Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic...

12
Integrated Systems and Technologies Nitroreductase, a Near-Infrared Reporter Platform for In Vivo Time-Domain Optical Imaging of Metastatic Cancer Emmet McCormack 1 , Elisabeth Silden 1 , Richard M. West 5 , Tina Pavlin 2 , David R. Micklem 2 , James B. Lorens 2 , Bengt Erik Haug 3 , Michael E. Cooper 5 , and Bjørn Tore Gjertsen 1,4 Abstract The ability to visualize reporter gene expression in vivo has revolutionized all facets of biologic investigation and none more so than imaging applications in oncology. Near-infrared reporter gene imaging may facilitate more accurate evaluation of chemotherapeutic response in preclinical models of orthotopic and metastatic cancers. We report the development of a cell permeable, quenched squarine probe (CytoCy5S), which is reduced by Escherichia coli nitroreductase (NTR), resulting in a near-infrared uorescent product. Time-domain molecular imaging of NTR/CytoCy5S reporter platform permitted noninvasive monitoring of disease progression in orthotopic xenografts of disseminated leukemia, lung, and metastatic breast cancer. This methodology facilitated thera- peutic evaluation of NTR genedirected enzymatic prodrug therapy with conventional metronidazole antibiotics. These studies show NTR/CytoCy5S as a near-infrared gene reporter system with broad preclinical and prospective clinical applications within imaging, and gene therapy, of cancer. Cancer Res; 73(4); 127686. Ó2012 AACR. Introduction Noninvasive imaging, permitting spatiotemporal monitor- ing of both disease progression and therapeutic efcacy (1), has had a major impact upon human drug development (2). In particular, optical imaging of uorescent and bioluminescent reporters has emerged as one of the most important preclinical imaging modalities, with considerable developments in sen- sitive instrumentation, e.g., time-domain optical imaging (3), contributing to its success. The relative simplicity of optical hardware, ease of use, and high-throughput potential has rendered optical imaging as an inexpensive modality accessi- ble to both academic and industrial environments alike. While reporter genes remain the cornerstone of the optical approach, an ever-expanding palette of targeted and activatable probes, exploiting the near infrared spectrum of light, enable a more complete investigation of disease pathology in situ (4). Anticancer drug screening in subcutaneous tumor models is neither representative of human cancer nor predictive of clinical success (5). A consequent paradigm shift toward drug evaluation in more clinically relevant orthotopic and meta- static models has placed a greater emphasis upon imaging cancer cells in dense parenchymatous organs, demanding sensitive, deep tissue imaging. However, the major caveats of visualizing uorescent proteins for this application has been autouorescence and signicant attenuation of both their excitation and emitted light in mammalian tissue by hemo- globin. While use of time-domain optical imaging can delineate autouorescence from uorescent protein uorescence on the basis of uorescence lifetime (3, 6), signicant efforts have been placed on development of far-redshifted uorescent proteins that might exploit the far-red and near-infrared window (approximately 630900 nm) where hemoglobin absorption is minimal (711). Bioluminescence imaging, requiring no excitation source, has proven to be one of the most sensitive platforms for small animal imaging (12). How- ever, bioluminescence emission maxima are still less than 620 nm, which in addition to signicant scattering of biolumines- cent photons, limit spatial resolution and detection at depth. Nitroreductase (NTR), a avoprotein that reduces nitroaro- matic prodrugs in the presence of NADPH or NADH to highly toxic metabolites, has found clinical application as a prodrug therapeutic strategy in cancer (13). Subsequently, a number of quenched uorogenic probes have been designed as substrates for NTR (1418), and pilot studies probing their use in NTR- mediated prodrug therapy have been carried out (18). These recent developments suggest that NTR and suitable quenched near-infrared substrate may be exploited as the basis for a reporter gene based in vivo optical imaging platform. Here, we present NTR reporter gene imaging as a method for visualization of disseminated, orthotopic, and metastatic can- cer in vivo, using CytoCy5S as a quenched, near-infrared substrate. Synthetic methods of squarine-based NTR substrate development, characterization, and mechanism of NTR Authors' Afliations: 1 Institute of Medicine, Hematology Section; 2 Department of Biomedicine, and 3 Department of Chemistry, University of Bergen; 4 Department of Internal Medicine, Hematology Section, Hauke- land University Hospital, Bergen, Norway; and 5 GE Healthcare, The May- nard Centre, Cardiff, Wales, United Kingdom Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Authors: Emmet McCormack, Institute of Medicine, Hematology Section, University of Bergen, N-5021 Bergen, Norway. Phone: 475-597-5000; Fax: 475-597-2950; E-mail: [email protected], and Bjørn Tore Gjertsen, E-mail: [email protected] doi: 10.1158/0008-5472.CAN-12-2649 Ó2012 American Association for Cancer Research. Cancer Research Cancer Res; 73(4) February 15, 2013 1276 on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Transcript of Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic...

Page 1: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

Integrated Systems and Technologies

Nitroreductase, a Near-Infrared Reporter Platform for In VivoTime-Domain Optical Imaging of Metastatic Cancer

EmmetMcCormack1, Elisabeth Silden1, RichardM.West5, Tina Pavlin2, David R.Micklem2, James B. Lorens2,Bengt Erik Haug3, Michael E. Cooper5, and Bjørn Tore Gjertsen1,4

AbstractThe ability to visualize reporter gene expression in vivohas revolutionized all facets of biologic investigation and

none more so than imaging applications in oncology. Near-infrared reporter gene imaging may facilitate moreaccurate evaluation of chemotherapeutic response in preclinicalmodels of orthotopic andmetastatic cancers.Wereport the development of a cell permeable, quenched squarine probe (CytoCy5S), which is reduced by Escherichiacoli nitroreductase (NTR), resulting in a near-infrared fluorescent product. Time-domain molecular imaging ofNTR/CytoCy5S reporter platform permitted noninvasive monitoring of disease progression in orthotopicxenografts of disseminated leukemia, lung, and metastatic breast cancer. This methodology facilitated thera-peutic evaluation of NTR gene–directed enzymatic prodrug therapywith conventionalmetronidazole antibiotics.These studies showNTR/CytoCy5S as a near-infrared gene reporter systemwith broad preclinical andprospectiveclinical applications within imaging, and gene therapy, of cancer. Cancer Res; 73(4); 1276–86. �2012 AACR.

IntroductionNoninvasive imaging, permitting spatiotemporal monitor-

ing of both disease progression and therapeutic efficacy (1), hashad a major impact upon human drug development (2). Inparticular, optical imaging of fluorescent and bioluminescentreporters has emerged as one of themost important preclinicalimaging modalities, with considerable developments in sen-sitive instrumentation, e.g., time-domain optical imaging (3),contributing to its success. The relative simplicity of opticalhardware, ease of use, and high-throughput potential hasrendered optical imaging as an inexpensive modality accessi-ble to both academic and industrial environments alike. Whilereporter genes remain the cornerstone of the optical approach,an ever-expanding palette of targeted and activatable probes,exploiting the near infrared spectrum of light, enable a morecomplete investigation of disease pathology in situ (4).

Anticancer drug screening in subcutaneous tumormodels isneither representative of human cancer nor predictive ofclinical success (5). A consequent paradigm shift toward drug

evaluation in more clinically relevant orthotopic and meta-static models has placed a greater emphasis upon imagingcancer cells in dense parenchymatous organs, demandingsensitive, deep tissue imaging. However, the major caveats ofvisualizing fluorescent proteins for this application has beenautofluorescence and significant attenuation of both theirexcitation and emitted light in mammalian tissue by hemo-globin.While use of time-domain optical imaging can delineateautofluorescence from fluorescent protein fluorescence on thebasis of fluorescence lifetime (3, 6), significant efforts havebeen placed on development of far-red–shifted fluorescentproteins that might exploit the far-red and near-infraredwindow (approximately 630–900 nm) where hemoglobinabsorption is minimal (7–11). Bioluminescence imaging,requiring no excitation source, has proven to be one of themost sensitive platforms for small animal imaging (12). How-ever, bioluminescence emission maxima are still less than 620nm, which in addition to significant scattering of biolumines-cent photons, limit spatial resolution and detection at depth.

