9
Therapeutics, Targets, and Chemical Biology Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared Fluorescence Imaging for Image-Guided Surgery of Prostate Cancer Susanne Lutje 1 , Mark Rijpkema 1 , David M. Goldenberg 2 , Catharina M. van Rij 1 , Robert M. Sharkey 2 , William J. McBride 2 , Gerben M. Franssen 1 , Cathelijne Frielink 1 , Wijnand Helfrich 3 , Wim J.G. Oyen 1 , and Otto C. Boerman 1 Abstract Radical removal of malignant lesions may be improved using tumor-targeted dual-modality probes that contain both a radiotracer and a uorescent label to allow for enhanced intraoperative delineation of tumor resection margins. Because pretargeting strategies yield high signal-to-background ratios, we evaluated the feasibility of a pretargeting strategy for intraoperative imaging in prostate cancer using an antiTROP-2 x anti- HSG bispecic antibody (TF12) in conjunction with the dual-labeled diHSG peptide (RDC018) equipped with both a DOTA chelate for radiolabeling purposes and a uorophore (IRdye800CW) to allow near-infrared optical imaging. Nude mice implanted s.c. with TROP-2expressing PC3 human prostate tumor cells or with PC3 metastases in the scapular and suprarenal region were injected i.v. with 1 mg of TF12 and, after 16 hours of tumor accumulation and blood clearance, were subsequently injected with 10 MBq, 0.2 nmol/mouse of either 111 In- RDC018 or 111 In-IMP288 as a control. Two hours after injection, both microSPECT/CT and uorescence images were acquired, both before and after resection of the tumor nodules. After image acquisition, the biodistribution of 111 In-RDC018 and 111 In-IMP288 was determined and tumors were analyzed immunohistochemically. The biodistribution of the dual-label RDC018 showed specic accumulation in the TROP-2expressing PC3 tumors (12.4 3.7% ID/g at 2 hours postinjection), comparable with 111 In-IMP288 (9.1 2.8% ID/g at 2 hours postinjection). MicroSPECT/CT and near-infrared uorescence (NIRF) imaging conrmed this TROP-2specic uptake of the dual-label 111 In-RDC018 in both the s.c. and metastatic growing tumor model. In addition, PC3 metastases could be visualized preoperatively with SPECT/CT and could subsequently be resected by image- guided surgery using intraoperative NIRF imaging, showing the preclinical feasibility of pretargeted dual- modality imaging approach in prostate cancer. Cancer Res; 74(21); 18. Ó2014 AACR. Introduction Despite advances in diagnostic procedures and clinical management, prostate cancer remains associated with signif- icant morbidity and is the second leading cause of cancer- related deaths in men in the Western world. At present, extensive research is focused on the develop- ment of new molecular imaging techniques to improve detec- tion and staging of this disease. In this regard, radiolabeled antibodies that target the prostate cancerassociated cell surface antigens seem to be particularly promising. This is supported by the clinical application of the PSMA-directed antibody capromab pendetide, also known as Prostascint (7E11-C53) or the antibody J591 (1). However, antibodies typically show slow clearance from the circulation (t 1 / 2 ¼ 23 days), which requires a relatively long interval between time of injection of the radiolabeled antibody and subsequent image acquisition to achieve adequate contrast. In addition, the slow blood clearance results in limited target-to-background ratios. To overcome these limitations and to improve targeting of tumors, various pretargeting techniques have been developed. Previously, we reported a study in which we evaluated a pretargeting approach that targets TROP-2expressing pros- tate cancer (2). TROP-2, also known as EGP-1 (epithelial glycoprotein-1), is a 46-kDa transmembrane glycoprotein expressed on diverse carcinomas, such as of the lung, bladder, breast, cervix, ovary, stomach, and prostate (3). We previously applied the trivalent bispecic Ab (bsAb) TF12, which consists of two anti-TROP-2 Fab fragments and one anti-HSG (hista- minesuccinylglycine) Fab fragment. In this strategy, an unlabeled bsAb with afnity for both the tumor cell and a radiolabeled diHSG peptide was injected. After TF12 had accumulated in the tumor and was cleared from the blood, 1 Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, the Netherlands. 2 Immunomedics, Morris Plains, New Jersey. 3 University of Groningen, University Medical Center Groningen, Department of Surgery, Laboratory for Translation Surgical Oncology, Groningen, the Netherlands. Corresponding Author: Susanne Lutje, Department of Radiology and Nuclear Medicine, Radboud University Medical Center, PO Box 9101, 6500 HB Nijmegen, the Netherlands. Phone: 31-24-3615054; Fax: 31- 24-3618942; E-mail: [email protected] doi: 10.1158/0008-5472.CAN-14-0594 Ó2014 American Association for Cancer Research. Cancer Research www.aacrjournals.org OF1 Research. on March 4, 2020. © 2014 American Association for Cancer cancerres.aacrjournals.org Downloaded from Published OnlineFirst September 24, 2014; DOI: 10.1158/0008-5472.CAN-14-0594

Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared ...cancerres.aacrjournals.org/content/canres/early/2014/10/15/0008-5472... · sections. The tissue sections were fixed with

  • Upload
    others

  • View
    8

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared ...cancerres.aacrjournals.org/content/canres/early/2014/10/15/0008-5472... · sections. The tissue sections were fixed with

Therapeutics, Targets, and Chemical Biology

Pretargeted Dual-Modality Immuno-SPECT and Near-Infrared Fluorescence Imaging for Image-Guided Surgery ofProstate Cancer

Susanne L€utje1, Mark Rijpkema1, David M. Goldenberg2, Catharina M. van Rij1, Robert M. Sharkey2,William J. McBride2, Gerben M. Franssen1, Cathelijne Frielink1, Wijnand Helfrich3, Wim J.G. Oyen1, andOtto C. Boerman1

