Open Access
1 January 2008 In vivo optical imaging of CD13/APN-expression in tumor xenografts
Angelika von Wallbrunn, Jens Waldeck, Carsten Hoeltke, Michael Zühlsdorf, Rolf Mesters, Walter L. Heindel M.D., Michael Schäfers, Christoph Bremer
Author Affiliations +
Abstract
The metalloexopeptidase CD13/aminopeptidase N (APN) has been shown to be involved in cancer angiogenesis, invasion, and metastasis. Therefore, a CD13/APN–targeted NGR-peptide was labeled with the cyanine dye Cy 5.5 and applied to image tumor xenografts with different APN-expression levels using both planar and tomographic optical imaging methods. In vitro, the peptide-dye conjugate showed a clear binding affinity to APN-positive HT-1080 cells, while negative MCF-7 cells and predosing with the free NGR-peptide revealed little to no fluorescence. In vivo, tumor xenografts (n≥5) were clearly visualized by two-dimensional (2-D) planar fluorescence reflectance imaging (FRI) and three-dimensional (3-D) fluorescence mediated tomography (FMT) up to 24 h after injection. FMT also allowed us to quantify fluorochrome distribution in deeper tissue sections, showing an average fluorochrome concentration of 306.7±54.3 nM Cy 5.5 (HT-1080) and 116.0±18.3 nM Cy 5.5 (MCF-7) in the target tissue after 5 h. Competition with the free NGR-peptide resulted in a reduction of fluorochrome concentration in HT-1080 tumor tissue (195.3±21.9 nM; 5 h). We thus conclude that NGR-Cy 5.5 combined with novel tomographic optical imaging methods allows us to image and quantify tumor-associated CD13/APN expression noninvasively. This may be a promising strategy for a sensitive evaluation of tumor angiogenesis in vivo.

1.

Introduction

Optical imaging is a desirable technique for in vivo cancer diagnostics since it is a highly sensitive, inexpensive imaging method that can potentially resolve relevant oncological target structures in vivo.1 With the injection of a fluorescent compound (fluorochrome), optical imaging allows specific tagging of particular receptors, antibodies, genes, or drugs, thereby measuring compound biodistribution and pharmacokinetics and hence leading to a better understanding of these processes.1, 2, 3, 4 Combining new quantitative fluorescence imaging methods with target specific fluorochromes facilitates the visualization and quantification of specific receptor expression in vivo. Especially, fluorescence mediated tomography (FMT) as a newly emerging optical imaging technique has been shown to be capable of resolving fluorescence signatures three-dimensionally and quantitatively.5, 6, 7, 8, 9

The CD13/aminopeptidase N (APN) is a metalloexopeptidase that removes unsubstituted, N-terminal amino acids with neutral side chains (Ala> Phe> Leu> Gly) from peptides and amide or arylamide derivatives of amino acids.10 APN has been shown to be involved in cancer angiogenesis, invasion, and metastasis.11, 12, 13 A unique peptide that contains the NGR (asparagine-glycine-arginine)-motive binds to APN in tumor vasculature.12, 14 Tumor homing peptides containing the NGR motive, such as CNGRC and GNGRG, have been used for delivering cytokines, chemotherapeutic drugs, apoptotic peptides, and liposomes to a CD13 isoform, highly secreted on the surface of various tumor cells and tumor blood vessels. 12, 14, 15, 16, 17, 18, 19, 20 Taking this into account, we chose a cyclic NGR peptide labeled with the cyanine dye Cy 5.5 for measuring and quantifying tumor homing of the conjugate with FMT, thereby comparing this technique with conventionally used fluorescence reflectance imaging (FRI) methods. Specific tumor targeting of the cyclo-[Cys-Asn-Gly-Arg-Cys]-Gly-Lys-Cy 5.5 conjugate (NGR-Cy 5.5) was measured on HT-1080 fibrosarcoma and MCF-7 adenocarcinoma cell assays in vitro and on tumor xenografts in vivo.

2.

Materials and Methods

2.1.