Nitroreductase (NTR), a flavoprotein that reduces nitroaro-matic prodrugs in the presence of NADPH or NADH to highlytoxic metabolites, has found clinical application as a prodrugtherapeutic strategy in cancer (13). Subsequently, a number ofquenched fluorogenic probes have been designed as substratesfor NTR (14–18), and pilot studies probing their use in NTR-mediated prodrug therapy have been carried out (18). Theserecent developments suggest that NTR and suitable quenchednear-infrared substrate may be exploited as the basis for areporter gene based in vivo optical imaging platform.

Here, we present NTR reporter gene imaging as amethod forvisualization of disseminated, orthotopic, and metastatic can-cer in vivo, using CytoCy5S as a quenched, near-infraredsubstrate. Synthetic methods of squarine-based NTR substratedevelopment, characterization, and mechanism of NTR

Authors' Affiliations: 1Institute of Medicine, Hematology Section;2Department of Biomedicine, and 3Department of Chemistry, Universityof Bergen; 4Department of Internal Medicine, Hematology Section, Hauke-land University Hospital, Bergen, Norway; and 5GE Healthcare, The May-nard Centre, Cardiff, Wales, United Kingdom

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

Corresponding Authors: Emmet McCormack, Institute of Medicine,Hematology Section, University of Bergen, N-5021 Bergen, Norway.Phone: 475-597-5000; Fax: 475-597-2950; E-mail:[email protected], and Bjørn Tore Gjertsen, E-mail:[email protected]

doi: 10.1158/0008-5472.CAN-12-2649

�2012 American Association for Cancer Research.

CancerResearch

Cancer Res; 73(4) February 15, 20131276

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 2: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

reduction are described for CytoCy5S. In our method, weadopted time-domain imaging, which provides greater sensi-tivity and significantly higher spatial resolution and detectionat depth as compared with epi-illumination imaging (19, 20).Time-domain imaging of the NTR/CytoCy5S platform permit-ted longitudinal in vivo imaging of disseminated and ortho-topic cancermodels of lung, breast, and leukemia. Comparisonof time-domain gated NTR/CytoCy5S fluorescence imaging tofluorescence protein imaging with enhanced GFP showedgreater sensitivity, detection at depth, and resolution ofmetas-tasis. Finally, we illustrate therapeutic efficacy of gene-directedenzyme prodrug therapy in vivo using the NTR/CytoCy5Smethodology to visualize therapeutic efficacy.

Materials and MethodsFor synthesis of CytoCy5S, nuclear magnetic resonance

(NMR), high resolution mass spectrometry (HRMS), chro-matographic analysis and purification, NTR-CytoCy5S enzymeassay, liquid chromatography/mass spectrometry (LC/MS),spectrophotometry, and Western Blot analysis, see Supple-mentary Materials and Methods.

Cell lines and cell cultureThe promyelocytic leukemia NB4 cell line was a gift fromDr.

Lanotte (Hospital Saint Luis, Paris, France). IPC-81NTRþGFPþ

was kindly provided by Prof. SteinOveDøskeland (University ofBergen, Norway), whereas NCI-H460Lucþ and MDA-MB-231Lucþ cells were kindly provided by Prof. James Lorens(University of Bergen, Bergen, Norway). NB4 and NCI-H460cells were maintained in RPMI-1640 medium (Sigma-Aldrich,Inc.). MDA-MB-231 and IPC-81 cells were maintained in Dul-becco'sModified Eagle's Medium. Themedia were supplemen-ted with 10% heat-inactivated FBS, or 10% horse serum (IPC-81; PAA Laboratories GmbH), 1% penicillin/streptomycin (PS;Sigma-Aldrich), and 1% L-glutamine (Sigma-Aldrich), and thecells were incubated in a humidified atmosphere at 37�C in 5%CO2. Authentication of all cell lines was done by DNA finger-printing using the AmpFlSTR Profiler Plus PCR Amplificationkit (Applied Biosystems), conducted in April 2012.

GFPþNTRþ and GFPþNTR� vector constructionThe retroviral expression vector L149 pTra Puro2AGFP2A-

Luciferase2ANTR (Entrez: EU753858; GFPþNTRþ) was madein several stages (details on request) by cloning the codingsequences of puromycin-N-acetyl-transferase, EGFP, fireflyluciferase, and NTR into pTra (21) to allow Tet-regulatedexpression. Each open-reading frame was separated from thenext by a linker encoding the 2A region (XXSGLRSGQLLN-FDLLKLAGDVESNPGP) from foot-and-mouth disease virus.This sequence is cleaved cotranslationally, resulting in theproduction of approximately stoichiometric amounts of eachprotein (22). Cleavage of the 2A linker leaves a short tag on theC-terminus of the preceding protein and a single proline fusedto the N-terminus of the following protein. Although puromy-cin resistance, green fluorescence, and NTR activity werereadily detected in the presence of the retroviral Tet ActivatorCttaIH (23), no luciferase activity could be detected. This is

most probably due to the N-terminal proline or C-terminal 2Atag interfering with folding/activity, as the presence of NTRimplies successful translation of luciferase.

A GFPþNTR� control vector L134 pTra Puro2AGFP (GFPþ)is identical to L149 except that GAATTCTAGAGCGGCCGCreplaces the sequence between the EcoRI site following EGFPand the unique NotI site. This construct expresses puromycinresistance and green fluorescence.

Retroviral transfection of NB4 cells and selection of NB4GFPþNTRþ cells by FACS

NB4, NCI-H460Lucþ, and MDA-MB-231Lucþ cells stablyexpressing nitroreductase (NTR) and enhanced GFP (eGFP)were engineered by retroviral transduction with the GFPþ

NTRþ (L149) and Tet Activator CttaIH vector. Production ofinfectious retroviral vector particles in 293-based Phoenix Apackaging cells and infection of cells were carried out asdescribed (22). Likewise, NB4NTR- GFPþ (L134) cells (control)were generated.

Flow cytometric analysisAccuri (Accuri Cytometers Ltd.) or FACSCalibur (BD Bio-

sciences) flow cytometers were used to quantify GFP fluores-cence [FL1 channel; 488 nm excitation laser, 530/30 band-pass(BP) filter)] and NIR fluorescence [NTR/CytoCy5S; 635 nmexcitation laser, 661/16 BP filter (Calibur), or 640 nm excitationlaser, 675/12.5 BP filter (Accuri)] from transduced cells. A totalof 1 � 106 cells were preincubated with CytoCy5S (1 mmol/L)for 1 hour before analysis as indicated. In the study comparingCytoCy5S fluorescence after metronidazole (Mtz; 0.6 mmol/L)treatment, 3.5� 105 cells were incubated�metronidazole for2 hours, and the last hour with CytoCy5S (1 mmol/L). Whenanalyzed on the cytometer, 10,000 events per sample wereacquired, and data analyzed using FlowJo (TreeStar) softwareversion 8.8.6. Stably high expressing GFPþNTRþ and GFPþ

cells were isolated by a Fluorescence Activated Cell Sorter(FACSAria; BD Biosciences) using a 488 laser for GFP sortingand 638nm laser for sortingNTRþ cells preincubated for 1 hourwith 1 mmol/L CytoCy5S.

