999; E-mail: [email protected]. lial-mesenchymal transition and leads to tumor cell inva- doi: 10.1158/1078-0432.CCR-10-1343 sion and metastasis. N-cadherin overexpression via ©2010 American Association for Cancer Research. cadherin switching was observed in various invasive cancer

www.aacrjournals.org 5177

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

Zhang et al.

uate the antitumor and antimetastatic activity of PF- Translational Relevance 03732010 as well as the underlying mechanism. Noninva- sive bioluminescence imaging (BLI) technology was used – P-cadherin mediated adherens junctions and the as- to longitudinally monitor disease progression. We also used sociated signaling pathway play diverse roles in the 3′-[18F]fluoro-3′-deoxythymidine ([18F]FLT)–positron regulation of tumor cell proliferation, invasiveness, emission tomography imaging (PET) to assess the anti- and metastatic potential. Upregulation of P-cadherin proliferative activity of PF-03732010 and determine the was frequently observed in various malignancies, in- potential clinical utility of this imaging modality as a cluding breast, colon, lung, and pancreatic tumors, surrogate biomarker for proof of mechanism (23). These and P-cadherin increase correlated with poor survival findings provide insights into the P-cadherin signaling of breast cancer patients. PF-03732010, a novel, highly pathway and warrant the clinical investigation of PF- selective human monoclonal antibody against P- 03732010 as a novel agent for anticancer therapy. cadherin, showed antitumor and antimetastatic activity in a diverse panel of P-cadherin–overexpressing tumor Materials and Methods models without introducing any adverse effects. Mech- anistic studies revealed that PF-03732010 disrupted the The PF-03732010 humanized monoclonal antibody β P-cadherin signaling pathway, suppressed -catenin was generated by affinity purification at Pfizer St. Louis transcriptional targets, and resulted in an antimetastatic Therapeutics group (Pfizer, Inc.). D-Luciferin was and antiproliferative activity and the induction of purchased from Caliper Life Sciences. The antibodies apoptosis. Thus, antagonizing P-cadherin with used include anti-β-catenin (BioSource International), PF-03732010 represents a novel approach for antican- anti-Ki67 (Lab Vision), and anti-vimentin (Dako). Anti- cer therapy targeting tumors with high P-cadherin P-cadherin, anti-E-cadherin, anti-N-cadherin, anti- expression. cyclin D1, anti-caspase-3, anti-survivin, and anti-Bcl-2 were purchased from Cell Signaling. Unless otherwise noted, all cell lines and fine chemicals were purchased from American Type Culture Collection and Sigma- cell lines and tumors and, therefore, is emerging as a poten- Aldrich, respectively. tial therapeutic target. ADH-1, a peptide antagonist that disrupts N-cadherin–mediated adhesion, has been shown In vitro selectivity over E-cadherin to inhibit cell growth and motility in vitro, as well as tumor E-cadherin antibody–binding assessment was tested via growth and invasion in vivo (11, 12); ADH1 is currently un- fluorescence-activated cell sorting analysis. A549 cells were dergoing phase 1 clinical trials. blocked with serum and incubated with either 50 μg/mL P-cadherin (CDH3), which shares moderate homology PF-03732010 or an E-cadherin–specific antibody (R&D with N-cadherin (41%) and E-cadherin (46%) in the extra- Systems) at 5 μg/mL for 1 hour. Cells were subsequently cellular domain (13), was often reported to correlate with processed according to the manufacturer's instructions and increased tumor cell motility and invasiveness when over- analyzed using a FACSCalibur system (BD Biosciences). expressed (14–16). P-cadherin–associated tumor cell growth, motility, and invasiveness is likely or at least par- Transfection of tumor cell lines tially mediated by the relocalization of membrane β-cate- DU145, H1650, and HCT116 cell lines, which express nin to the cytoplasm and subsequently into the nucleus high levels of endogenous P-cadherin, were transfected where it partners with the T-cell factor and activates tran- with luciferase expression vector as previously reported scription of the target (14, 17), including vimentin (24). The paired isogenic cell lines, MDA-MB-231-CDH3 (18), cyclin D1, survivin, and BCL-2 (19). The upregula- (P-cadherin)/MDA-MB-231-pCL (vector), PC3M-CDH3/ tion of P-cadherin was frequently observed in various ma- PC3M-pCL (from PC3M-luc-C6, Xenogen-Caliper Compa- lignant tumors, including breast, colon, lung, and ny), and MDA-MB-435HAL-CDH3/MDA-MB-435HAL- pancreatic tumors, and correlated with poor survival of pCL (25, 26), were derived so that they differ only in P- breast cancer patients (15, 20, 21). In contrast, significant- cadherin expression. Full-length human or mouse P-cad- ly low levels of the P-cadherin expression were de- herin cDNAs were cloned into the retroviral transfer plas- tected in a diverse panel of normal tissues (22). Thus, mid pCLNCX (Imgenex), and retroviral vectors were disruption of P-cadherin signaling represents an intriguing prepared in the PT67 packaging cell line (Clontech), ac- opportunity for the development of novel targeted thera- cording to the manufacturer's protocols. 4T1-CDH3 cells peutic agents. were transfected to express high levels of P-cadherin. Tar- PF-03732010, a humanized monoclonal antibody, was get cell lines were transduced with retroviral vectors, and discovered to antagonize P-cadherin–regulated cell-cell drug-resistant pools of cells were selected with geneticin at adhesion and the associated signaling pathway. Because 0.5 mg/mL. DU145, H1650, and HCT116, along with the P-cadherin is implicated in tumor cell invasion, prolifera- MDA-MB-435HAL and PC3M isogenic pairs, stably ex- tion, and metastatic progression, in this report, we used pressed luciferase to allow disease progression to be mon- spontaneous and experimental metastasis models to eval- itored by BLI.