Synthesis and Characterization of NGR-Cy 5.5

Peptides were purchased as trifluoroacetate (TFA) salts in > 90% purity from Bachem Distribution Services GmbH, Weil am Rhein, Germany. Cy 5.5 monoreactiv NHS-ester was purchased from Amersham Biosciences Europe GmbH, Freiburg, Germany. All chemicals were used as purchased without further purification. Peptides were dissolved in aqueous bicarbonate buffer ( 0.1M , pH 8.4; 1.3mg500μL ) at room temperature. To this, a solution of the dye in DMSO (1.9mg200μL) was added and stirred for 60min at room temperature in the dark. Purification was accomplished by gradient HPLC using a Knauer system with two K-1800 pumps, an S-2500 UV-detector and a RP-HPLC Nucleosil 100-5 C18 column (eluent A: water, 0.1% TFA; eluent B: acetonitrile, 0.1% TFA). Elution times were 40to60min , dependent on solution quantity, at a flow rate of 1mLmin ). The product fractions were pooled, lyophilized, redissolved in phosphate-buffered saline (PBS) at 1.0mg500μL , and frozen at 20°C . The identities of the labeled products were confirmed by high-resolution electrospray mass spectrometry (HR-ES-MS) on a QUATTRO LCZ (Waters Micromass, Manchester, United Kingdom) spectrometer with a nanospray capillary inlet, mz=557.5[M]3 , 837.0[M+H]2 . The fully assembled tracer was assessed by fluorometry and photometry for spectral characteristics (Hitachi, F-4500 fluorescence spectrometer and U-3310 UV/VIS spectrophotometer, Tokyo, Japan).

2.2.

Cell Lines and Reagents

Human fibrosarcoma HT-1080 (ATCC: CCL-1321, Manassas, Virginia) and human adenocarcinoma MCF-7 (ATCC: HTB-22, Manassas, Virginia) cells were cultured in RPMI-1640 or MEM (Invitrogen Corporation, San Diego, California) supplemented with 10% fetal calf serum, penicillin and streptomycin (Biochrom AG, Berlin, Germany). Cells were grown routinely in a monolayer culture at 37°C in a 5% CO2 humidified air atmosphere. All antibodies were specific for CD13/APN: clone WM15 antibodies (phycoerythrin- and unconjugated) were from Pharmingen (BD Biosciences, San Jose, California), and antibody H-300 for Western blotting and immunohistochemistry was from Santa Cruz Biotechnology (Santa Cruz, California).

2.3.

FACS Analysis by Flow Cytometry

Cells were washed with PBS and subsequently harvested in 5mL Versen-buffer ( 13.7mM NaCl, 10mM EDTA, 2.6mM KCl, 8.1mM NaH2PO4 , and 1.4mM KH2PO4 , pH 7.2). Aliquots of 2.5×105 cells were blocked with mouse IgG in PBS/BSA (0.1%) for 15min at 4°C , washed twice with PBS with Ca2+ and Mg2+ , and resuspended in 150μL binding buffer ( 150mM NaCl, 10mM MgCl2 , 10mM CaCl2 , 0.5mM MnCl2 , pH 7.2, diluted 1:20 in PBS/BSA 0.1%). Antibodies (3μgmL) were added, and cells were incubated for 45min at 4°C . Cells were washed, resuspended in PBS with 0.1% BSA, and analyzed on a Becton Dickinson FACSCalibur (BD Biosciences, San Diego, California).

2.4.

Western Blot

Confluent (80 to 100%) HT-1080 and MCF-7 cells were treated with 0.5mL trypsin and harvested by scraping in cold PBS. After two centrifugation steps ( 1500rpm , 5min , 4°C ), cells were resuspended in lysis buffer [0.5% Tween 20, 50mM Tris-HCl pH 8, 250mM NaCl, 5mM EDTA pH 8, 50mM NaF, 0.5mM Na3VO4 , 0.9mgml protease inhibitor cocktail (Sigma, St. Louis, Missouri)], and the protein concentration of each sample was determined. To concentrate the protein, a two-fold volume of cold acetone was added to each sample and placed in 20°C for 16h , followed by centrifugation at 10,000g , 4°C for 30min . Protein pellets were dissolved in 100μL of 6× sodium dodecyl sulfate (SDS) sample buffer (New England Biolabs, Beverly, Massachusetts). Following precipitation, proteins were electrophoretically separated in nonreducing 10% SDS-PAGE and transferred to PVDF membranes (Millipore Corporation, Bedford, Massachusetts). After blocking, immunoblots were incubated with CD13/APN polyclonal antibody H-300 and with peroxidase-conjugated goat anti-rabbit IgG secondary antibody (Sigma, St. Louis, Missouri). Peroxidase activity was revealed using ECL chemiluminescence (Amersham, Buckinghamshire, United Kingdom) and x-ray film, as described by the manufacturers. THP-1 cell lysat (BD Biosciences, San Diego, California) was used as a positive control.