Fluorescence microscopyImages of cell fluorescence were acquired with a Zeiss Axio

Observer Z1 inverted microscope (Carl Zeiss MicroimagingGmbH) and analyzed by the AxioVision 4.8.2 software. ForNTR-CytoCy5S fluorescence, 5 � 104 cells were preincubatedin 1mmol/LCytoCy5S for 1 hour beforewashingwith 1�PBS forlive cell imaging, or CytoCy5Swas added just before imaging forlongitudinal fluorescence microscopy as indicated. The cellswere kept at 37�C during imaging. NTR-CytoCy5S fluorescencewas imaged with a BP 540–580 excitation filter and BP 630–675 nm emission filter GFP with a BP 450–490 nm filter and BP515–565 nm emission filter and 40, 6-diamidino-2-phenylindole(DAPI) with a 365 nm excitation filter and BP 420–470 emissionfilter. Phase contrast images were also acquired.

Cell viability analysisNTRþ and NTR� cells were incubated for 24 hours with

metronidazole (0.3, 0.6, and 2.4 mmol/L) or CytoCy5S (0, 0.001,

NTR Optical Imaging of Metastatic Cancer

www.aacrjournals.org Cancer Res; 73(4) February 15, 2013 1277

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 3: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

0.01, 0.1, and 1 mmol/L) and cell viability evaluated by labelingcells with the DNA intercalating dye Hoechst 33342. Nuclearmorphology was investigated as described (24). 300 to 400 cellswere counted in each well to determine the fraction of cellswith normal/abnormal nuclear morphology. NB4 cell viabilityfollowing 24 hours incubation withmetronidazole (0.3, 0.6, and2.4 mmol/L) was also determined using the Annexin-V, PacificBlue (Life Technologies Ltd), and propidium iodide (PI; Sigma-Aldrich) assay. Cells were washed in PBS and resuspended inbinding buffer (2.5% Annexin-V, Pacific Blue). Samples wereincubated for 15 minutes at room temperature and addedbinding buffer with PI (final concentration 0.2 mg/mL). Thedata were acquired on a BD LSR II Fortessa flow cytometer (BDBiosciences) and analyzed using Cytobank (www.cytobank.org). The percentage of living cells is displayed relative tountreated control cells. 3H-thymidine incorporation in cellswas used to examine effect of NTR and/or CytoCy5S onproliferation. NTRþ and NTR� cells (NB4, NCI-H460, andMDA-MB-231) were seeded in 96-well plates and left to settlefor 20 hours before treatment. The cells were treated withCytoCy5S (0, 0.001, 0.01, 0.1, and 1 mmol/L) or metronidazole(0, 0.3, 0.6, 2.4 mmol/L) for 24 hours, and 3H-thymidine (1 mCiper well; TRA310, Amersham International) was added the last18 hours of the treatment period before harvesting and analysisof the cells using a Packard Microplate Scintillation andLuminescence counter (PerkinElmer Life And AnalyticalSciences, Inc.).

General animal careAll experiments were approved by The Norwegian Animal

Research Authority and conducted according to The EuropeanConvention for the Protection of Vertebrates Used for Scien-tific Purposes. NOD/LtSz-Prkdcscid (NOD/SCID), NOD/LtSz-Prkdcscid/B2mnull (NOD/SCID/B2m), and NOD/LtSz-Prkdcscid/IL-2Rgnull (NSG) mice (Gades Institute, Universityof Bergen. NOD-scid IL-2Rgnull (originally a generous gift fromProf. Leonard D. Shultz, Jackson Laboratories) were housedin groups of 5 or less in individually ventilated cages (Tech-niplast) and kept on a 12-hour dark/light schedule at a con-stant temperature of 21�C and 50% relative humidity. Themicehad continuous access to food and autoclaved water. Duringdepilation and imaging, mice were anaesthetized with 1%isofluorane.

Leukemia, lung, and breast subcutaneous andorthotopic xenograft models

NOD/SCID and NOD/SCID/B2m mice were irradiated froma photon radiation source (BCC Dynaray CH4, 4 megavoltphoton irradiation source), with a sublethal dose of 2.5 Gybefore subcutaneous or intraperitoneal injections of leukemiccells, respectively. A total of 10 � 106 or 5 � 106 NB4NTRþGFPþ

cells were suspended in 200 mL of sterile 1� PBS beforeinjection intraperitoneally or intravenously and in 100 mL1� PBS with 10% Matrigel (Invitrogen) for subcutaneousinoculation. NCI-H460NTRþLucþGFPþ cells were suspended in20 mL 1� PBS with 25% Matrigel for intrapulmonary injection.A total of 1 � 106 MDA-MB-231NTRþLucþGFPþ cells were sus-pended in 50 mL 1� PBS with 25% Matrigel for intramam-

mary injection. Mice treated with metronidazole were given50 mg/kg intravenously twice daily when tumors reached 100to 150 mm3. All cell suspensions were injected with a 28 Gsyringe. Inoculated animals weremonitored closely for signs ofadvancing disease such as weight loss, ruffled fur and lethargy,in general. Mice were sacrificed according to Institutionalguidelines.

Time-domain optical imagingGFP and bioluminescence images were acquired as

previously described (3, 25). Time-domain imaging of NTR/CytoCy5S fluorescence was acquired 30 to 60 minutes follow-ing administration of 69 mg CytoCy5S in a 5% Cremaphore ELsolution in saline (100 mL) intravenously via the dorsal tail vein.Bioluminescence images were acquired 10 minutes followingadministration of 150 mg/kg D-luciferin (Promega) intraperi-toneally. All imaging was conducted using Optix MX2 Time-DomainMolecular Imager (ART Inc.). All imageswere acquiredwith raster scan points 1 mm apart and integration time of0.3 to 1.5 seconds per raster point. Typical scanning times for awhole body image with integration times of 0.3 second perraster point were approximately 5 minutes. NTR/CytoCy5Sfluorescence images were acquired using 670 or 635 nm pulsedlaser diodes as excitation sources with 700 or 650 LP filters,respectively. Fluorescence lifetime gating was conducted withOptix Optiview (software versions 1.04.01, 2.00.01, 2.01.00 and2.02.00 ART Inc.), gating for peak NTR/CytoCy5S fluorescencelifetime of approximately 1.2 ns between 1.0 and 1.3 ns.

MR imaging of miceMR imageswere acquired using a 7T horizontal boremagnet

(Pharmascan 70/16, Bruker BioSpin) operating at 300 MHz. A38-mm diameter linear volume resonator was used for trans-mission and reception of radiofrequency (RF) radiation. After aTriPilot scan, a T1-weighted FLASH pulse sequence (echo time(TE)/repetition time (TR)¼ 3.416ms/247ms, 25� flip angle, 0.7mm slice thickness, 8 consecutive slices, 3.0� 3.0 cm2

field-of-view, 117 � 117 mm2 in-plane resolution) was used to scanaxially through the lungs to localize the lung tumor. Theacquisition was respiratory triggered to minimize motionartifacts. Breathing rate limited the maximum TR time andtherefore the number of slices collected in each image. Oncethe tumor was localized, we identified the slice with thelargest tumor cross section. We then imaged this slice usingretrospective respiratory and cardiac triggering (Bruker Intra-gate ig-FLASH-cine pulse sequence, TE/TR ¼ 2.989 ms/8 ms,10� flip angle, 0.7 mm slice thickness, 1 slice, 6.0 � 6.0 cm2

field-of-view, 234 � 234 mm2 in-plane resolution) to get amotion-free image of the tumor. Finally, we imaged the tumoragain using our T1-weighted FLASH pulse sequence (sameparameters as for the axial T1-weighted FLASH), but this timeusing sagittal and coronal slice orientation.