5178 Clin Cancer Res; 16(21) November 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

P-cadherin IgG with Antitumor, Antimetastatic Activity

Cell aggregation and spheroid disruption assays Prostate orthotopic surgical implants were done by in- To perform the aggregation assay, cells were detached jecting5×105 tumor cells into male severe combined and resuspended as single cells at a concentration of 1 × immunodeficient mice. In the SRC model, lung metasta- 106 cells/mL. Cells were plated into 96-well poly-HEMA– ses were visualized at 21 to 28 days after tumor implant. coated plates and incubated with and without the test At later stages of the SRC and experimental metastasis agent(s) at 37°C on a horizontal shaker for 16 hours to disease models, mice exhibited physiologic signs consis- facilitate aggregate formation. For the spheroid disruption tent with advanced metastatic disease, moribund mice experiments, cell aggregates were formed before being were euthanized, and the survival times were recorded treated, and activity was expressed as fold increase in dis- for Kaplan-Meier survival analyses. rupted cells compared with untreated controls. Cells or aggregates were fixed with 4% formaldehyde, BLI and the degree of aggregation was scanned and calculat- In vivo BLI was conducted using the IVIS100 system with ed using a Cellomics ArrayScan (Thermo Fisher Scientif- Living Image acquisition and analysis software (Caliper ic) equipped with Target Acquisition software. Maximal Life Sciences). Anesthetized mice were i.p. injected with inhibition was defined as the value when EGTA was 75 mg/kg D-luciferin and imaged 10 minutes after luciferin present, during which the maximum number of single injection. nonaggregated cells was produced, whereas the negative control was the value when the smallest number of Quantitative PCR and Western blot analysis cell-cell aggregates was formed in complete medium. The circulating tumor cells (CTC) in mouse whole EC50 values were determined using Graphpad Prism blood were evaluated by real-time quantitative PCR software. (qPCR) analysis of human Alu DNA sequences, which has been previously validated by laser scanning cytometry In vivo studies and drug administration (29). Western blot analysis was done according to the All animal experimental procedures complied with the antibody manufacturer's instructions. Tumors were snap- Guide for the Care and Use of Laboratory Animals (Insti- frozen and pulverized in a liquid nitrogen-cooled mortar tute for Laboratory Animal Research, 1996) and were ap- before analysis. proved by the Pfizer Global Research and Development Institutional Animal Care and Use Committee. For phar- Immunofluorescence and immunohistochemical macokinetic analysis, serum samples were collected until staining ready for ELISA analysis. PF-03732010 exhibited a half-life For in vitro immunofluorescence staining, cells were of 5.7 to 6.6 days in mice via s.c. or i.v. dosing (Supple- plated on coverslips in triplicate before treatment. When mentary Fig. S1). The once-weekly s.c. dosing schedule ready, cells were fixed in 4% formaldehyde and permea- was also used in the subsequent in vivo experiments for ef- bilized in 100% methanol before staining. Tumor sam- ficacy and pharmacodynamic end point evaluations. For ples were collected after 4 to 5 weeks of treatment and the efficacy assessments, mice (Charles River) were ran- either frozen in OCT medium (VWR) or prepared into domly assigned to groups (12 mice per group) before formalin-fixed, paraffin-embedded tissue blocks. The treatment, such that the mean tumor size was equal for staining procedure was done according to the manufac- all groups. In studies assessing efficacy, PF-03732010 turer's instructions. Fluorescence images were captured was administered s.c. once per week for 6 to 9 weeks. using a Nikon Eclipse TE2000 fluorescent microscope For all studies in this report, no adverse effects were ob- with Q-Capture software, and image analysis was done served in mice treated with PF-03732010. using Image Pro Plus 5.1 (Media Cybernetics). Quantita- tive analysis of immunohistochemical staining was done Tumor models using the Chromovision automated cell imaging system. Severe combined immunodeficient-beige mice were used for all studies in this report, except when using [18F]FLT-PET imaging 4T1-CDH3 cells, for which normal BALB/c mice were [18F]FLT-PET and computed tomography (CT) imaging used. For the mammary orthotopic implant, 2 × 106 cells was done with a microPET Focus F220 scanner (Siemens were combined with Matrigel and injected into the mice Medical Solutions) and a GE eXplore microCT scanner under the mammary fat pad. Details for the prostate (GE Healthcare), respectively, as described previously orthotopic and subrenal capsule (SRC) tumor models (30). The standardized uptake value (SUV) was calculated were described previously (27, 28). For the H1650 ortho- using the following formula: topic or lung metastasis models, 2 × 106 cells were i.v. injected through the tail vein of anesthetized mice. Tu- SUV ¼ CPETðTÞ=ðID=WÞ; mors in the lungs were identified via bioluminescence within 2 weeks after tumor inoculation. For the HCT116-luc liver metastasis model, mice were intrasple- where CPET is the measured activity in the volume of inter- nically injected with 2 × 103 cells, and liver metastases est, ID is the injected dose (μCi), and W is the mouse body were visualized within a week after tumor cell implant. weight.

www.aacrjournals.org Clin Cancer Res; 16(21) November 1, 2010 5179

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

Zhang et al.

Data analysis To confirm that PF-03732010 does not bind to E- Tumor growth inhibition (TGI) was calculated as 100 × cadherin, cell-based flow cytometric analysis was done (1 − ΔT/ΔC). The ΔC (ΔT) was obtained by subtracting using A549 cells, which exhibit high E-cadherin expres- the mean tumor volume in the vehicle (treated) group sion but no detectable P-cadherin expression. Figure 1A on the first day of treatment from the mean tumor volume shows that binding of PF-03732010 to A549 cells was at the day of assessment. Statistical analyses were con- not detected. ducted using GraphPad Prism with one-way ANOVA anal- ysis, followed by Dunnet's t test to compare the tumor In vitro functional assay of PF-03732010 sizes between the vehicle- and drug-treated groups. P-cadherin expression in the cell lines used in this report was tested by immunoblot analysis (Supplementary Fig. S3). High levels of P-cadherin were observed in Results HCT116, DU145, and H1650, as well as the cell lines engineered to express P-cadherin (CDH3). In contrast, Affinity and specificity of PF-03732010 against vector-transfected (pCL) lines showed minimal expression P-cadherin of P-cadherin. The single-chain Fc versions of PF-03732010 bind to In cell-based functional assays, PF-03732010 signifi- both human and mouse P-cadherin/Fc proteins, with dis- cantly disrupted the MDA-MB-435HAL-CDH3 cell aggre- sociation constant values (Kd) of 2.5 and 1.8 nmol/L, re- gates with an IC50 of 5 nmol/L (Fig. 1B and C). spectively, suggesting similar affinities toward both PF-03732010 also disrupted existing P-cadherin–mediat- proteins. Because the extracellular domain of human and ed adhesion. When the preestablished DU145 spheroids mouse mature P-cadherin proteins share 87% homology were treated for 24 hours, PF-03732010 (>5 nmol/L) ex- (31), PF-03732010 showed comparable cross reactivity hibited an 11-fold to 15-fold increase in disrupted with human and mouse P-cadherins. The selectivity of spheroids when compared with vehicle-treated cells PF-03732010 was tested on immobilized P-cadherin ver- (Fig. 1C, bottom). DU145 cells treated with PF-03732010 sus other related cadherins via an ELISA format (Supple- (>5 nmol/L) for 24 hours showed a reduction in mem- mentary Fig. S2), and PF-03732010 did not exhibit brane-localized β-catenin (Fig. 1D). significant binding to E-cadherin, N-cadherin, or VE- Together, PF-03732010 showed a similar degree of inhi- cadherin recombinant proteins across a broad range of bition of cellular aggregation, disruption of spheroids, and concentrations. suppression of membrane β-catenin levels across a panel