AbstractRadical removal of malignant lesions may be improved using tumor-targeted dual-modality probes that

contain both a radiotracer and a fluorescent label to allow for enhanced intraoperative delineation of tumorresection margins. Because pretargeting strategies yield high signal-to-background ratios, we evaluated thefeasibility of a pretargeting strategy for intraoperative imaging in prostate cancer using an anti–TROP-2 x anti-HSG bispecific antibody (TF12) in conjunction with the dual-labeled diHSG peptide (RDC018) equipped withboth a DOTA chelate for radiolabeling purposes and a fluorophore (IRdye800CW) to allow near-infrared opticalimaging. Nude mice implanted s.c. with TROP-2–expressing PC3 human prostate tumor cells or with PC3metastases in the scapular and suprarenal region were injected i.v. with 1mg of TF12 and, after 16 hours of tumoraccumulation and blood clearance, were subsequently injected with 10 MBq, 0.2 nmol/mouse of either 111In-RDC018 or 111In-IMP288 as a control. Two hours after injection, both microSPECT/CT and fluorescence imageswere acquired, both before and after resection of the tumor nodules. After image acquisition, the biodistributionof 111In-RDC018 and 111In-IMP288 was determined and tumors were analyzed immunohistochemically. Thebiodistribution of the dual-label RDC018 showed specific accumulation in the TROP-2–expressing PC3 tumors(12.4 � 3.7% ID/g at 2 hours postinjection), comparable with 111In-IMP288 (9.1 � 2.8% ID/g at 2 hourspostinjection). MicroSPECT/CT and near-infrared fluorescence (NIRF) imaging confirmed this TROP-2–specificuptake of the dual-label 111In-RDC018 in both the s.c. and metastatic growing tumor model. In addition, PC3metastases could be visualized preoperatively with SPECT/CT and could subsequently be resected by image-guided surgery using intraoperative NIRF imaging, showing the preclinical feasibility of pretargeted dual-modality imaging approach in prostate cancer. Cancer Res; 74(21); 1–8. �2014 AACR.

IntroductionDespite advances in diagnostic procedures and clinical

management, prostate cancer remains associated with signif-icant morbidity and is the second leading cause of cancer-related deaths in men in the Western world.At present, extensive research is focused on the develop-

ment of new molecular imaging techniques to improve detec-tion and staging of this disease. In this regard, radiolabeledantibodies that target the prostate cancer–associated cellsurface antigens seem to be particularly promising. This is

supported by the clinical application of the PSMA-directedantibody capromab pendetide, also known as Prostascint(7E11-C53) or the antibody J591 (1). However, antibodiestypically show slow clearance from the circulation (t1/2 ¼ 2–3 days), which requires a relatively long interval between timeof injection of the radiolabeled antibody and subsequent imageacquisition to achieve adequate contrast. In addition, the slowblood clearance results in limited target-to-background ratios.To overcome these limitations and to improve targeting oftumors, various pretargeting techniques have been developed.Previously, we reported a study in which we evaluated apretargeting approach that targets TROP-2–expressing pros-tate cancer (2). TROP-2, also known as EGP-1 (epithelialglycoprotein-1), is a 46-kDa transmembrane glycoproteinexpressed on diverse carcinomas, such as of the lung, bladder,breast, cervix, ovary, stomach, and prostate (3). We previouslyapplied the trivalent bispecific Ab (bsAb) TF12, which consistsof two anti-TROP-2 Fab fragments and one anti-HSG (hista-mine–succinyl–glycine) Fab fragment. In this strategy, anunlabeled bsAb with affinity for both the tumor cell and aradiolabeled diHSG peptide was injected. After TF12 hadaccumulated in the tumor and was cleared from the blood,

1Department of Radiology and Nuclear Medicine, Radboud UniversityMedical Center, Nijmegen, the Netherlands. 2Immunomedics, MorrisPlains, New Jersey. 3University of Groningen, University Medical CenterGroningen, Department of Surgery, Laboratory for Translation SurgicalOncology, Groningen, the Netherlands.

Corresponding Author: Susanne L€utje, Department of Radiology andNuclear Medicine, Radboud University Medical Center, PO Box 9101,6500 HB Nijmegen, the Netherlands. Phone: 31-24-3615054; Fax: 31-24-3618942; E-mail: [email protected]

doi: 10.1158/0008-5472.CAN-14-0594

�2014 American Association for Cancer Research.

CancerResearch

www.aacrjournals.org OF1

Research. on March 4, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 24, 2014; DOI: 10.1158/0008-5472.CAN-14-0594

Page 2: Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared ...cancerres.aacrjournals.org/content/canres/early/2014/10/15/0008-5472... · sections. The tissue sections were fixed with

the 111In-labeled diHSG peptide IMP288 was administered,which was rapidly and selectively trapped at the tumor. Thismethod allowed for imaging within 1 to 2 hours after injectionof the radiolabeled peptide and higher target-to-backgroundratios thanwith the directly labeled anti-TROP2 antibody hRS7(2, 4).

At present, surgical removal of prostate cancer lesions is thetreatment of choice for patients with low- and intermediate-risk localized disease, and in selected patients with high-risklocalized disease (5). The use of intraoperative imaging mightguide the surgeon in the detection of malignant tissue, whichmight improve the outcome, and reduce morbidity and treat-ment-related side effects. However, the application of nuclearimaging–based pretargeting strategies seems to be limited forintraoperative imaging purposes as tumor tissue cannot bedelineated precisely by radionuclide detection in the intrao-perative setting.