HistologyOrgan or tumor samples collected following euthanasia

were transferred to a tube containing 4% formalin for para-ffin-embedding, cryosectioning, and subsequent immunohis-tochemistry of the samples. Sections were stained with hema-

McCormack et al.

Cancer Res; 73(4) February 15, 2013 Cancer Research1278

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 4: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

toxylin and eosin (H&E), and results were analyzed by standardlight microscopy (Olympus BX51, Olympus America Inc.).

StatisticsIn cell viability and proliferation assays, triplicates were

analyzed for each sample and results given as mean � SEM.Statistical significance of differences in averages betweentreatment groups in vitro and in vivo was determined usinga 2-tailed Student t test (GraphPad Prism 5.0, GraphPadSoftware). For all statistical analysis, P < 0.05 was consideredsignificant.

ResultsSynthesis and characterization of NTR-activatedfluorescent probeNitroreductase catalyses the reduction of nitro-aryl groups

to the corresponding hydroxylamine, providing the basis of agene-directed enzymatic prodrug therapy (13). We postulatedthat introduction of an electron withdrawing dinitrophenylmoiety into the far-red chromophore Cy5 would effectivelyquench its fluorescence, and upon intracellular enzymaticreduction by NTR, yield a near-infrared fluorescent productproviding the basis of an NTR-based reporter gene platform(Supplementary Figs. S1A, S1B, S2A, and S2B). Thus, introduc-tion of dinitro-aryl- and squarate functionality into the Cy5chromophore yielded CytoCy5S (Supplementary Fig. S1A), aproposed cell permeable substrate for NTR (26). LC/MS anal-ysis of CytoCy5S showed a peak at 6.32minutes using detectionat 630 nm, which upon initial incubation with NTR in thepresence of b-NADH and b-NADPH resulted in 2 additionalpeaks with retention times of 3.08 and 1.42 minutes, respec-tively (Supplementary Fig. S1C). MS analysis revealed the 3.08-minute product resultant of 2 ions with m/z of 677 and 675,consistent with the mono hydroxylamine and the mono-nitroso reduction products, respectively. Analysis of the peakappearing at 1.42 minutes suggests reduction of both nitrogroups to the corresponding hydroxylamines, giving an m/z of663 for the protonated species (Supplementary Fig. S1C). Thesedata are consistent with an overall reduction mechanism ofCytoCy5S to the dihydroxyl product via nitroso intermediates(Supplementary Fig. S1B). After the 48-minute reaction time,only minor amounts of both starting material and the mono-reduction products could be detected (Supplementary Fig.S1D). Spectrophotometric analysis of the final reduction prod-uct revealed excitation and emission maxima of 631 and 688nm, respectively (Supplementary Fig. S1E), and NTR-depen-dent increase in fluorescence in enzymatic assays (Supple-mentary Fig. S1F). Furthermore, kinetic analysis showed arapid fluorescence induction upon addition of CytoCy5S sub-strate to NTR (Supplementary Fig. S1G).

In vitro evaluation of CytoCy5S in NTR-expressing celllinesHaving shownCytoCy5S as a substrate for NTR resulting in a

highly fluorescent near-infrared product, we retrovirally trans-duced a diverse array of cancer cells lines to explore its cellpermeability and application as a cellular gene reporter assay.Accordingly, the cell lines NB4 (human acute promyelocytic

leukemia), IPC-81 (rat acute myeloid leukemia), NCI-H460Lucþ

(human large cell lung carcinoma), and MDA-MB-231Lucþ

(human breast carcinoma) were retrovirally transduced withthe tTA-dependent L149 plasmid expressing both GFP andNTR and tTAplasmids (Supplementary Fig. S2A, S2B, and S2D).Western blots of transfected cell lines confirmed NTR and GFPexpression, as exemplified for NB4NTRþGFPþ cells (Supplemen-tary Fig. S2C). Fluorescence microscopy of GFP (green) andNTR/CytoC5S (red) fluorescence illustrated bright fluores-cence in both channels (Supplementary Fig. S2D), whereasoverlay (yellow/orange) of GFP and CytoCy5S fluorescencecorrelated well. Stable transduction of these cell lines with thetTA-dependent L149 and tTAplasmids did not alter cell growthin comparison with controls (Supplementary Fig. S4A). Incu-bation of cell lines with 0.1 mmol/L CytoCy5S and fluorescencemicroscopy showed a rapid induction of bright fluorescence inNTR-expressing cell lines (Supplementary Fig. S2E). Flowcytometry analysis of NB4NTRþGFPþ and NB4WT cells followingincubation with 0.1 mmol/L CytoCy5S showed more than 100-fold increase in fluorescence in NB4NTRþGFPþ over NB4WT

(Supplementary Fig. S6) corresponding to enzymatic results(Supplementary Fig. S1F). Crucially, incubation of cells withCytoCy5S did not affect the proliferation or viability of NTRþ orNTR� cell lines (Supplementary Fig. S4B and S4C). To evaluatethe use of the NTR/CytoCy5S system for time-domain opticalimaging applications, pellets of defined numbers of NTRþ cells(0.01–1 � 106 cells) were immersed in a liposyn/Indian inkphantom, as previously described (3) and imaged using a time-domainmolecular imagerwith excitation source of 670 nmandLP filter of 700 nm (Supplementary Fig. S2F). Excellent linearcorrelation between cell number and fluorescence intensitywere observed and imaging at depths of 6 to 8 mm wasachievable with 0.1–1� 106 cells (Supplementary Fig. S2G andS2H).

Time-domain imaging of NTR/CytoCy5S fluorescenceWe next examined the suitability of the NTR/CytoCy5S

fluorescent reporter gene platform for near-infrared in vivooptical imaging. NOD/SCID mice were implanted subcutane-ously with NB4NTRþGFPþcells and imaging was conducted with670 nm pulsed laser diode source and 700 nm long passcollection filter following injection of 69 mg CytoCy5S intra-venously (Supplementary Fig. S3A). As previously observedin vitro, induction of fluorescence was rapid with brighttumoral fluorescence noted 10 minutes after CytoCy5S injec-tion. Stable fluorescence was observed between 1 and 4 hours(Supplementary Fig. S3A and S3C) andwas still evident 8 hoursafter injection. Similar observations were made imagingNSG mice 8 days following orthotopic injection with NCI-H460NTRþLucþGFPþ into the left lung parenchyma (Supplemen-tary Fig. S3D). Induction of gastrointestinal fluorescence wasalso noted after CytoCy5S injection (Supplementary Fig. S3A,bottom), as previously reported (18). Analysis of the CytoCy5Sfluorescence lifetimes from both NTR-expressing subcutane-ous tumors and gastrointestinal fluorescence showed distinctdistributions up to 130 minutes (Supplementary Fig. S3B) afterwhich time they coalesce, suggesting fluorescence lifetimegatingmay be possible to discriminate between these 2 sources

NTR Optical Imaging of Metastatic Cancer

www.aacrjournals.org Cancer Res; 73(4) February 15, 2013 1279

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 5: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

of fluorescence up to 2 hours following CytoCy5S administra-tion.