Fig. 1. In vitro characteristics of PF-03732010. A, PF-03732010 does not show nonspecific binding to E-cadherin. B and C, PF-03732010 inhibits MDA-MB- 435HAL-CDH3 cell aggregate formation. C, PF-03732010 disrupts DU145 spheroids. D, PF-03732010 modulates membrane β-catenin level in DU145 cells. Images are at 40× magnification.

5180 Clin Cancer Res; 16(21) November 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

P-cadherin IgG with Antitumor, Antimetastatic Activity

Fig. 2. PF-03732010 exhibits antitumor and antimetastatic properties in multiple P-cadherin–expressing tumor models. Representative BLI images of selected tumor models (A) and the quantitative assessment of the relative tumor sizes in all tested tumor models (B). In each orthotopic model, treatment was initiated when primary tumors were established. In the HCT116 and DU145 metastasis models, PF-03732010 was administered prophylactically at 24 h before tumor cell inoculation, and the TGI was determined during the late stage of the exponential growth period in vehicle-treated mice. *, P < 0.05 versus vehicle group. Tumor sizes in the MDA-MB-231-pCL, MDA-MB-231-CDH3, and 4T1-CDH3 mammary orthotopic models were assessed with calipers; tumors or metastases in other models were quantified with BLI. of P-cadherin–expressing cell lines, including HCT116, In all tested models, treatment of PF-03732010 (s.c. injec- DU145, MDA-MB-231-CDH3, PC3M-CDH3, and MDA- tion) was done once per week until the study ended. MB-435HAL-CDH3. The representative BLI images of selected tumor models (Fig. 2A) and the quantitative measurements of the tumor The antitumor and antimetastatic properties of PF- burdens in all tested models (Fig. 2B) showed the antitu- 03732010 against multiple tumor models, including mor and antimetastatic efficacy of PF-03732010 (20 mg/ mammary, non–small cell lung, colon, and prostate kg) in a panel of orthotopic and metastasis models. cancer models In HCT116 liver metastasis and DU145 lung metastasis The primary tumor sizes in the 4T1-CDH3, MDA-MB- models, PF-03732010 displayed significant (P <0.05) 231-CDH3, and MDA-MB-231-pCL mammary orthotopic antimetastatic activity. PF-03732010 also exhibited signif- models were assessed by caliper measurement. For other icant (P < 0.05) antitumor efficacy against tumor growth tumor models, luciferase-expressing DU145, PC3M-pCL, in 4T1-CDH3, MDA-MB-231-CDH3, DU145, PC3M- PC3M-CDH3, H1650, and HCT116 lines were used to en- CDH3, and H1650 orthotopic models. Dose-dependent able BLI assessment of the primary tumor and metastasis. efficacy was shown in the PC3M-CDH3 orthotopic model.

www.aacrjournals.org Clin Cancer Res; 16(21) November 1, 2010 5181

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

Zhang et al.

PF-03732010 at 5 and 20 mg/kg (data not shown) in- The antitumor and antimetastatic properties of duced 49% and 73% TGI (P < 0.05), respectively. In con- PF-03732010 in MDA-MB-435HAL-CDH3 SRC and trast, in the MDA-MB-231-pCL and PC3M-pCL orthotopic experimental metastasis tumor models models, which expressed low levels of P-cadherin, PF- We next tested the antitumor and antimetastatic proper- 03732010 (20 mg/kg) did not exhibit antitumor activity. ties of PF-03732010 in SRC tumor models using antigen- Because 4T1-CDH3, MDA-MB-231-CDH3, DU145, PC3- expressing MDA-MB-435HAL-CDH3 cells, which form CDH3, H1650, and HCT116 cell lines express high levels spontaneous metastases in lungs. Treatment of PF- of P-cadherin, these results suggest that the efficacy of PF- 03732010 was initiated on day 9 after tumor cell implan- 03732010 was dependent on the blockade of P-cadherin tation, and representative BLI images of the primary and signaling. secondary tumor burdens on day 50 (Fig. 3A) showed In the PC3M-CDH3 tumor-bearing mice, qPCR analysis the antitumor and antimetastatic properties of PF- of the human Alu gene in mouse whole blood (at day 34) 03732010. H&E staining of lungs from a separate cohort showed that PF-03732010 at 5 and 20 mg/kg significantly of five mice in each group (Fig. 3B) indicated a marked reduced the number of human CTCs compared with vehi- reduction of the tumor burden in PF-03732010–treated cle treatment (data not shown). groups compared with vehicle controls, in agreement with

Fig. 3. PF-03732010 displays antitumor and antimetastatic properties and survival benefit in the MDA-MB-435HAL-CDH3 SRC model. Values represent the mean ± SEM from 12 mice. *, P < 0.05 versus vehicle group. The representative BLI images (A), H&E staining of tumor cell infiltration into lungs (B), and qPCR analysis of human CTCs (C) on day 50. Line plots show the dose-dependent inhibition of PF-03732010 against primary tumor growth in SRC (D), tumor burden in lungs (E), and Kaplan-Meier survival analysis using moribund status as the end point.