Near-infrared fluorescence (NIRF) imaging with fluoro-phores conjugated to tumor-targeting agents is rapidly emerg-ing as a new sensitive intraoperative imaging technique forimproved and real-time detection of malignant lesions duringsurgery (6–9). However, the penetration depth of both excita-tion and emission light in tissues is usually limited to severalmillimeters (6). One strategy to deal with this limited pene-tration depth is combining NIRF and radionuclide imagingtechniques. With this dual-modality approach, the burden ofthe disease could be assessed preoperatively, followed bylocalization of the tumor lesions intraoperatively using agamma probe, and subsequent intraoperative guidance forsurgical removal of the tumor lesions based on the fluorescentcomponent of the dual-label agent.

Here, we evaluated a bispecific antibody-based pretargetingstrategy for prostate cancer preclinically, using a novel hapten,designated RDC018, a peptide that is based on the extensivelycharacterized tetrapeptide IMP288 with two HSG haptensand a 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid(DOTA) chelate. RDC018 is an IMP288 analog that is C-termi-nally conjugated with the fluorophore IRDye800CW (Fig. 1).The DOTA chelate of RDC018 was labeled with 111In. Thepretargeting characteristics of the TF12/RDC018 approachwere evaluated in a mouse tumor model bearing TROP-2–expressing prostate cancer metastases. In addition, the feasi-bility of this dual-label pretargeting strategy for image-guidedsurgery based on microSPECT/CT and NIRF imaging wasinvestigated.

Materials and MethodsPretargeting reagents TF12 and RDC018

BsAb TF12, having both TROP-2- and HSG-binding specifi-cities, was produced using the dock-and-lock technology asdescribed previously by Rossi and colleagues (10). The NIRfluorescent peptide RDC018 is a peptide-hapten derived fromIMP288, a DOTA-conjugated D-Tyr-D-Lys-D-Glu-D-Lys tetra-peptide, in which both lysine residues are substituted with anHSG moiety via their e-amino group. In addition to the DOTAchelator for radiolabeling, RDC018 also contains a NIR fluo-rescent IRDye800CW moiety (Fig. 1).

Cell cultureThe TROP-2–expressing human prostate cancer cell line

PC3, originally derived from a PC bone metastasis wasobtained from the American Type Culture Collection (CRL1435). Cells were cultured in RPMI-1640 medium, supplemen-tedwith 10% fetal calf serum (Life Technologies) and 2mmol/Lglutamine.

Mouse modelsMale BALB/c nude mice (Janvier), 8- to 9-week-old,

housed in individual ventilated cages (5 mice per cage)under nonsterile standard conditions with free access tostandard animal chow and water, were adapted to labora-tory conditions for 1 week before experimental use. Micewere subcutaneously inoculated with 3� 106 PC3 cells (rightflank) suspended in 200 mL of 67% complete RPMI-1640medium with 33% Matrigel (BD Biosciences). PC3 xenograftsgrew to approximately 0.1 g in 7 days after tumor cellinoculation. To induce metastatic prostate cancer growth,mice were anesthetized with isoflurane and 5� 105 PC3 cellsin 50 mL of PBS were inoculated with a 30-gauge needlethrough the diaphragm into the left cardiac ventricle aftermidline incision (11). PC3 metastases were allowed to growfor 10 to 12 weeks after tumor cell inoculation.

All experiments have been approved by the InstitutionalAnimalWelfare Committee of the RadboudUniversity MedicalCenter and were conducted in accordance to the guidelines ofthe Revised Dutch Act on Animal Experimentation.

Radiolabeling of IMP288 and RDC018IMP288 was labeled with 111In (Covidien) at a specific

activity of 60.5 MBq/nmol under strict metal-free conditions.Briefly, 60 MBq of 111In was added to 2.4 mg of IMP288 in0.1 mol/L MES buffer (pH 5.4; three times the volume of 111In-chloride) during 20 minutes of incubation at 95�C. Followingincubation, 50 mmol/L ethylenediaminetetraacetic acid(EDTA) was added to the labeling reaction to a final concen-tration of 1mmol/L EDTA to chelate unincorporated 111In. Thelabeling efficiency, determined by instant thin-layer chroma-tography on silicagel strips (ITLC-SG; Gelman Sciences) using0.1 mol/L ammonium acetate (NH4Ac) buffer with 0.1 mol/LEDTA (pH, 5.5) as the mobile phase, reached 97%.

RDC018 was radiolabeled under metal-free conditions at aspecific activity of 25.5 MBq/nmol. For this reaction, 127 MBqof 111In was added to 7.2 mg of RDC018 in 0.1 mol/L MES bufferpH 5.4 (three times the volume of 111In-chloride) for 20minutesof incubation at 95�C. For imaging and image-guided surgery,2.5 mg of RDC018 was radiolabeled with 32 MBq (specificactivity, 18.5 MBq/nmol). After incubation, 50 mmol/L EDTAwas added to the labeling reaction to a final concentration of 1mmol/L EDTA. The labeling efficiency for the RDC018 labelingreaction ranged between 87% and 97%.

ImmunoreactivityTo demonstrate bispecific immunoreactivity of TF12, a

serial dilution of PC3 cells in RPMI medium containing 0.5%BSA (1.6� 106 to 2.6� 107 cells in 0.5 mL) was incubated withTF12 (50 mg/mL) for 30 minutes at 37�C. Cells were washed

L€utje et al.

Cancer Res; 74(21) November 1, 2014 Cancer ResearchOF2

Research. on March 4, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 24, 2014; DOI: 10.1158/0008-5472.CAN-14-0594

Page 3: Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared ...cancerres.aacrjournals.org/content/canres/early/2014/10/15/0008-5472... · sections. The tissue sections were fixed with

twice in RPMI medium containing 0.5% BSA followed byincubation with 111In-RDC018 (250 Bq). To determine non-specific binding, a duplicate of the lowest cell concentrationwas incubated without TF12. After incubation, cells werecentrifuged and washed with 500 ml RPMI medium containing0.5% BSA. The activity in the vials and in the cell pellet wasdetermined in the gamma counter (Wizard 300 1480; LKB-Wallac).