Fluorescence lifetime gating of NTR/CytoCy5Sfluorescence in vivo

The observation of gastrointestinal CytoCy5S-inducedfluorescence would seriously impede the application of theNTR/CytoCy5S reporter system in models with peritonealinvolvement. Thus, to rigorously examine whether fluores-cence lifetime gating could be used to differentiate betweenCytoCy5S-induced gastrointestinal fluorescence and tumoralNTR fluorescence, we imaged control mice with no cancercells, mice-bearing vector control (NB4GFPþ) or NTRþ

(NB4NTRþGFPþ) blood cancer cells (n ¼ 4 per group) in theperitoneal cavity, one hour � following the administration ofCytoCy5S (Fig. 1A). Fluorescence was noted in all animals;however, the distribution of fluorescence lifetimes observed inthe animals differed in mice-bearing NTRþ cells (Fig. 1B) aspreviously noted at this time point (Supplementary Fig. S3B).While all control groups showed distributions with multiplefluorescence lifetimes at this time point, only one predominantlifetime (peak of 1.2 ns) was evident in mice bearing NTR-positive tumors. Exploiting the differences in fluorescencelifetime distributions between control and NTR-positivetumors (Fig. 1C), fluorescence lifetime gating (1.0–1.3 ns) wasused to identify only those pixels exhibiting NTR/CytoCy5Slifetime, and used to generate gated fluorescence intensitymaps of scanned mice (Fig. 1D). Subsequently all micewere imaged 30 to 60 minutes after CytoCy5S injection and

this gating strategy was used in all ensuing work describedhere.

Longitudinal NTR/CytoCy5S imaging of disseminatedand metastatic disease progression in vivo

Having established fluorescence lifetime gating as a robustmethod to discriminate CytoCy5S induced gastrointestinalfluorescence from NTR-expressing cancer cells, we evaluatedthe system in longitudinal monitoring of disseminated andmetastatic cancer progression, and validated results withcommonly used GFP or luciferase gene reporters. NSG micewere injected intravenously with IPC-81NTRþGFPþ (n¼ 4), IPC-81WT (n ¼ 4) rat leukemia cells or NB4NTRþGFPþ human acutepromyelocytic cells (n ¼ 7) and disease course imaged withNTR/CytoCy5S (30 minutes following CytoCy5S i.v. injection)andGFP-gatedfluorescence. Femoralfluorescencewas evidentas early as 5 to 7 days following IPC-81NTRþGFPþ cell injection(Fig. 2A) with increase in NTR/CytoCy5S-gated fluorescence(r2¼ 0.6316) notedas a functionof time (Fig. 2D). Leukemiawasfirst evident after 17 to 24 days using GFP-gated fluorescenceimaging in the same mice (Fig. 2C). IPC-81WT showed minimalNTR/CytoCy5S fluorescence (Fig. 2B and D). Ex vivo imagingand H&E staining of bonemarrow samples from the femur andsterna of mice confirmed leukemia (Fig. 2E). Similarly, NTR/CytoCy5S-gated images of NB4NTRþGFPþ mice revealed earlierdetection of leukemia, particularly in the spines and femurs,than respective GFP-gated images at day 14 and moreextensive disease patterns at day 18 (Fig. 2F). In vivo tumorswere confirmed by ex vivo imaging and histology (Fig. 2G).

Figure 1. In vivo time-domainresolution of fluorescent lifetimes.A, comparative near-infraredfluorescence intensity images(same scale) of control mice(� CytoCy5S) and mice bearingeither GFPþ or NTRþGFPþ tumorsintraperitoneally (þ CytoCy5Sinjected i.v.; n ¼ 4 per group). Redboxes indicate scan areas. B,fluorescence lifetimes from eachanimal differ both in fluorescenceintensity and distribution betweencontrols and NTRþ tumor bearingmice. C, gating for the NTR/CytoCy5S-specific fluorescencelifetime in all images presented inA results in completediscrimination of NTR/CytoCy5Sfluorescence (all images have thesame fluorescence scale) fromboth endogenous and exogenousautofluorescence (D).

McCormack et al.

Cancer Res; 73(4) February 15, 2013 Cancer Research1280

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 6: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

NCI-H460NTRþLucþGFPþ cells, expressing NTR, GFP, andluciferase, were injected orthotopically into the left lungparenchyma of NSG mice (n ¼ 5) and imaged on days 5, 8,and 14 for NTR/CytoCy5S fluorescence (Fig. 3A and F). Bio-luminescence imaging andT1-weighted FLASHMRI confirmedtumor placement (example in Fig. 3B and C). At days 5 and 8,NTR/CytoCy5S-gated fluorescence images were comparablewith acquired bioluminescence images. Six days later, bothbioluminescence and NTR/CytoCy5S-gated fluorescenceimages revealed extensive pulmonary metastasis in dorsal andventral aspects. Necropsy of the mice and ex vivo imagingverified extensive lung metastasis (Fig. 3D and E), which wasconfirmed histologically (Fig. 3G and H). Encouragingly,NTR/CytoCy5S-gated dorsal images demonstrated definitivedetection of a pulmonary tumor on day 5, which was consis-tent with MRI imaging, bioluminescence, and primary tumorlocation at later time points. These results suggest a role forNTR/CytoCy5S in multimodal imaging applications.Finally, we investigated NTR/CytoCy5S-gated imaging of the

MDA-MB-231NTRþLucþGFP metastatic mammary carcinoma

model (Fig. 4). A total of 1 � 106 cells were injected orthoto-pically into the mammary fat pad of NSG mice (n ¼ 4) andNTR/CytoCy5S-gated fluorescence images acquired on aweek-ly basis. Previously, axial lymph node metastasis has beenformerly shown by in vivo optical imaging of the MDA-MB-231 orthotopic breast cancer model (27, 28). NTR/CytoCy5S-gated fluorescence images revealed progressive primary tumorfluorescence up toweek 7 (Fig. 4C)with initial axial lymphnodemetastasis atweek 8 (see arrowFig. 4A). Detection of additionalmetastasis in this region was observed by week 9 (see inset). Atweek 12, multiple tumors along the draining lymphatic vesselwere noted and a further distal auxiliary lymphatic metastasis.Interestingly, fluorescence was also observed in the liver with afaint area of fluorescence in the upper thorax (Fig. 4B). Ex vivoimaging following necropsy revealed fluorescence associatedwith primary tumor section, several lymph node tumors alongthe draining lymphatic vessel, in addition to liver fluorescenceand small foci in the lungs (Fig. 4D and E). Microscopicexamination of histology sections from these organs confirmedlymph and liver metastasis (Fig. 4F).

Figure 2. In vivo imaging of disseminated leukemiawithNTR/CytoCy5S. A, leukemiawas detectable inmice (n¼ 4) as early as 5 to 7days following intravenousinoculation with 5 � 106 IPC-81NTRþGFPþ cells, with time-domain NTR/CytoCy5S imaging, whereas (B) IPC-81WT remained undetectable. C, analogousdiscrimination of leukemia was only possible after 17 to 24 days with imaging of GFP expression. D, Images present the entire scan area andfluorescence quantifications determined from these images. E, histology (H&E staining) of femurs [scale bar, 200 mm for large image and 100 mm for thefemur magnification (right)]. F, similarly, earlier detection of NB4NTRþGFPþ leukemia (5 � 106 cells per mouse; n ¼ 7) in spinal and femoral bone marrowwas possible by imaging NTR/CytoCy5S fluorescence (white circles). G, ex vivo images and H&E histology from spinal, ovarian, and lymph nodetumors from 2 NB4NTRþGFPþ mice (scale bar, 100 mm). All NTR/CytoCy5S fluorescence images were acquired 30 minutes following CytoCy5S injection. Allfluorescence images are presented on Log scale.