5182 Clin Cancer Res; 16(21) November 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

P-cadherin IgG with Antitumor, Antimetastatic Activity

the BLI images. In addition, the reduction in the human Alu gene, detected by qPCR analysis, suggested a reduced level of CTCs in mice treated with PF-03732010 (Fig. 3C). Significant inhibition (P < 0.05) of the primary tumor burden was observed at each dose level (Fig. 3D) evaluat- ed at or beyond day 36 after tumor implantation. On day 50, PF-03732010 at 10, 20, and 40 mg/kg showed TGI va- lues of 52%, 66%, and 68%, respectively, against the pri- mary tumor growth. In addition, PF-03732010 showed significant dose-dependent inhibition of tumor growth at secondary organ sites starting on day 28, with TGI va- lues on day 50 of 54%, 73%, and 82% for the 10, 20, and 40 mg/kg–treated groups, respectively (Fig. 3E). The medi- an survival time for mice treated with PF-03732010 at 10, 20, and 40 mg/kg significantly (P < 0.05) increased to 92, 101, and 96 days, respectively, compared with 71 days for the vehicle-treated mice. To further understand how the kinetics of metastasis was mitigated, we tested PF-03732010 in the MDA-MB- 435HAL-CDH3 experimental lung metastasis model by di- rectly injecting cells into the vasculature, which omits the first steps of the metastatic cascade and enables the assess- ment of tumor cell colonization in host organs. PF- 03732010 was administered either 24 hours before tumor cell injection using the prophylactic model or after tumors were established in lungs (at day 14) in the established tu- mor model. Figure 4A depicts the effect of PF-03732010 on tumor burden in the prophylactic setting. PF-03732010 at the 10 and 20 mg/kg dose levels significantly (P < 0.01) inhib- ited tumor cell colonization (80% and 90% inhibition, re- spectively) compared with the vehicle-treated group, and the median survival time significantly (P < 0.01) improved to 87 and 93 days, respectively, over 54 days for the vehi- cle-treated mice (Fig. 4B). In the therapeutic setting, PF- 03732010 significantly inhibited tumor growth (P < 0.01) in lungs at both 10 and 20 mg/kg dose levels with TGI values of 77.2% and 86.8%, respectively (Fig. 4C). Both evaluated dose levels also improved (P < 0.05) the median survival time to 74 days, compared with 55 days for vehicle-treated mice. The overall results in the experimental metastasis model show that PF-03732010 not only inhibits tumor cell col- onization in lungs but also inhibits tumor growth after be- ing established in the host organ.

PF-03732010 negatively modulates P-cadherin and β -catenin levels, results in antiproliferation and Fig. 4. PF-03732010 inhibits tumor cell colonization and lung tumor antimetastatic activities, as well as increased burden growth in the MDA-MB-435HAL-CDH3 experimental metastasis apoptosis model. Values represent the mean ± SEM from 12 mice. In the prophylactic P-cadherin mediates adherens junctions through direct model, the representative BLI images (A) and line plots (B) depict the binding of β-catenin with cadherin cytoplasmic domains. disease progression and the treatment effect. The IgG was also tested in the therapeutic setting (C). The difference in BLI measurements of the In addition to serving a functional role through P-cadherin control groups between B and C was due to interexperimental variability. to stabilize cell-cell adhesion, β-catenin acts as a signal- ing molecule to mediate cell invasion, migration, prolif- At the efficacious doses (10, 20, and 40 mg/kg), PF- eration and survival through the transcription of target 03732010 suppressed the levels of P-cadherin, β-catenin, genes, including vimentin, cyclin D1, survivin, and vimentin, and Ki67 and increased caspase-3 activation, BCL-2. as shown in representative immunofluorescence or

www.aacrjournals.org Clin Cancer Res; 16(21) November 1, 2010 5183

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

Zhang et al.

immunohistochemical images (Fig. 5A) and semiquanti- showed that the degradation of β-catenin induced by PF- tative analysis (Fig. 5B and C) of the biomarkers in the 03732010 (20 mg/kg) was accompanied by a decrease in MDA-MB-435HAL-CDH3 SRC tumor model. These data cyclin D1 levels and the increase in caspase-3 activation, show that the disruption of the P-cadherin/β-catenin sig- indicating that tumor cell antiproliferation and apoptosis naling pathway corresponds with diminished tumor cell were associated with the blockade of β-catenin signaling. invasiveness, proliferation, and increased apoptosis. E- Suppression of vimentin levels was also observed after cadherin levels were below the level of detection in this treatment (not shown). PF-03732010 concurrently modu- model; therefore, we were unable to determine whether lated the levels of survivin and BCL2, suggesting that apo- E-cadherin plays a role in this signaling cascade. ptosis may result from suppression of these antiapoptotic In the PC3M-CDH3 prostate orthotopic model (Fig. proteins. Diminished P-cadherin levels were also observed 5D), Western blot and immunohistochemical analyses after the treatment with PF-03732010, whereas E-cadherin

Fig. 5. PF-03732010 suppresses P-cadherin/β-catenin signaling and reduces levels of β-catenin signaling-mediated target proteins, including vimentin, cyclin D1, survivin, and Bcl2. Values represent the mean ± SEM from six mice. The representative images of tumors treated with vehicle or 20 mg/kg PF-03732010 (A) and semiquantitative analyses (B, C) of P-cadherin, β-catenin, vimentin, Ki67, and caspase-3 staining at all doses in the MDA-MB- 435HAL-CDH3 SRC tumors. Western blot analysis of P-cadherin, cyclin D1, survivin, and Bcl2 levels and the representative immunohistochemical images of β-catenin, E-cadherin, and caspase-3 staining (D) in PC3M-CDH3 tumors. Images of P-cadherin (A), β-catenin (A, D), P-cadherin/β-catenin (A), vimentin (A), and E-cadherin (D) are at 40× magnification. The remaining immunohistochemical images (A, D) are at 10× magnification. All sample analyses were done after mice were treated with vehicle or PF-03732010 for 5 wk.