Immunohistochemical analysis of TROP-2–expressingtumorsExpression of TROP-2 in subcutaneous PC3 tumors was

determined immunohistochemically on 5-mm frozen tissuesections. The tissue sections were fixed with acetone (100%)for 10 minutes at �20�C. After overnight drying, the sectionswere blocked with 20% normal goat serum (Bodinco) for 30minutes and stained with the anti–TROP-2 humanized mono-clonal antibody hRS7 (4 mg/mL antibody in PBS 1% BSA) for 1hour at room temperature under light-protected conditions.Subsequently, the tissue sections were washed in PBS andstained with a secondary goat-anti-human HRP (Abcam) at adilution of 1:100 in PBS, 1% BSA, for 30 minutes at roomtemperature. After incubation, the tissue sectionswerewashedwith PBS three times. Finally, tumor sections were incubatedwith 0.019 g of 3,3-diaminobenzidine tetrahydro-chloride(DAB) substrate (Bright DAB; Immunologic) in 25 mL of PBSand 62.5 mL of H2O2 for 10 minutes for development, followedby a hematoxylin counterstaining. Immunohistochemicalanalysis of the resected s.c. PC3 tumors revealed TROP-2expression in all tumors (not shown).

Pretargeted immuno-SPECT/CT and NIRF imagingIn the first experiment, male BALB/c nudemice (n¼ 4) with

subcutaneous PC3 xenografts in the right flank were intrave-nously injected into the tail veinwith 1mgof TF12 (6.4 nmol) in200 mL of PBS followed by i.v. injections of either 111In-RDC018[10MBq, 0.6mg (0.2 nmol)] or 111In-IMP288 [10MBq, 0.4mg (0.2nmol)] 16 hours later. Two additional groups of mice (n ¼ 2)did not receive TF12 before injection of the radiolabeledpeptide to determine the non-TF12-mediated localization ofthe peptide. One of these groups received only 111In-RDC018(10 MBq, 0.6 mg/mouse) and one group received only 111In-IMP288 (10 MBq, 0.4 mg/mouse).

Two hours after injection of 111In-RDC018, mice were anes-thetized with isoflurane and imaged on a small-animal micro-SPECT/CT scanner (U-SPECT II; MILabs) with a 1.0-mmdiameter pinhole collimator tube (acquisition time, 30 min-utes) in prone position. After microSPECT/CT imaging, themice were euthanized by O2/CO2 asphyxiation and NIRFimages were acquired on the IVIS Imaging System (acquisitiontime, 5 minutes; binning, medium; Fstop, 2; excitation, 745 nm;excitation autofluorescence, 675 nm; emission, ICG; lamp level,high; FOV, D). MicroSPECT/CT scans were reconstructed withMILabs reconstruction software, which uses an ordered-subsetexpectation maximalization algorithm.

Biodistribution of pretargeted 111In-RDC018 and111In-IMP288

Subsequently, the biodistribution in mice (n ¼ 4) with s.c.PC3 tumors of 111In-RDC018 was compared with that of111In-IMP288. Tissues of interest (tumor, muscle, lung, spleen,

NN

NHNH

HNHN

HN

HN

NHNH

NH

N

NHNH

NH

HN

HN

O

O

O

O

O

OO

N

N N

N

OHO

HO

OH

OH

O

O O

O O

O

OO O

O–

O–

S

O

N

OS

N+

S

SO

O

O

O

O

O

Figure 1. Structural formula of RDC018.

Pretargeted Dual-Modality Imaging of Prostate Cancer

www.aacrjournals.org Cancer Res; 74(21) November 1, 2014 OF3

Research. on March 4, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 24, 2014; DOI: 10.1158/0008-5472.CAN-14-0594

Page 4: Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared ...cancerres.aacrjournals.org/content/canres/early/2014/10/15/0008-5472... · sections. The tissue sections were fixed with

kidney, liver, pancreas, stomach, duodenum, and prostate)were dissected, weighed, and the radioactivity was measuredin a g-counter to determine the biodistribution of 111In-IMP288and 111In-RDC018. Blood samples were obtained by heartpuncture. For calculation of the uptake of radioactivity in eachtissue as a fraction of the injected dose, an aliquot of theinjection dose was counted simultaneously.

Dual-modality microSPECT/CT and NIRF image-guidedsurgery

In the second experiment, male BALB/c nude mice (n¼ 20)with PC3 metastases in the submandibular, shoulder, andadrenal gland region that developed after injection of PC3cells into the right cardiac ventricle were injected intravenous-ly with the bsAb TF12 (1 mg/mouse), followed by i.v. injectionof 111In-RDC018 (0.6 mg/mouse, 20 MBq/mg) at 16 hours afterinjection of TF12. Mice were imaged 2 hours later on both theIVIS Imaging System and a small-animal microSPECT/CTscanner with the same settings as in the first experiment.MicroSPECT/CT scans were performed preoperatively, fol-lowed by NIRF imaging of the mice in the supine positionafter surgical removal of skin, abdominal muscle layers, andperitoneum. After NIRF image acquisition, the visualizedtumor lesions were resected, followed by NIRF imaging tocheck whether residual tumor tissue was left in situ. In addi-tion, microSPECT/CT scans were performed after resection ofthe tumor lesions, using the same scanning parameters thatwere used for preoperative evaluation.

Statistical analysesStatistical analyses were performed with GraphPad Prism,

version 5.03 (GraphPad). Results are presented as mean �

standard deviation (SD). To determine statistical differences,unpaired t tests were used.