Tumor tissue was frozen in liquid nitrogen, and 0.1to0.5g of each tissue was homogenized by ultrasound using an Ultra-Turrax T8 (IKA-Labortechnik, Staufen, Germany). The homogenate was resuspended in 300μL lysis buffer, additionally sonicated twice at 20kHz for 5s , and centrifuged for 30to60min at 10,000g to remove cell debris. The supernatant was taken for determining protein concentration following SDS-PAGE and Western blotting.

2.5.

Aminopeptidase N Activity Assay

Surface aminopeptidase activity of intact cells was measured by plating 5000 cells/well in a 96-well plate (Lumox-Multiwell, Greiner Bio-One GmbH, Frickenhausen, Germany). Cells were allowed to recover for 24h , followed by washing with PBS and incubating with 100μM L-alanine-4-methyl-7-coumarinylamide-(MCA)-trifluoroacetate (Fluka) in PBS with 0.5% BSA and 20mM HEPES (pH 7.2).21 The release of fluorescent product 7-amido-4-methylcoumarin was monitored on a Kodak Gel Logic 200 Imaging System (Kodak, New Haven, Connecticut), with λexc of 360nm and λem of 460nm after 90min . To assay for inhibition of enzymatic activity due to binding of CD13-specific antibody WM15 or the NGR-peptide, cells were pretreated in PBS containing a excess of antibody (5μgmL) or peptide (25μgmL) at 37°C for 1h .

2.6.

In Vitro CD13/APN Ligand Binding Studies

Cells were seeded in 24-well plates and incubated in culture medium overnight. For binding studies, cells were washed twice with PBS and resuspended in 150μL cation-buffer diluted 1:20 in PBS/BSA (0.1%). Cy 5.5 (up to 30μM ) or NGR-Cy 5.5 (12.5μM) was added into each well. After an incubation period of 1h at 37°C , cells were washed twice with PBS and resuspended in binding buffer. For blocking studies, free NGR-peptide (125μM) was added to each well for 45min at 37°C . Cells could be directly visualized by fluorescence microscopy ( 40× objective magnification, Nikon TE 2000-S, Tokyo, Japan). The microscope was equipped with a mercury vapor lamp, 620775nm and 545675nm (excitation/emission) filters (AHF Analysentechnik, Tuebingen, Germany), a Nikon DXM1200F camera, and ACT1/DXM1200F software (Nikon, Tokyo, Japan).

2.7.

Immunohistochemistry

Tumors were removed and embedded in buffered formalin (4%, pH 7.0). Immunohistochemistry has been carried out commercially by Habedank-LID (Berlin, Germany) using the specific anti–CD13mAb H-300. For the in vivo visualization of NGR-Cy 5.5 uptake in tumor tissue, tumors were removed 24h p.i., embedded in Tissue-Tek O.C.T. (Sakura, Zoeterwoude, Netherlands), snap-frozen, and serial-sectioned (10μm) . After drying, tissue fluorescence was evaluated by fluorescence microscopy.

2.8.

Tumor Xenografts

All animal studies were approved by the institutional review boards (# G76/2005). Female athymic nude mice (nunu) were obtained from Charles River Laboratories (Sulzfeld, Germany). At 4to6weeks of age, 3×106 HT-1080 or MCF-7 cells suspended in 100μL PBS and Matrigel (BD Biosciences, San Diego, California) were injected subcutaneously in the flank and thoracic wall, respectively. When tumors reached 4to6mm in diameter, the tumor-bearing mice were anesthetized by an i.p. injection of ketamine ( 125mgkg bw) and xylazine ( 12.5mgkg bw) and subjected to in vivo imaging studies.