5184 Clin Cancer Res; 16(21) November 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

P-cadherin IgG with Antitumor, Antimetastatic Activity

Fig. 6. [18F]FLT-PET/CT imaging shows the antiproliferative effect of PF-03732010 in the MDA-MB-435HAL-CDH3 SRC model. Values represent the mean ± SEM from eight mice. *, P < 0.05; **, P < 0.01, between the vehicle and treated groups. The representative images (A) and quantitative analyses (B) of [18F]FLT uptake in the primary tumor (SRC). The representative images (C) and quantitative analyses (D) of [18F]FLT uptake and the CT intensity in the lung tumor burdens. The imaging analysis was done after mice were treated with vehicle or PF-03732010 for 5 wk. expression remained unchanged. PF-03732010 caused ferative activity of PF-03732010 as a downstream effect similar results in the DU145 (Supplementary Fig. S4) of blocking P-cadherin signaling. Similar results were and H1650 orthotopic models. obtained on day 55.

PF-03732010 shows antiproliferative activity via [18F] FLT-PET imaging in the MDA-MB-435HAL-CDH3 SRC Discussion tumor model In MDA-MB-435HAL-CDH3 SRC tumors, PF-03732010 Here, we report the preclinical characteristics of PF- at 10 and 20 mg/kg significantly (P < 0.01) reduced the 03732010, a human monoclonal antibody against uptake of [18F]FLT by 26.8% and 23.4%, respectively, in P-cadherin. PF-03732010 efficiently blocks P-cadherin– primary tumors on day 42 (Fig. 6A and B). The observed mediated cell-cell adhesion by antagonizing the β-catenin reduction in [18F]FLT uptake also occurred in the lung signaling pathway, resulting in significant antiproliferative tumor burden (Fig. 6C and D). PF-03732010 at 10 and and antimetastatic activity. Because the IgG shows no no- 20 mg/kg suppressed [18F]FLT uptake in the lung tumor ticeable signs of adverse effects at the efficacious doses, burden by 31.7% and 37.8% (P < 0.01), respectively, com- antagonizing P-cadherin with PF-03732010 represents a pared with the vehicle treatment, showing the antiproli- novel approach to anticancer therapy.

www.aacrjournals.org Clin Cancer Res; 16(21) November 1, 2010 5185

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

Zhang et al.

PF-03732010 showed high specificity and selectivity for the metastatic cascade, malignant cells break away from P-cadherin targeting. The IgG failed to bind to the most the primary tumor, shed into the bloodstream or migrate closely target-related family members, including E-cadher- through the lymphatic system, and eventually colonize at in, N-cadherin, and VE-cadherin, as assessed by ELISA the preferred organ microenvironment, as proposed in Pa- using recombinant proteins. In PC3M-pCL and MDA-MB- get's “seed and soil” theory (36). PF-03732010 reduced 435HAL-pCL models, which express high level of N-cadher- lymph node metastases via BLI in the PC3M-CDH3 and in but lack P-cadherin, treatment with PF-03732010 DU145 prostate orthotopic models and lowered CTC le- showed no therapeutic benefit. These data suggest that vels in mouse whole blood of PC3M-CDH3 tumor- the IgG-associated efficacy was not due to the off-target bearing mice. The antimetastatic property of the antibody activity by antagonizing N-cadherin. Specifically, PF- was further confirmed in the MDA-MB-435HAL-CDH3 03732010 also lacked the ability to bind to endogenous SRC model; via BLI and CT imaging, as well as H&E stain- E-cadherin in cell-based fluorescence-activated cell sorting ing of lungs, PF-03732010 significantly inhibited tumor analyses, confirming the high selectivity of the antibody cell infiltration into the lungs at all doses tested (10, 20, to P-cadherin over E-cadherin. Because E-cadherin shares and 40 mg/kg). Also using this model, PF-03732010 re- some homology with P-cadherin and E-cadherin function- duced CTC levels in whole blood. Supported by the in vitro ally suppresses tumorigenesis or metastasis (32), the high assay results (data not shown), the ability of PF-03732010 specificity of PF-03732010 for P-cadherin over E-cadherin to suppress CTCs in mouse whole blood could be partially would allow a better therapeutic window to be achieved. caused by antibody-dependent cell-mediated cytotoxicity. PF-03732010 impairs P-cadherin–mediated cell func- Among the millions of cells that primary tumors release tion, and at nanomolar concentrations, the antibody also into the circulation, only a small minority of them will ad- blocked P-cadherin–mediated cell-cell adhesion and dis- here at a distant organ and eventually establish metastases rupted preestablished spheroids. (37). We therefore assessed whether the antimetastatic In tumor models, PF-03732010 induced significant TGI, characteristic of PF-03732010 was entirely due to the re- although this antiproliferative activity was not observed duction of CTCs in whole blood. In the MDA-MB- when cells were treated with PF-03732010 in vitro.This 435HAL-CDH3 prophylactic metastasis model, mice were discrepancy suggests that fully functioning P-cadherin sig- pretreated with PF-03732010 to ensure that plasma levels naling may require the cell-cell and cell-stroma crosstalk in of the IgG were maintained above the effective biological an intact tumor architecture during tumorigenesis and me- concentration when the tumor cells were i.v. injected (24 tastasis, a process that may not be recapitulated under hours posttreatment). PF-03732010 significantly inhibited in vitro conditions. In the experimental system, we used tumor cell adhesion in the lungs, and similar results were P-cadherin–deficient MDA-MB-231, 4T1, PC3M, and observed in the HCT-116 and DU145 experimental metas- MDA-MB-435HAL cells to construct cell lines that stably tasis models. Notably, under the same settings, MDA-MB- overexpress P-cadherin. PF-03732010 showed significant 435HAL-pCL cells did not respond to PF-03732010 in the antitumor activity in the 4T1-CDH3 orthotopic, MDA- prophylactic metastasis model, suggesting that the block- MB-231-CDH3 orthotopic, PC3M-CDH3 orthotopic, and ade of MDA-MB-435HAL-CH3 colonization in the lungs the MDA-MB-435HAL-CDH3 SRC models, as well as in was due to impaired P-cadherin signaling. Overall, the H1650 and DU145 orthotopic models, in which tumor antimetastatic activity of PF-03732010 is attributed to its cells endogenously expressed high levels of P-cadherin. ability to reduce tumor cells in the circulatory system and By comparison, tumor models with minimal P-cadherin mitigate tumor cell colonization in the host organ. expression, including the MDA-MB-231-CDH3 orthoto- P-cadherin knockdown in cells resulted in reduced tu- pic, PC3M-pCL orthotopic, MDA-MB-435HAL-pCL SRC mor cell invasiveness regardless of E-cadherin expression (Supplementary Fig. S5A and B), and the MDA-MB- levels (17). Indeed, loss of P-cadherin expression after 435HAL-pCL experimental metastasis (Supplementary PF-03732010 treatment resulted in decreased metastasis Fig. S5C and D) models, showed no antitumor response without any affect on the E-cadherin level, presumably, to PF-03732010; these results suggest that PF-03732010 in part, due to the reduced tumor cell adhesiveness. More only inflicts efficacy in tumors with high P-cadherin ex- importantly, the antimetastatic activity was derived from pression. P-cadherin is frequently upregulated in a diverse the diminished level of β-catenin, which not only plays panel of malignancies (15, 33, 34) and often associated a functional role in stabilizing P-cadherin–mediated with high histologic grade, invasiveness, and poor survival cell-cell adhesion but also regulates molecular signaling rate (14, 35). The broad spectrum activity of PF-03732010 pathways to promote tumor growth, invasiveness, and me- provides rationale and opportunity in the clinical develop- tastasis (17, 19). It has been shown that P-cadherin failed ment of the antibody for treating patients with tumors to induce invasion when the catenin-binding domain was with high P-cadherin expression. mutated (38). When released from the plasma membrane, Increasing evidence indicates that high P-cadherin ex- β-catenin is stabilized by Wnt signaling and subsequently pression in human cancer cells correlates with increased translocates into the nucleus, where it acts as a coactivator cell motility (15) and invasiveness (20), which led us to with T-cell factor and induces transcriptional activation of investigate whether PF-03732010 slows down metastasis target genes (32, 39). This pathway activation is critical for by blocking the P-cadherin signaling pathway. During epithelial-mesenchymal transition, a mechanism that