NTR Optical Imaging of Metastatic Cancer

www.aacrjournals.org Cancer Res; 73(4) February 15, 2013 1281

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 7: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

Gene-directed enzyme prodrug therapy evaluation withNTR/CytoCy5S

NTR is typically used as the activating enzyme in the gene-directed enzyme prodrug therapy (GDEPT) approach to cancerchemotherapy. We therefore investigated the use of the NTR/CytoCy5S platform for active imaging of GDEPT (Fig. 5). Thenitroimidazole, metronidazole (Fig. 5A), has previously beendescribed as an NTR substrate in GDEPT therapeuticapproaches (29) and conditional cell ablation in Zebrafish(30). Incubation of NB4 wt or NB4NTRþGFPþ cells with increas-ing concentrations of metronidazole (0.3–2.4 mmol/L) for 24hours resulted in NTR-dependent cell death, evaluated bymicroscopic examination following Hoechst 33342 (P <0.01–0.05; Fig. 5B), flow cytometry with AnnexinV/PI viabilityassay (P < 0.001–0.05; Supplementary Fig. 5A and B), and 3H-thymidine incorporation proliferative assay (P < 0.01; Supple-mentary Fig. 5C). Similar results were observed with adherentcell lines expressingNTR (Supplementary Fig. S5D and S5E). Toassess any potential metronidazole-competitive inhibition ofCytoCy5S reduction by NTR via decrease in fluorescenceoutput, NB4NTRþGFPþ cells were incubated � metronidazole(0.6 mmol/L) for 2 hours with CytoCy5S for the last hour andfluorescence evaluated by flow cytometry (Fig. 5C). No differ-ences were observed in median NTR/CytoCy5S fluorescenceintensity � metronidazole. Subsequently, NSG mice bearing

subcutaneous NB4NTRþGFPþ tumors were randomized (P ¼0.24) into control (vehicle, 134 � 15 mm3, n ¼ 7) or metroni-dazole-treated (50 mg/kg, b.i.d., 107 � 25, n ¼ 8) groups andtreated for 10 consecutive days (Fig. 5D). Caliper measure-ments of tumor volume showed significant inhibition of tumorgrowth between the NB4NTRþGFPþ groups (P < 0.01–0.05) from2 days following initiation of treatment through to terminationof the study (Fig. 5E). Similarly, NTR/CytoCy5S-gated fluores-cence images correlated well with caliper measurements (Fig.5H; Pearson r¼ 0.88), with treated mice illustrating significantdifferences in fluorescence intensities throughout treatment(Fig. 5F; P < 0.001–0.05). NSG mice xenografted with the NTR�

cell line, NB4GFPþ, subcutaneously were similarly divided (P¼0.29) into control (95.9 � 16 mm3, n ¼ 4) and metronidazole(125 � 17 mm3, n ¼ 4) groups and treated analogously. Nosignificant differences in tumor progression were noted (Fig.5G). These results establish NTR/CytoCy5S-gated fluorescenceimaging as a powerful gene reporter imaging technique per-mitting temporal monitoring of NTR-targeted GDEPT efficacy.

DiscussionClear delineation of disseminated or metastatic cancer

lesions in preclinical xenografts, particularly when metastaticsites are not known a priori, is often problematic with

Figure 3. In vivo imaging oforthotopic, metastatic lungcarcinoma xenografts (n ¼ 5) withNTR/CytoCy5S. NTR/CytoCy5Sfluorescence (A) andbioluminescence (B) images ofNCI-H460NTRþLucþGFPþ orthotopicxenografts 5, 8, and 14 days afterimplant. Images represent theentire scan areas and from wherefluorescence was quantified. AllNTR/CytoCy5S fluorescenceimages were acquired 30 minutesfollowing CytoCy5S and 10minutes following D-Luciferinadministration. C, axial and sagittalT1-weighted FLASH images oftumor location in the left lung(red circle). D and E, ex vivofluorescence (D) andbioluminescence (E) images ofexcised lungs. F, NTR/CytoCy5Sfluorescence time course of NCI-H460NTRþLucþGFPþ orthotopicxenografts (n ¼ 5) quantified fromthe entire scan area. Error barsrepresent SEM. G and H, H&Ehistology show extensivepulmonary metastasis (scale bars,1 mm for G and 200 mm for H).Data presented from 2 mice(n¼ 1, optical imaging; n¼ 1, MRI).Results were repeated in 5independent experiments.

McCormack et al.

Cancer Res; 73(4) February 15, 2013 Cancer Research1282

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 8: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

application of exogenous fluorochromes. Far-red or near-infrared reporter gene labeling of cancer cells better suit suchwhole body imaging applications (31). Accordingly, we expectwhole-body, time-domain imaging of NTR reporter geneexpression to broaden the possibilities of noninvasive opticalimaging and be a practical and useful addition to currentfluorescence and bioluminescence methodologies.The current study has not presented data comparing red

fluorescence protein andNTR/CytoCy5S imaging and there areseveral reasons for this. Despite innovative engineering of far-red proteins with the specific aim of deep tissue whole bodyimaging, their adaption to macroscopic imaging has generallybeen limited thus far (32). A recent study concluded thatdespite increases in detected fluorescence gained from switch-ing to red-shifted protein from GFPs, benefits were compro-mised by increases in autofluorescence in transilluminationgeometry (33). While we anticipate that time-domain imagingof infrared fluorescent protein (iRFP; ref. 11) will amelioratefluorescence detection, a report comparing time-domainimaging of the far-red Katushka and GFP concluded thatlongitudinal imaging was superior with GFP (34). We haveshown here the superiority of the NTR/CytoCy5Smethodologycompared with GFP (Fig. 2) and the possibility for multimodalimaging with NTR/CytoCy5S and fluorescent proteins.A caveat to the NTR reporter platform is the necessity of a

fluorogenic substrate such as CytoCy5S. However, substrateinjection has not prevented the luciferases in becoming the

most commonly used preclinical optical reporter gene (12, 35).Furthermore, the rapid induction of fluorescence and consis-tency of the NTR/CytoCy5S signal over several hours (Supple-mentary Fig. S3) makes this imaging platform very attractivefor raster scanning imaging techniques, which typically requirelong timeframes of 5 to 20 minutes to acquire whole bodyimages of a single mouse. Undoubtedly, a significant benefit ofthe NTR/CytoCy5S platform as shown here has been thesensitivity of the imaging technique to detect disseminatedand metastatic cancer cells in organs of high blood volume.Here, we have verified the sensitivity of the NTR/CytoCy5Simaging platform to detect metastasis in the liver (Fig. 4) inaddition to leukemia in bone marrow (Fig. 2) confirmed by exvivo imaging and histology. Indeed, an intriguing future per-spective may include use of the NTR/CytoCy5S reporter plat-form to image the in vivo leukemogenesis of leukemic stemcells, disease evolution following their ablation by GDEPT, inaddition to NTR/CytoCy5S-mediated differential populationsorting, and quantification by flow cytometry. Further evolu-tion of the methodology using further near-infrared–shiftedfluors such as Cy5.5 and Cy7 may yield multiple substrates formultimodal optical imaging of NTR gene expression.

In summary, by combining the NTR enzyme and itsquenched fluorogenic substrate CytoCy5S, with time-domainoptical imaging, we have generated a sensitive, near-infraredapproach permitting preclinical imaging of orthotopic andmetastatic tumors. Validation of the method in several

Figure 4. In vivo NTR/CytoCy5Sfluorescence imaging of metastaticbreast carcinoma. A, NTR/CytoCy5Sfluorescence imaging of MDA-MB-231NTRþLucþGFPþorthotopicxenografts from week 8. Evidence ofprimarily axial lymph nodemetastasis is evident from week 9.Images present the entire scan area.B, imaging of NTR/CytoCy5Sfluorescence shows detection ofaxial lymph node metastasis fromweek 8 (arrow) with detection ofbrachial lymph metastasis on week9. By week 12, multiple metastasisare evident in the lymphoid organsand liver (white arrows). C, NTR/CytoCy5S fluorescencequantification of tumor growth fromthe entire scan area before week 8(n¼ 4). Error bars represent SEM. D,ex vivo NTR/CytoCy5S fluorescenceimages of suspected metastasis inorgans described in E. F, H&Ehistologies reveal the presence ofMDA-MB-231 cells (arrowheads) inprimary, lymph, and liver tissues(scale bar, 400 mm and 100 mm formagnified views). All NTR/CytoCy5Sfluorescence images acquired 30minutes following CytoCy5Sinjection.