2.9.

In Vivo Optical Imaging Systems – Fluorescence Reflectance Imaging

The combination of x-ray and optical fluorescence reflectance imaging (FRI) was performed using the Image Station In Vivo FX Imaging System (Kodak Molecular Imaging Systems, New Haven, Connecticut), which is equipped to provide multiwavelength fluorescence, luminescence, x-ray, and radioisotopic imaging. Fluorescence is provided by a 150-W halogen illuminator with selectable bandpass excitation and emission filters. For Cy 5.5 excitation, 625±18nm and emission 700±17.5nm were employed. X-ray energy is provided by a 35-Kvp , 150-μamp , microfocus source in an enclosed chamber above the animal. A specialized radiographic phosphor screen is moved into the imaging path under the animal, which transduces the x-ray energy to light. Light from the fluorescence and/or the radiographic screen is captured with the 4-million-pixel cooled CCD camera equipped with a 10× zoom lens. Images were captured 1, 3, 5, and 24h after probe injection. Image acquisition times were 30s per animal at the emission wavelength. Optical images were co-registered with x-ray background images, and regions of interest were selected and analyzed with the Kodak MI 4.0 software.

2.J.

Fluorescence Mediated Tomography

All tomographic optical imaging studies were performed with a small-animal fluorescence mediated tomography (FMT) system from VisEn Medical, Inc. (Woburn, Massachusetts) that has been described in detail elsewhere.6 The central piece of the system is the animal-imaging chamber configured in a parallel plate geometry. The chamber is scanned by a movable optical fiber that can excite a user-defined matrix of typically 50,000 to 100,000 source-detector projection pairs. The light source is software-selectable from two high-power laser diodes at 670nm and 745nm , operating in the range of 5mWto150mW . Detection is performed with a thermoelectrically cooled CCD camera by means of an imaging lens and appropriate bandpass filters (three-cavity interference at 675nm or 755nm for excitation measurements, and 705nm or 770nm for emission measurements). Tomographic reconstruction is based on the normalized Born approximation, a diffraction optical tomographic technique that uses a diffusion-type theoretical photon propagation model in biological tissue. The underlying assumption of the mathematical model has been propagation in a homogeneous diffuse medium with average optical absorption and scattering properties those of small rodents at the relevant wavelengths. However, it has been demonstrated that because of the appropriate data normalization scheme applied, the algorithm is very accurate even in highly heterogeneous media and in vivo.5 The instrument is calibrated on each channel using euthanized mice with surgical implants of known fluorochrome concentration and is able to recover fluorescence depth, size, and concentration in three dimensions from tissue at any depth. The system exhibits strict linearity over a broad range of biologically relevant concentration and detection sensitivities in the order of femtomoles.22 Animal scan times are in the range of 2to5min ; tomographic reconstruction times on a standard desktop PC are about 1to3min .

For in vivo experiments, 2nmol NGR-Cy 5.5 were injected into the tail vein of each mouse ( n=5 for MCF-7, n=6 for HT-1080 xenografts). FRI and FMT were performed 1, 2, 5, and 24h (FRI additional 5, 10, and 30min ) post injection. Blocking studies were accomplished by injecting 10mgkg NGR-peptide in 100μL PBS (200nmol) 10min before injection of the labeled peptide ( n=6 for HT-1080 xenografts). For biodistribution studies, FRI images of isolated organs (tumor, muscle, heart, spleen, kidney, liver, and lung) were carried out 24h after injection of the tracer.

2.K.

Statistical Analysis

Data are presented as mean±SEM (standard error of the mean). Statistical analysis of in vivo tumor fluorescence was conducted using a two-tailed, unpaired student t-test. A p-value 0.05 was considered to be significant.

3.

Results

3.1.

Synthesis and Characterization of NGR-Cy 5.5

The NHS ester of the near infrared fluorophore Cy 5.5 is reacted with an equimolar amount of the cyclo-[Cys-Asn-Gly-Arg-Cys]-Gly-Lys (NGR) peptide and purified by reversed phase HPLC (purity 95% ). The retention time was 28.5min . Yields of the NGR-Cy 5.5 conjugate were typically 75 to 90%, as calculated with ε675=250,000(molL)1cm1 from the absorption spectra measured with the PBS solution. The excitation and emission spectrum of the targeted probe remained unchanged as compared to the nonmodified fluorophore showing excitation and emission peaks at 675nm or 694nm , respectively.