5186 Clin Cancer Res; 16(21) November 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

P-cadherin IgG with Antitumor, Antimetastatic Activity

contributes to metastasis (40). During epithelial-mesen- The antiproliferative activity of PF-03732010 was fur- chymal transition, one of the activated targets of β-catenin ther assessed by using the hybrid [18F]FLT-PET/CT imag- signaling is vimentin (18), which has been implicated in ing in the MDA-MB-435HAL-CDH3 SRC model. In tumor cell invasiveness and migratory potential. In several agreement with the results from Ki67 immunohisto- tested xenografts, including MDA-MB-435HAL-CH3 SRC, chemical staining, PF-03732010 significantly (P < 0.05 PC3M-CDH3. and DU145 orthotopic models, efficacious versus vehicle group) suppressed [18F]FLT uptake in doses of PF-03732010 concurrently suppressed membrane both primary and secondary tumors, indicative of anti- β-catenin and vimentin levels and resulted in antimeta- proliferative activity as a downstream effect of blocking static activity, further confirming that the IgG-induced dis- P-cadherin/β-catenin signaling. The results also suggest ruption of β-catenin signaling contributes to antibody that [18F]FLT-PET imaging may be used in early clinical efficacy. In addition, as Bcl-2 overexpression leads to en- development of PF-03732010 as a supplementary surro- hanced metastatic potential in human cancers (41, 42), gate biomarker. Comparing to the conventional biopsy the suppression of Bcl-2 could account for the PF- approach, the imaging technology in some cases pre- 03732010–induced antimetastatic property. The cause of sents advantages due to the noninvasive nature and less the IgG-induced suppression of β-catenin levels is yet to heterogeneity. In the clinical setting, showing the suffi- be fully understood; one possibility is that once β-catenin cient reduction in [18F]FLT uptake in patients on treat- is released into the cytoplasm, it is stabilized by Wnt sig- ment of PF-03732010 at biologically effective doses nalingin acoordinated fashion.Theblockade of P-cadherin/ would constitute proof of mechanism (48). β-catenin signaling disrupts this process and results in the Taken together, these data highlight the critical role of subsequent degradation of β-catenin by nonspecific P-cadherin/β-catenin signaling in regulating tumor cell enzymes (43). metastasis, proliferation, and apoptosis. PF-03732010 β-catenin is well known for controlling a wide array of antagonizes P-cadherin signaling and leads to the antitu- cellular functions. High levels of nuclear β-catenin have mor and antimetastatic efficacy in target-associated tumor been frequently observed in cells with β-catenin or APC models without introducing noticeable adverse effect. This gene mutations (44). We were unable to detect nuclear work provides the rationale and guidance for the clinical β-catenin levels in the cell lines used in this report, includ- development of PF-03732010, in which tumors with high ing MDA-MB-435-CDH3, PC3-CDH3, and DU145, possi- P-cadherin expression will be essential criteria for patient bly due to their functional (wild-type) β-catenin and APC selection. Future work is warranted to seek a refined ther- proteins. One noteworthy observation is that, although apeutic strategy for patient stratification and combination the cellular distribution of β-catenin was primarily de- therapies, aiming to reach the full potential for clinical de- tected on the membrane, nuclear β-catenin remains tran- velopment of the IgG. PF-03732010 is currently under scriptionally active, which is consistent with previous Phase 1 trial development. findings (45). The supporting data are that the decrease in membrane β-catenin by PF-03732010 not only resulted Disclosure of Potential Conflicts of Interest in diminished levels of vimentin (Fig. 5A) but also the re- duction in other β-catenin transcription-mediated target All authors are current or former employees and shareholders of Pfizer, Inc. proteins (Fig. 5D), including cyclin D1, survivin, and Bcl-2 (46, 47). Cyclin D1 mediates tumor cell proliferation Acknowledgments and survival, and Bcl-2 prevents tumor cells from undergo- ing apoptosis. These observations provide evidence that, We thank many Pfizer colleagues for their contributions to this work, including P-cadherin project team, Bioimaging CoE team, Investigational in addition to the reduced metastatic potential, suppres- Pathology in Drug Safety Research and Development, Pfizer Worldwide sing β-catenin levels by PF-03732010 also resulted in anti- Comparative Medicine La Jolla Division, and Protein Therapeutics CoE team. proliferative activity and apoptosis in tumor cells. The The costs of publication of this article were defrayed in part by the β payment of page charges. This article must therefore be hereby marked concurrent decrease in -catenin and Ki67 levels, as well advertisement in accordance with 18 U.S.C. Section 1734 solely to as the increase of caspase-3 staining, further confirmed indicate this fact. the biological relevance of a β-catenin signaling blockade Received 05/18/2010; revised 08/11/2010; accepted 08/16/2010; with the PF-03732010–induced antitumor activity. published OnlineFirst 09/09/2010.