NTR Optical Imaging of Metastatic Cancer

www.aacrjournals.org Cancer Res; 73(4) February 15, 2013 1283

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 9: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

Figure 5. In vivo NTR/CytoCy5Sfluorescence imaging of gene-directed enzyme prodrug therapy.A, metronidazole is anitroimidazole substrate of NTR,whose reduction products destroyhelical DNA structure. B, only NTR-expressing NB4 cells are sensitiveto metronidazole as assayed bynuclear morphology cell deathassay (Hoechst 33342). C,incubation of NB4NTRþGFPþ cellswith CytoCy5S in the presence ofmetronidazole (MTZ) does not alterfluorescence by flow cytometry.D, representative NTR/CytoCy5Sfluorescence images of miceimplanted with NB4NTRþGFPþ cells(5�106) s.c. and treatedwith eitherMTZ (n¼8 tumors, 50mg/kg, twicea day) for 10 days or vehiclecontrol (n ¼ 7 tumors). The shavedhalf of themice represents the scanareas and from where NTR/CytoCy5S-gated fluorescencewas quantified. E, tumor volumemeasurements correlated well withresults observed fromfluorescence imaging. F, followinginitiation of therapy, treated miceshowed significantly lessfluorescence than vehicle controlsuntil termination of treatment.G, mice implanted with NB4GFPþ

and treated analogously toNB4NTRþGFPþ bearing miceshowed no significant differencesin tumor growth. H, correlation oftumor volume andNTR/CytoCy5S-gated fluorescence. All NTR/CytoCy5S fluorescence imagesacquired 30 minutes followingCytoCy5S injection. PC, photoncounts. (�, P < 0.05; ��, P < 0.01;���, P < 0.001). Error Barsrepresent SEM.

McCormack et al.

Cancer Res; 73(4) February 15, 2013 Cancer Research1284

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 10: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

preclinical models of disseminated, orthotopic, andmetastaticcancers showed sensitive primary tumor and metastasis visu-alization. Furthermore, we established that the NTR/CytoCy5Sreporter platform could be used to image GDEPT therapeuticefficacy concomitantly with prodrugs, without interference.This is an important aspect to the current study. While NTR isa promising clinical candidate for GDEPT cancer therapy(36–38), a caveat to preclinical development of NTR-basedGDEPT strategies has been an inability to visualize NTRþ cellsunder GDEPT. We believe use of this platform will accelerateGDEPT interventions towards clinical development and mayprovide the basis for a positron emission tomography–basedradiotracer suitable for clinical imaging of NTR-based GDEPTapproaches.

Disclosure of Potential Conflicts of InterestR.M. West is employed (other than primary affiliation; e.g., consulting) in GE

Healthcare as R&D scientist and has ownership interest (including patents) inUS7579140 B2. No potential conflicts of interest were disclosed by the otherauthors.

Authors' ContributionsConception and design: E. McCormack, E. Silden, R.M. West, D.R. Micklem,J. Lorens, M.E. Cooper, B.T. GjertsenDevelopment of methodology: E. McCormack, E. Silden, D.R. Micklem,J. LorensAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): E. McCormack, E. Silden, T. Pavlin, J. Lorens, B.E.Haug

Analysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): E. McCormack, E. Silden, B.E. Haug, B.T. GjertsenWriting, review, and/or revision of the manuscript: E. McCormack,E. Silden, R.M. West, B.E. Haug, M.E. Cooper, B.T. GjertsenAdministrative, technical, or material support (i.e., reporting or orga-nizing data, constructing databases): E. McCormack, E. Silden, R.M. West,B.T. GjertsenStudy supervision: E. McCormack, E. Silden, M.E. Cooper, B.T. Gjertsen

AcknowledgmentsThe authors thankM. Popa, L. Vikebø, M. Boge, K. Jacobsen, T. Osdal, and L.E.

Pindard for expert assistance in all preclinical work; M. Enger, A. Sulen, and R.Brendsdal Forthun for flow cytometry assistance; and E. Fick for histologyprocessing. The authors also thankM. Briggs and R. Ismail for critical discussionof the manuscript and A.Molven for cell line validation and fingerprinting. Allimaging and flow cytometry sorting was conducted at the Molecular ImagingCenter (MIC), the Department of Biomedicine, University of Bergen (Bergen,Norway).

Grant SupportThis work was supported by the Norwegian Cancer Society (grant number

421828, 732200); the Western Health Board of Norway (grant number 911182),Bergen Research Foundation and MedViz (www.medviz.uib.no), a medicalimaging and visualization R&D cluster in Western Norway founded by Hauke-land University Hospital, University of Bergen, and Christian MichelsenResearch.

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 July 7, 2012; revised November 28, 2012; accepted November 29, 2012;published OnlineFirst December 10, 2012.

References1. Weissleder R, Pittet MJ. Imaging in the era of molecular oncology.

Nature 2008;452:580–9.2. Willmann JK, van Bruggen N, Dinkelborg LM, Gambhir SS. Molecular

imaging in drug development. Nat Rev Drug Discov 2008;7:591–607.3. McCormack E, Micklem DR, Pindard LE, Silden E, Gallant P, Belenkov

A, et al. In vivo optical imaging of acute myeloid leukemia by greenfluorescent protein: time-domain autofluorescence decoupling, fluor-ophore quantification, and localization. Mol Imaging 2007;6:193–204.

4. Luo S, Zhang E, Su Y, Cheng T, Shi C. A review of NIR dyes in cancertargeting and imaging. Biomaterials 2011;32:7127–38.

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

6. KumarAT,ChungE,RaymondSB, vandeWater JA,ShahK, FukumuraD, et al. Feasibility of in vivo imaging of fluorescent proteins usinglifetime contrast. Opt Lett 2009;34:2066–8.

7. Campbell RE, Tour O, Palmer AE, et al. A monomeric red fluorescentprotein. Proc Natl Acad Sci U S A 2002;99:7877–82.

8. Shaner NC, Campbell RE, Steinbach PA, Giepmans BN, Palmer AE,Tsien RY. Improved monomeric red, orange and yellow fluorescentproteins derived from Discosoma sp. red fluorescent protein. NatBiotechnol 2004;22:1567–72.

9. Wang L, Jackson WC, Steinbach PA, Tsien RY. Evolution of newnonantibody proteins via iterative somatic hypermutation. Proc NatlAcad Sci U S A 2004;101:16745–9.

10. Merzlyak EM, Goedhart J, Shcherbo D, Bulina ME, Shcheglov AS,Fradkov AF, et al. Bright monomeric red fluorescent protein with anextended fluorescence lifetime. Nat Methods 2007;4:555–7.

11. Filonov GS, Piatkevich KD, Ting LM, Zhang J, Kim K, Verkhusha VV.Bright and stable near-infrared fluorescent protein for in vivo imaging.Nat Biotechnol 2011;29:751–61.

12. Prescher JA, Contag CH. Guided by the light: visualizing biomolecularprocesses in living animalswithbioluminescence.CurrOpinChemBiol2010;14:80–9.