ResultsTumor growth and development of metastases

Subcutaneous PC3 tumors in the rightflank reached a tumorsize of approximately 0.1 g at 7 days after tumor cell inocu-lation. To induce metastases, mice (n¼ 20) received injectionsof PC3 cells into the left cardiac ventricle. The mice weredissected after 10 to 12 weeks and soft-tissue PC3 metastaseswere most frequently found in the submandibular, shoulder,and adrenal gland region. In addition, several PC3 metastaseswere found in the rectovesical pouch. Besides the formationof soft-tissue PC3 metastases at different anatomic sites,microCT imaging showed the development of PC3 bonemetastases in the vertebral column (n ¼ 3) and in the ribs(n ¼ 2) of the mice.

Biodistribution of 111In-RDC018 and 111In-IMP288Analysis of the bispecific immunoreactivity of TF12 revealed

that in the in vitro assay up to 82% of the added peptidespecifically bound to the PC3 cells (results not shown).

111In-RDC018 and 111In-IMP288 specifically accumulated inthe s.c. TF12-pretargeted TROP-2–expressing PC3 tumors(right flank). Uptake of 111In-RDC018 and 111In-IMP288 in thePC3 tumor was comparable at 2 hours after injection [12.4 �3.7% ID/g (n ¼ 4) and 9.1 � 2.8% ID/g (n ¼ 4), respectively],Fig. 2A. Tumor uptake of 111In-RDC018 and 111In-IMP288in nonpretargeted tumors was significantly lower, reaching2.1� 0.3% ID/g (P¼ 0.020) and 2.2� 0.0% ID/g (P¼ 0.029) for111In-RDC018 and 111In-IMP288, respectively. Overall, the

Figure 2. A, biodistribution of 111In-RDC018 and 111In-IMP288 in BALB/c nude mice with TF12-pretargeted s.c. PC3 xenografts (right flank) measured at 2hours after injection of the radiolabeled haptens (n ¼ 4). Two groups of mice did not receive TF12 before injection of the radiolabeled haptens to determinenonspecific binding. Tumor uptake for both 111In-RDC018 and 111In-IMP288 was significantly higher in the TF12-pretargeted tumors compared withnonpretargeted tumors. B, representative SPECT/CT (panel 1) and NIRF image (panel 2) of a BALB/c nude mouse with a s.c. PC3 xenograft (right flank)pretargeted with TF12 and 2 hours after injection of 111In-RDC018. In addition to tumor uptake, on the SPECT image also, rest activity originating fromrenal excretion of the hapten can be observed.

L€utje et al.

Cancer Res; 74(21) November 1, 2014 Cancer ResearchOF4

Research. on March 4, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 24, 2014; DOI: 10.1158/0008-5472.CAN-14-0594

Page 5: Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared ...cancerres.aacrjournals.org/content/canres/early/2014/10/15/0008-5472... · sections. The tissue sections were fixed with

biodistribution profiles of 111In-RDC018 and 111In-IMP288show similar patterns (Fig. 2A). One essential difference inthe biodistribution profiles of 111In-RDC018 and 111In-IMP288after pretargeting with TF12 is renal accumulation. Whereasthe accumulation of 111In-IMP288 in the kidneys was relativelylow (2.4 � 0.6% ID/g), renal uptake of 111In-RDC018 wassignificantly higher, reaching 7.6 � 0.9% ID/g at 2 hours afterinjection (P<0.0001). In addition, blood levels of 111In-RDC018were significantly higher compared with 111In-IMP288 [4.7 �0.6% ID/g and 1.7 � 0.9% ID/g (P ¼ 0.0016), respectively],indicating slower blood clearance. Moreover, liver uptake of111In-RDC018 and 111In-IMP288 differed significantly, reaching2.8� 0.3% ID/g for 111In-RDC018 and 1.1� 0.4% ID/g for 111In-IMP288 (P ¼ 0.0006).Tumor-to-blood ratios of 111In-RDC018 and 111In-IMP288

in the TF12-pretargeted tumors were 2.6 � 0.7 and 5.9 � 2.0(P ¼ 0.021), respectively.

Pretargeted immuno-SPECT/CT and NIRF imagingFive BALB/c nude mice with s.c. TROP-2–expressing PC3

tumors (rightflank)were imaged by bothfluorescence imagingandmicroSPECT/CT at 2 hours after injection of 111In-RDC018(18 hours after pretargeting with TF12). A typical set ofmicroSPECT/CT images with corresponding fluorescenceimages of mice in prone and left lateral positions is shownin Fig. 2B. PC3 xenografts were clearly and specifically visual-ized with both imaging modalities.

Dual-modality microSPECT/CT and NIRF image-guidedsurgeryBALB/c nude mice with PC3 metastases were imaged by

both fluorescence imaging and microSPECT/CT at 2 hoursafter injection of radiolabeled RCD018. A typical example of a

microSPECT/CT image with corresponding NIRF image of amouse with two bone metastases in the vertebral column isshown in Fig. 3. Another example of dual-modality micro-SPECT/CT and NIRF imaging of a mouse with TF12-pretar-geted bone metastases originating from the ribs is depictedin Fig. 4.

After preoperative evaluation of tumor lesions by micro-SPECT/CT, image-guided surgery of the tumor lesions wasperformed. First, the exact tumor location was identified byNIRF imaging. Subsequently, these tumor lesions wereresected and NIRF andmicroSPECT/CT imaging was repeatedto ensure radical surgical resection. In Fig. 5, the feasibility ofthe dual-modality probe for intraoperative image-guided sur-gical resection of the metastatic tumor nodules is shown.

DiscussionIn the present preclinical study, the in vivo tumor-targeting

characteristics were evaluated of the dual-modality fluores-cent 111In-labeled hapten RDC018 in TROP-2–expressing pros-tate cancer pretargeted with the trivalent bsAb TF12. Thefeasibility of this approach for image-guided surgery wasdemonstrated inmice bearing TROP-2–expressingmetastases.