References 1. Wheelock MJ, Knudsen KA, Johnson KR. Membrane-cytoskeleton tissue segregation in relation to malignancy. Int J Dev Biol 2004; interactions with cadherin cell adhesion proteins: roles of catenins 48:397–409. as linker proteins. Curr Top Membr 1996;43:169–85. 5. Wheelock MJ, Shintani Y, Maeda M, Fukumoto Y, Johnson KR. 2. Hajra KM, Fearon ER. Cadherin and catenin alterations in human Cadherin switching. J Cell Sci 2008;121:727–35. cancer. Genes Cancer 2002;34:255–68. 6. Perrais M, Chen X, Perez-Moreno M, Gumbiner BM. E-cadherin 3. Conacci-Sorrell M, Zhurinsky J, Ben-Ze'ev A. The cadherin-catenin homophilic ligation inhibits cell growth and epidermal growth factor adhesion system in signaling and cancer. J Clin Invest 2002;109: receptor signaling independently of other cell interactions. Mol Biol 987–91. Cell 2007;18:2013–25. 4. Foty RA, Steinberg MS. Cadherin-mediated cell-cell adhesion and 7. Stockinger A, Eger A, Wolf J, Beug H, Foisner R. E-cadherin

www.aacrjournals.org Clin Cancer Res; 16(21) November 1, 2010 5187

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

Zhang et al.