13. Searle PF, Chen MJ, Hu L, Race PR, Lovering AL, Grove JI, et al.Nitroreductase: a prodrug-activating enzyme for cancer gene therapy.Clin Exp Pharmacol Physiol 2004;31:811–6.

14. Cui L, Zhong Y, Zhu W, Xu Y, Du Q, Wang X, et al. A new prodrug-derived ratiometric fluorescent probe for hypoxia: high select-ivity of nitroreductase and imaging in tumor cell. Org Lett 2011;13:928–31.

15. Thorne SH, Barak Y, LiangW, BachmannMH, Rao J, Contag CH, et al.CNOB/ChrR6, a new prodrug enzyme cancer chemotherapy. MolCancer Ther 2009;8:333–41.

16. Cellier M, Fabrega OJ, Fazackerley E, James AL, Orenga S, Perry JD,et al. 2-Arylbenzothiazole, benzoxazole and benzimidazole derivativesas fluorogenic substrates for the detection of nitroreductase andaminopeptidase activity in clinically important bacteria. Bioorg MedChem 2011;19:2903–10.

17. James AL, Perry JD, Jay C, Monget D, Rasburn JW, Gould FK.Fluorogenic substrates for the detection of microbial nitroreductases.Lett Appl Microbiol 2001;33:403–8.

18. Bhaumik S, Sekar TV, Depuy J, Klimash J, Paulmurugan R. Noninva-sive optical imaging of nitroreductase gene-directed enzyme prodrugtherapy system in living animals. Gene Ther 2012;19:295–302.

19. de la Zerda A, Bodapati S, Teed R, Schipper ML, Keren S, Smith BR,et al. A comparison between time domain and spectral imagingsystems for imaging quantum dots in small living animals.Mol ImagingBiol 2010;12:500–8.

20. KerenS,GheysensO, LevinCS,Gambhir SS. A comparison between atime domain and continuous wave small animal optical imaging sys-tem. IEEE Trans Med Imaging 2008;27:58–63.

21. Lorens JB, Bennett MK, Pearsall DM, Throndset WR, Rossi AB,ArmstrongRJ, et al. Retroviral delivery of peptidemodulators of cellularfunctions. Mol Ther 2000;1:438–47.

22. Lorens JB, Pearsall DM, Swift SE, Peelle B, Armstrong R, Demo SD,et al. Stable, stoichiometric delivery of diverse protein functions.J Biochem Biophys Methods 2004;58:101–10.

NTR Optical Imaging of Metastatic Cancer

www.aacrjournals.org Cancer Res; 73(4) February 15, 2013 1285

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 11: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

23. LorensJB, JangY,Rossi AB,PayanDG,Bogenberger JM.Optimizationof regulated LTR-mediated expression. Virology 2000;272:7–15.

24. Gjertsen BT, Cressey LI, Ruchaud S, Houge G, Lanotte M, DoskelandSO. Multiple apoptotic death types triggered through activation ofseparate pathways by cAMP and inhibitors of protein phosphatases inone (IPC leukemia) cell line. J Cell Sci 1994;107 (Pt 12):3363–77.

25. McCormack E, Haaland I, Venas G, Forthun RB, Huseby S, GausdalG, et al. Synergistic induction of p53 mediated apoptosis by val-proic acid and nutlin-3 in acute myeloid leukemia. Leukemia 2012;26:910–7.

26. West RM, Ismail Rinventors; GE Healthcare UK Limited, Amersham(GB), assignee. Method and reagent for measuring nitroreductaseenzyme activity. United States patent. US 7,579,140 B2. 2009.

27. Gjerdrum C, Tiron C, Hoiby T, Stefansson I, Haugen H, Sandal T, et al.Axl is an essential epithelial-to-mesenchymal transition-induced reg-ulator of breast cancer metastasis and patient survival. Proc Natl AcadSci U S A 2010;107:1124–9.

28. Liu J, Liao S, Huang Y, Samuel R, Shi T, Naxerova K, et al. PDGF-Dimproves drug delivery and efficacy via vascular normalization, butpromotes lymphatic metastasis by activating CXCR4 in breast cancer.Clin Cancer Res 2011;17:3638–48.

29. Bridgewater JA, Knox RJ, Pitts JD, Collins MK, Springer CJ. Thebystander effect of the nitroreductase/CB1954 enzyme/prodrug sys-tem is due to a cell-permeable metabolite. Hum Gene Ther 1997;8:709–17.

30. Willems B, Buttner A, Huysseune A, Renn J, Witten PE, Winkler C.Conditional ablation of osteoblasts in medaka. Dev Biol 2012;364:128–37.

31. Shcherbo D, Shemiakina II, Ryabova AV, et al. Near-infrared fluores-cent proteins. Nat Methods 2010;7:827–9.

32. Hoffman RM. Imaging cancer dynamics in vivo at the tumor andcellular level with fluorescent proteins. Clin Exp Metastasis 2009;26:345–55.

33. Deliolanis NC, Wurdinger T, Pike L, Tannous BA, Breakefield XO,Weissleder R, et al. In vivo tomographic imaging of red-shifted fluo-rescent proteins. Biomed Opt Express 2011;2:887–900.

34. Hojman P, Eriksen J, Gehl J. In vivo imaging of far-red fluorescentproteins after DNA electrotransfer tomuscle tissue. Biol ProcedOnline2009;11:253–62.

35. ZhaoH, Doyle TC, CoquozO, Kalish F, Rice BW, ContagCH. Emissionspectra of bioluminescent reporters and interaction with mammaliantissue determine the sensitivity of detection in vivo. J Biomed Opt2005;10:41210.

36. Patel P, Young JG,Mautner V, AshdownD,BonneyS, PinedaRG, et al.A phase I/II clinical trial in localized prostate cancer of an adenovirusexpressing nitroreductase with CB1954 [correction of CB1984]. MolTher 2009;17:1292–9.

37. Palmer DH, Mautner V, Mirza D, Oliff S, Gerritsen W, van der Sijp JR,et al. Virus-directed enzyme prodrug therapy: intratumoral admin-istration of a replication-deficient adenovirus encoding nitroreduc-tase to patients with resectable liver cancer. J Clin Oncol 2004;22:1546–52.

38. Onion D, Patel P, Pineda RG, James N, Mautner V. Antivector andtumor immune responses following adenovirus-directed enzyme pro-drug therapy for the treatment of prostate cancer. Hum Gene Ther2009;20:1249–58.

McCormack et al.

Cancer Res; 73(4) February 15, 2013 Cancer Research1286

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649

Page 12: Nitroreductase,aNear-InfraredReporterPlatformfor …...Time-Domain Optical Imaging of Metastatic Cancer EmmetMcCormack 1 ,ElisabethSilden 1 ,RichardM.West 5 ,TinaPavlin 2 ,DavidR.Micklem

2013;73:1276-1286. Published OnlineFirst December 10, 2012.Cancer Res   Emmet McCormack, Elisabeth Silden, Richard M. West, et al.   Time-Domain Optical Imaging of Metastatic Cancer

In VivoNitroreductase, a Near-Infrared Reporter Platform for

  Updated version

  10.1158/0008-5472.CAN-12-2649doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://cancerres.aacrjournals.org/content/suppl/2012/12/10/0008-5472.CAN-12-2649.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/73/4/1276.full#ref-list-1

This article cites 37 articles, 7 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/73/4/1276.full#related-urls

This article has been cited by 3 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://cancerres.aacrjournals.org/content/73/4/1276To request permission to re-use all or part of this article, use this link

on July 12, 2020. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org Downloaded from

Published OnlineFirst December 10, 2012; DOI: 10.1158/0008-5472.CAN-12-2649