In a previous study, the potential for imaging pretargetedprostate cancer with the bsAb TF12 and the radiolabeledhapten IMP288was evaluated (2). Inmice with s.c. PC3 tumors,it was shown that this pretargeting approach allows for rapidand sensitive imaging of TROP-2–expressing prostate cancer.For intraoperative imaging purposes, radionuclide detectioncan be used to localize tumor nodules with a gamma probe;however, precise delineation of tumor lesions or assessment oftumor cell–containing surgical margins remains challenging.NIRF imaging is a sensitive technique that may be exploited to

Figure 3. Sequentially acquireddual-modality microSPECT/CTand NIRF image with 111In-RDC018 in amousewith two TF12-pretargeted tumors. PC3 bonemetastases located in twovertebrae with soft tissueoutgrowth were visualized 2 hoursafter injection of 111In-RDC018. A,CT scan illustrating the alterationsin bone structure in the twovertebrae. B, microSPECT/CTscan showing uptake in the bonemetastases with soft tissueoutgrowth, aswell as accumulationin kidneys and bladder due to renalexcretion of 111In-RDC018. C,corresponding NIRF imageshowing specific uptake in bothPC3 metastases and kidneys.

Pretargeted Dual-Modality Imaging of Prostate Cancer

www.aacrjournals.org Cancer Res; 74(21) November 1, 2014 OF5

Research. on March 4, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 24, 2014; DOI: 10.1158/0008-5472.CAN-14-0594

Page 6: Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared ...cancerres.aacrjournals.org/content/canres/early/2014/10/15/0008-5472... · sections. The tissue sections were fixed with

improve visualization of small to microscopic malignantlesions and potentially of positive resection margins duringsurgery. So far, several preclinical studies have evaluatedmethods to intraoperatively visualize tumor tissue and resec-tion margins in prostate cancer (12–14). Targeted approachesusing antibodies or peptides are particularly promising toapply dual-modality imaging. Very recently, the first clinicaltrial using the anti-VEGF antibody bevacizumab tagged withthe NIRF dye IRDye800CW (clinicaltrials.gov, NCT01508572)has begun, with the purpose to show the feasibility of tumor-targeted intraoperative fluorescence imaging in patients withbreast cancer.

Because the penetration depth of emitted light in tissue islimited to several millimeters, the combination with a radio-label would allow intraoperative probe-guided tumor detec-tion and image-guided surgery.

In the present study, analysis of biodistribution of 111In-RDC018 and 111In-IMP288 showed that the addition of theIRDye800CWfluorophore to the IMP288diHSGpeptide haptenonly slightly affected the in vivo behavior of the molecule. Theoverall biodistribution profile of both peptides was similar;however, RDC018 showed enhanced hepatic uptake. Previous-ly, this has also been observed for IRDye800CW-conjugatedantibodies (15) andmay be due to the increased lipophilicity ofRDC018 compared with IMP288.

Biodistribution studies demonstrated high and specific PC3tumor targeting for both 111In-RDC018 as well as 111In-IMP288.Using this subcutaneous tumor model, we provide proof-of-principle that 111In-RDC018 can be used for rapid dual-modal-ity imaging of TROP-2–expressing tumors. As early as 2 hoursafter injection, high tumor-to-background ratios wereachieved. With both imaging modalities, the fluorescent111In-labeled hapten-peptide RDC018 accumulated specificallyin the TROP-2–expressing PC3 tumors, whereas no specifictumor targeting with 111In-RDC018 was observed in mice withsubcutaneous tumors that were not pretargeted with TF12.Accordingly, the nonpretargeted PC3 tumors were not visual-ized by NIRF imaging. These results clearly demonstrate thatthe accumulation of 111In-RDC018 is TF12mediated. While theuptake of 111In-RDC018 in other tissues remained low, inter-mediate accumulation in the kidneys was observed, which ismost likely caused by renal clearance of the peptide. Theenhanced renal retention of RDC018 compared with that ofIMP288 suggests enhanced tubular reabsorption of the dual-labeled peptide in the kidneys, which may be due to thepresence of the IRDye800CW moiety in RDC018. Similarly,blood levels of RDC018 are higher than those obtained withIMP288 in the same model, most likely because theIRDye800CW moiety of the RDC018 peptide caused theenhanced residence time in the circulation. A clearing agent

Figure 4. Sequentially acquireddual-modality microSPECT/CTand NIRF image with 111In-RDC018 (12MBq, 0.5mg/mouse) ina mouse with two TF12-pretargeted PC3 bone metastasesoriginating from the ribs. Imageswere acquired 2 hours afterinjection of 111In-RDC018. A,microSPECT/CT scan showingkidneys, bladder, and two tumormanifestations (green and bluearrow). B, photographs of exposedmetastases. C, correspondingNIRF images showing specificuptake of 111In-RDC018 in bothPC3 metastases.

L€utje et al.

Cancer Res; 74(21) November 1, 2014 Cancer ResearchOF6

Research. on March 4, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 24, 2014; DOI: 10.1158/0008-5472.CAN-14-0594

Page 7: Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared ...cancerres.aacrjournals.org/content/canres/early/2014/10/15/0008-5472... · sections. The tissue sections were fixed with

may not help in this case, as it is unlikely that the enhancedblood levels are due to complex formation of the peptide withTF12 in the circulation, because the affinity of TF12 for theHSGhapten is the same for IMP288 and RDC018.High accumulation of small-sized fluorescent tracers has

been observed previously. Banerjee and colleagues synthesizeda dual-label PSMA ligand (based on glutamate urea), whichwastagged with both 111In and IRDye800CW for targeting PSMA-expressing prostate cancer in mice with subcutaneous xeno-grafts (16). In this study, high tumor uptake in the PSMA-expressing PC3-PIP xenografts was observed, whereas uptakein the PSMA-negative PC3-FLU tumors remained low. Bothtumor cell lines were derived from PC3 human prostate cancercells and only differ in their expression of PSMA (PIP: PSMAþ,FLU: PSMA�). PSMA-expressing tumors could be visualizedspecifically on both SPECT/CT and NIRF imaging. However,intense radiotracer uptake was observed in the kidneys, whichwere attributed to the tracer's route of excretion and specificuptake by PSMA expressed in the kidneys.In addition to biodistribution and NIRF imaging experi-