regulates cell growth by modulating proliferation-dependent {β}- agents in the MDA-MB-435-HAL-Luc mammary fat pad and subrenal catenin transcriptional activity. J Cell Biol 2001;154:1185–96. capsule tumor models. Clin Cancer Res 2009;15:238–46. 8. Onder TT, Gupta PB, Mani SA, Yang J, Lander ES, Weinberg RA. 29. Nehmann N, Wicklein D, Schumacher U, Müller R. Comparison of Loss of E-cadherin promotes metastasis via multiple downstream two techniques for the screening of human tumor cells in mouse transcriptional pathways. Cancer Res 2008;68:3645–54. blood: quantitative real-time polymerase chain reaction (qRT-PCR) 9. Wong AST, Gumbiner BM. Adhesion-independent mechanism for versus laser scanning cytometry (LSC). Acta Histochemica. In Press suppression of tumor cell invasion by E-cadherin. J Cell Biol 2003; 2009, Corrected Proof. 161:1191–203. 30. Zhang C, Yan Z, Painter CL, et al. PF-00477736 mediates checkpoint 10. Hazan RB, Qiao R, Keren R, Badano I, Suyama K. Cadherin switch in kinase 1 signaling pathway and potentiates docetaxel-induced effi- tumor progression. Ann N Y Acad Sci 2004;1014:155–63. cacy in xenografts. Clin Cancer Res 2009;15:4630–40. 11. Augustine CK, Yoshimoto Y, Gupta M, et al. Targeting N-cadherin 31. Shimoyama Y, Yoshida T, Terada M, Shimosato Y, Abe O, Hirohashi enhances antitumor activity of cytotoxic therapies in melanoma S. Molecular cloning of a human Ca2+-dependent cell-cell adhesion treatment. Cancer Res 2008;68:3777–84. molecule homologous to mouse placental cadherin: its low expres- 12. Mariotti A, Perotti A, Sessa C, Ruegg C. N-cadherin as a therapeutic sion in human placental tissues. J Cell Biol 1989;109:1787–94. target in cancer. Exp Opin Invest Drugs 2007;16:451–65. 32. Jeanes A, Gottardi CJ, Yap AS. Cadherins and cancer: how does 13. Suzuki S, Sano K, Tanihara H. Diversity of the cadherin family: evi- cadherin dysfunction promote tumor progression[quest]. Oncogene dence for eight new cadherins in nervous tissue. Cell Regul 1991;2: 2008;27:6920–9. 261–70. 33. Sanders DSA, Ian P, Robert H, Janusz J. Aberrant P-cadherin ex- 14. Paredes J, Correia AL, Ribeiro AS, Albergaria A, Milanezi F, Schmitt pression is a feature of clonal expansion in the gastrointestinal tract FC. P-cadherin expression in breast cancer: a review. Breast Cancer associated with repair and neoplasia. J Pathol 2000;190:526–30. Res 2007;9:214. 34. Kovacs A, Dhillon J, Walker RA. Expression of P-cadherin, but not 15. Taniuchi K, Nakagawa H, Hosokawa M, et al. Overexpressed P- E-cadherin or N-cadherin, relates to pathological and functional cadherin/CDH3 promotes motility of pancreatic cancer cells by inter- differentiation of breast carcinomas. Mol Pathol 2003;56:318–22. acting with p120ctn and activating rho-family GTPases. Cancer Res 35. Alejandro Peralta S, Karen AK, Hernando S, Aaron CH, Albert AK. 2005;65:3092–9. P-cadherin expression in breast carcinoma indicates poor survival. 16. Ila SP, Pavneesh M, Spiro G, Monique AB, Colin DM. Cadherin Cancer 1999;86:1263–72. switching in ovarian cancer progression. Int J Cancer 2003;106: 36. Fidler IJ. The pathogenesis of cancer metastasis: the “seed and soil” 172–7. hypothesis revisited. Nat Rev Cancer 2003;3:453–8. 17. Cheung LWT, Leung PCK, Wong AST. Cadherin switching and acti- 37. Gupta GP, Massagué J. Cancer metastasis: building a framework. vation of p120 catenin signaling are mediators of gonadotropin- Cell 2006;127:679–95. releasing hormone to promote tumor cell migration and invasion in 38. Paredes J, Stove C, Stove V, et al. P-cadherin is up-regulated by the ovarian cancer. Oncogene 2010;29:2427–40. antiestrogen ICI 182,780 and promotes invasion of human breast 18. Gilles C, Polette M, Mestdagt M, et al. Transactivation of vimentin cancer cells. Cancer Res 2004;64:8309–17. by {β}-catenin in human breast cancer cells. Cancer Res 2003;63: 39. Cheon SS, Cheah AYL, Turley S, et al. Î2-Catenin stabilization dysre- 2658–64. gulates mesenchymal cell proliferation, motility, and invasiveness 19. Tsai Y-P, Yang M-H, Huang C-H, et al. Interaction between HSP60 and causes aggressive fibromatosis and hyperplastic cutaneous and {β}-catenin promotes metastasis. Carcinogenesis 2009;30: wounds. Proc Natl Acad Sci U S A 2002;99:6973–8. 1049–57. 40. Medici D, Hay ED, Olsen BR. Snail and slug promote epithe- 20. Paredes J, Albergaria A, Oliveira JT, Jeronimo C, Milanezi F, Schmitt lial-mesenchymal transition through {β}-catenin-T-cell factor-4- FC. P-cadherin overexpression is an indicator of clinical outcome in dependent expression of transforming growth factor-{β}3. Mol invasive breast carcinomas and is associated with CDH3 promoter Biol Cell 2008;19:4875–87. hypomethylation. Clin Cancer Res 2005;11:5869–77. 41. Pinkas J, Martin SS, Leder P. Bcl-2-mediated cell survival promotes 21. Hardy RG, Tselepis C, Hoyland J, et al. Aberrant P-cadherin expres- metastasis of EpH4 βMEKDD mammary epithelial cells. Mol Cancer sion is an early event in hyperplastic and dysplastic transformation in Res 2004;2:551–6. the colon. Gut 2002;50:513–9. 42. Del Bufalo D, Biroccio A, Leonetti C, Zupi G. Bcl-2 overexpression 22. Imai K, Hirata S, Irie A, et al. Identification of a novel tumor-associated enhances the metastatic potential of a human breast cancer line. antigen, cadherin 3/P-cadherin, as a possible target for immunother- FASEB J 1997;11:947–53. apy of pancreatic, gastric, and colorectal cancers. Clin Cancer Res 43. Maier TJ, Janssen A, Schmidt R, Geisslinger G, Grosch S. Targeting 2008;14:6487–95. the β-catenin/APC pathway: a novel mechanism to explain the cy- 23. Hargreaves RJ. The role of molecular imaging in drug discovery and clooxygenase-2-independent anticarcinogenic effects of celecoxib development. Clin Pharmacol Ther 2008;83:349–53. in human colon carcinoma cells. FASEB J 2005;19:1353–5. 24. Scatena CD, Hepner MA, Oei YA, et al. Imaging of bioluminescent 44. Morin PJ, Sparks AB, Korinek V, et al. Activation of β-catenin-Tcf LNCaP-luc-M6 tumors: a new animal model for the study of meta- signaling in colon cancer by mutations in β-catenin or APC. Science static human prostate cancer. Prostate 2004;59:292–303. 1997;275:1787–90. 25. Schmidt C, Settle S, Keene J, Westlin W, Nickols G, Griggs D. Charac- 45. Torres VA, Tapia JC, Rodriguez DA, et al. E-cadherin is required for terization of spontaneous metastasis in an aggressive breast carcino- caveolin-1-mediated down-regulation of the inhibitor of apoptosis ma model using flow cytometry. Clin Exp Metastasis 1999;17:537–44. protein survivin via reduced β-catenin-Tcf/Lef-dependent transcrip- 26. Murray L, Abrams T, Long K, et al. SU11248 inhibits tumor growth tion. Mol Cell Biol 2007;27:7703–17. and CSF-1R-dependent osteolysis in an experimental breast cancer 46. Kim PJ, Plescia J, Clevers H, Fearon ER, Altieri DC. Survivin and mo- bone metastasis model. Clin Exp Metastasis 2003;20:757–66. lecular pathogenesis of colorectal cancer. Lancet 2003;362:205–9. 27. Jenkins D, Yu S-F, Hornig Y, Purchio T, Contag P. In vivo monitoring 47. Li Q, Dashwood WM, Zhong X, Nakagama H, Dashwood RH. Bcl-2 of tumor relapse and metastasis using bioluminescent PC-3M-luc- overexpression in PhIP-induced colon tumors: cloning of the rat Bcl- C6 cells in murine models of human prostate cancer. Clin Exp Me- 2 promoter and characterization of a pathway involving [β]-catenin, tastasis 2003;20:745–56. c-Myc and E2F1. Oncogene 2007;26:6194–202. 28. Zhang C, Yan Z, Arango ME, Painter CL, Anderes K. Advancing bio- 48. Murphy PS, McCarthy TJ, Dzik-Jurasz ASK. The role of clinical im- luminescence imaging technology for the evaluation of anticancer aging in oncological drug development. Br J Radiol 2008;81:685–92.

5188 Clin Cancer Res; 16(21) November 1, 2010 Clinical Cancer Research

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research. Published OnlineFirst September 9, 2010; DOI: 10.1158/1078-0432.CCR-10-1343

PF-03732010: A Fully Human Monoclonal Antibody against P-Cadherin with Antitumor and Antimetastatic Activity

Cathy C. Zhang, Zhengming Yan, Qin Zhang, et al.

Clin Cancer Res 2010;16:5177-5188. Published OnlineFirst September 9, 2010.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-10-1343

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2014/10/08/1078-0432.CCR-10-1343.DC1

Cited articles This article cites 47 articles, 22 of which you can access for free at: http://clincancerres.aacrjournals.org/content/16/21/5177.full#ref-list-1

Citing articles This article has been cited by 12 HighWire-hosted articles. Access the articles at: http://clincancerres.aacrjournals.org/content/16/21/5177.full#related-urls

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://clincancerres.aacrjournals.org/content/16/21/5177. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 30, 2021. © 2010 American Association for Cancer Research.