ments, we also performed sequential dual-modality micro-SPECT/CT and NIRF imaging to demonstrate the feasibility ofthis pretargeting approach using the bsAb TF12 and the dual-label fluorescent hapten-peptide 111In-RDC018. Preferentialaccumulation of the dual-label hapten-peptide 111In-RDC018was confirmed with both NIRF and microSPECT/CT imagingin TROP-2–expressing soft-tissue and bone metastases. PC3metastases could be visualized clearly with both microSPECT/CT and NIRF imaging, showing accurate conformity of singletumor lesions by both imaging modalities. In addition, image-guided surgery based on microSPECT/CT and NIRF images

was performed to provide proof-of-principle for feasibility ofusing this pretargeting approach for intraoperative detectionof tumor lesions. It was shown that metastases imaged bymicroSPECT/CT and NIRF before surgery could be resectedwith image guidance using this pretargeting approach. More-over, NIRF imaging could be performed intraoperatively toensure that the tumor tissue was resected completely withoutleaving visible tumor-cell–containing resectionmargins in situ.MicroSPECT/CT imaging, which was performed after surgeryto exclude false-negative observations from intraoperativelyconducted NIRF imaging, confirmed complete resection of allmetastatic tumor lesions.

For clinical translation of the current approach, toxicitytests will have to be carried out. The toxicity of IMP288 wasinvestigated in mice: high doses were administered withoutany signs of toxicity. We have applied the IMP288 peptidelabeled with In-111 in patients (100 mg/patient), without anytoxicity (17). The toxicity of IRDye800CW has been studiedextensively in rats (18). Also, the first clinical studies withIRDye800CW-conjugated antibodies are ongoing and so far notoxicity has been reported.

In conclusion, compared with targeting approaches usingdual-labeled antibodies, pretargeting strategies may yield bet-ter signal-to-background ratios. Here, we show feasibility ofthe 111In-labeled near-infrared fluorescent hapten-peptideRDC018 for dual-modality detection of prostate cancer metas-tases in a mouse model of TROP-2–expressing soft-tissue andbony prostate cancer metastases pretargeted with the bsAbTF12. Both soft-tissue as well as bony prostate cancer lesionswere specifically and sensitively detected in vivo. Whereasradionuclide imaging may allow preoperative detection and

Figure 5. microSPECT/CT and NIRF image-guided surgery with 111In-RDC018 in a TF-12 pretargeted PC3 soft-tissue metastasis located below the scapularbone. Imageswere acquired at 2 hours after injection of 111In-RDC018. A,microSPECT/CT image before resection. B,NIRF image before resection illustratingthe limited penetration depth of the emitted fluorescent light, since the tumor is located below a bony structure. C, NIRF image after lifting the scapular bone tothe left-hand side, exposing the tumor; D, microSPECT/CT image after resection. E, corresponding NIRF image after resection.

www.aacrjournals.org Cancer Res; 74(21) November 1, 2014 OF7

Pretargeted Dual-Modality Imaging of Prostate Cancer

Research. on March 4, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 24, 2014; DOI: 10.1158/0008-5472.CAN-14-0594

Page 8: Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared ...cancerres.aacrjournals.org/content/canres/early/2014/10/15/0008-5472... · sections. The tissue sections were fixed with

intraoperative localization of tumor lesions, NIRF imagingenables subsequent accurate delineation of tumors and real-time assessment of resection margins. In addition, proof-of-principle for image-guided resection of metastatic lesionsusing this dual-modality pretargeting approach was provided.

Disclosure of Potential Conflicts of InterestD.M. Goldenberg and W.J. McBride have ownership interest (including

patents) in Immunomedics, Inc.W.J.G. Oyen received other commercial researchsupport. No potential conflicts of interest were disclosed by the other authors.

Authors' ContributionsConception and design: S. L€utje, M. Rijpkema, R.M. Sharkey, W. Helfrich,W.J.G. Oyen, O.C. BoermanDevelopment of methodology: S. L€utje, M. Rijpkema, D.M. Goldenberg,W.J. McBride, G.M. Franssen, O.C. BoermanAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): S. L€utje, D.M. Goldenberg, G.M. Franssen, C. FrielinkAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): S. L€utje, M. Rijpkema, C. Frielink, W.J.G. Oyen,O.C. Boerman

Writing, review, and/or revision of the manuscript: S. L€utje, M. Rijpkema,D.M. Goldenberg, C.M. van Rij, R.M. Sharkey, W.J. McBride, G.M. Franssen,C. Frielink, W. Helfrich, W.J.G. Oyen, O.C. BoermanStudy supervision: M. Rijpkema, O.C. Boerman

AcknowledgmentsThe authors thank Bianca Lemmers-van de Weem, Henk Arnts, Iris

Lamers-Elemans, and Kitty Lemmens-Hermans for their excellent technicalassistance with the animal experiments and Celeste Regino, Ph.D., forpreparing the peptide.

Grant SupportThis study was supported by the Dutch Cancer Society (grant KUN-2010-

4820) and was performed within the framework of CTMM, the Center forTranslational Molecular Medicine, PCMM project (grant 03O-203).

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

Received March 4, 2014; revised August 21, 2014; accepted August 27, 2014;published OnlineFirst September 24, 2014.

References1. Ravizzini G, Turkbey B, Kurdziel K, Choyke PL. New horizons in

prostate cancer imaging. Eur J Radiol 2009;70:212–26.2. van Rij CM, L€utje S, Frielink C, Sharkey RM, Goldenberg DM, Franssen

GM, et al. Pretargeted immuno-PET and radioimmunotherapy ofprostate cancer with an anti-TROP-2 x anti-HSG bispecific antibody.Eur J Nucl Med Mol Imaging 2013;40:1377–83.

3. Basu A, Goldenberg DM, Stein R. The epithelial/carcinoma antigenEGP-1, recognized by monoclonal antibody RS7-3G11, is phosphor-ylated on serine 303. Int J Cancer 1995;62:472–9.

4. van Rij CM, Sharkey RM, Goldenberg DM, Frielink C, Molkenboer JD,Franssen GM, et al. Imaging of prostate cancer with immuno-PET andimmuno-SPECT using a radiolabeled anti-EGP-1 monoclonal anti-body. J Nucl Med 2011;52:1601–7.

5. Heidenreich A, Bellmunt J, Bolla M, Joniau S, Mason M, Matveev V,et al. European Association of Urology. EAU guidelines on prostatecancer. Part 1: screening, diagnosis, and treatment of clinically loca-lised disease. Eur Urol 2011;59:61–71.

6. Keereweer S, Van Driel PB, Snoeks TJ, Kerrebijn JD, Baatenburg deJong RJ, Vahrmeijer AL, et al. Optical image-guided cancer surgery:challenges and limitations. Clin Cancer Res 2013;19:3745–54.

7. Keereweer S, Kerrebijn JD, van Driel PB, Xie B, Kaijzel EL, Snoeks TJ,et al. Optical image-guided surgery - where dowe stand?Mol ImagingBiol 2011;13:199–207.

8. Vahrmeijer AL, Hutteman M, van der Vorst JR, vande Velde CJ,Frangioni JV. Image-guided cancer surgery using near-infrared fluo-rescence. Nat Rev Clin Oncol 2013;10:507–18.

9. van Dam GM, Themelis G, Crane LM, Harlaar NJ, Pleijhuis RG, KelderW, et al. Intraoperative tumor-specific fluorescence imaging in ovariancancer by folate receptor-a targeting: first in-human results. Nat Med2011;17:1315–9.

10. Rossi EA, Goldenberg DM, Cardillo TM, McBride WJ, Sharkey RM,Chang CH. Stably tethered multifunctional structures of defined com-

positionmadeby the dock and lockmethod for use in cancer targeting.Proc Natl Acad Sci U S A 2006;103:6841–6.

11. Brown JM. In vivomodels of human prostate cancer bone metastasis.Methods Mol Med 2003;81:149–62.

12. L€utjeS,RijpkemaM,FranssenGM,FracassoG,HelfrichW,EekA, et al.Dual-modality image-guided surgery of prostate cancer with a radi-olabeled fluorescent anti-PSMA monoclonal antibody. J Nucl Med2014;55:995–1001.

13. Chen Y, Dhara S, Banerjee SR, Byun Y, Pullambhatla M, Mease RC,et al. A low molecular weight PSMA-based fluorescent imaging agentfor cancer. Biochem Biophys Res Commun 2009;390:624–9.

14. Nakajima T, Mitsunaga M, Bander NH, Heston WD, Choyke PL,Kobayashi H. Targeted, activatable, in vivo fluorescence imaging ofprostate-specificmembrane antigen (PSMA) positive tumors using thequenched humanized J591 antibody-indocyanine green (ICG) conju-gate. Bioconjug Chem 2011;22:1700–5.

15. Cohen R, Stammes MA, de Roos HC, Stigter-van Walsum M, VisserGWM, van Dongen GAMS. Inert coupling of IRDye800CW to mono-clonal antibodies for clinical imaging of tumor targets. EJNMMI Res2011;1:31.

16. Banerjee SR, Pullambhatla M, Byun Y, Nimmagadda S, Foss CA,Green G, et al. Sequential SPECT and optical imaging of experimentalmodels of prostate cancer with a dual modality inhibitor of the pros-tate-specific membrane antigen. Angew Chem Int Ed Engl 2011;50:9167–70.

17. Schoffelen R, BoermanOC, Goldenberg DM, Sharkey RM, vanHerpenCM, Franssen GM, et al. Development of an imaging-guided CEA-pretargeted radionuclide treatment of advanced colorectal cancer:first clinical results. Br J Cancer 2013;109:934–42.

18. Marshall MV, Draney D, Sevick-Muraca EM, Olive DM. Single-doseintravenous toxicity study of IRDye 800CW in Sprague-Dawley rats.Mol Imaging Biol 2010;12:583–94.

Cancer Res; 74(21) November 1, 2014 Cancer ResearchOF8

L€utje et al.

Research. on March 4, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 24, 2014; DOI: 10.1158/0008-5472.CAN-14-0594

Page 9: Pretargeted Dual-Modality Immuno-SPECT and Near- Infrared ...cancerres.aacrjournals.org/content/canres/early/2014/10/15/0008-5472... · sections. The tissue sections were fixed with

Published OnlineFirst September 24, 2014.Cancer Res   Susanne Lütje, Mark Rijpkema, David M. Goldenberg, et al.   CancerFluorescence Imaging for Image-Guided Surgery of Prostate Pretargeted Dual-Modality Immuno-SPECT and Near-Infrared

  Updated version

  10.1158/0008-5472.CAN-14-0594doi:

Access the most recent version of this article at:

   

   

   

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

  Subscriptions

Reprints and

  [email protected] at

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

  Permissions

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

.http://cancerres.aacrjournals.org/content/early/2014/10/15/0008-5472.CAN-14-0594To request permission to re-use all or part of this article, use this link

Research. on March 4, 2020. © 2014 American Association for Cancercancerres.aacrjournals.org Downloaded from

Published OnlineFirst September 24, 2014; DOI: 10.1158/0008-5472.CAN-14-0594