3.2.

CD13 Protein Expression in HT-1080 and MCF-7 Cells

To determine the cellular origin of the CD13/APN receptor and its activity in HT-1080 and MCF-7 cells, cell lines were analyzed for CD13 protein expression by FACS, Western blot, and immunohistochemistry as well as by measurement of the specific aminopeptidase activity. For flow cytometry, cells were incubated with PE-labeled WM15 antibody directed against the human APN, and binding was analyzed (Fig. 1 ). Approximately 87% of the HT-1080 cells expressed the APN-receptor. In contrast, the receptor was hardly detected on MCF-7 adenocarcinoma cells [0.24%; see also Fig. 1b]. CD13 Western blotting of HT-1080 and MCF-7 cells and tumor tissues with the H-300 antibody, recommended for detection of CD13 of mouse, rat, and human origin, confirmed the apparent Mr of APN at 150,000. THP-1 cell lysate was used as a positive control. The APN enzyme was expressed only on HT-1080 cells and tumor tissue, while MCF-7 cells and tumors are negative for CD13 expression (not shown). Proper catalytic function of the CD13 protein was verified using a specific neutral aminopeptidase activity assay [Ref. 23; Fig. 1c]. CD13 activity in HT-1080 and MCF-7 cells is correlated with the expression measured by flow cytometry and Western blot analysis. The enzyme activity in HT-1080 cells could be specifically blocked by the WM15 antibody24 and the NGR-peptide.

Fig. 1

Expression of CD13/APN in vitro. Human HT-1080 [(a), black curve] and MCF-7 [(a), red curve, which is enlarged in (b)] carcinoma cells were analyzed by flow cytometry using monoclonal antibody WM15-PE (CD13/APN specific) or isotype control (mouse-IgG1-PE). The results are presented in histograms (i.e., distribution of cells according to their fluorescence intensity). Numbers indicate the percentage of cells that were CD13/APN positive. (c) APN activity assay using L-alanine-4-methy-coumarinylamide-(MCA)-trifluoroacetate as substrate. The release of fluorescent product 7-amido-4-methylcoumarin was monitored on positive HT-1080 fibrosarcoma and negative MCF-7 adenocarcinoma cells. To assay for inhibition of enzymatic activity, binding of CD13-specific antibody WM15 and the NGR-peptide was measured on HT-1080 cells. (Color online only.)

011007_1_020801jbo1.jpg

3.3.

In Vitro NGR Binding Assays

Based on the CD13 protein expression data, the binding properties of cyclo-[Cys-Asn-Gly-Arg-Cys]-Gly-Lys-Cy 5.5 (NGR-Cy 5.5) to the cell lines were evaluated by fluorescence microscopy (Fig. 2 ). Negligible signals were detected when cells were incubated with the nonmodified Cy 5.5 dye alone (up to 30μM ; data not shown), and also for MCF-7 cells following incubation with up to 12.5μM of the labeled tracer [Fig. 2c]. The labeled peptide bound distinctly to HT-1080 cells [Fig. 2a]. The fluorescence was distributed over the cell surface and membrane-associated. Blocking of the signal was possible with a tenfold excess of unlabeled NGR-peptide [ 125μM ; Fig. 2b].

Fig. 2

Binding affinity of NGR-Cy 5.5 to tumor cell lines with different CD13/APN expression level. HT-1080 fibrosarcoma cells [(a) and (b)] and MCF-7 adenocarcinoma cells (c) were incubated with 12.5μM NGR-Cy 5.5 to assess the target affinity of the probe. In accordance with FACS analysis and Western blot data, HT-1080 cells producing high CD13/APN levels revealed strong cellular fluorescence [(a): fluorescence microscopy; left: phase contrast], which could be selectively blocked by coincubation with the unlabeled NGR-peptide (b). MCF-7 cells (c), which lack CD13 expression, did not bind the targeted fluorochrome (objective magnification: 40× ).

011007_1_020801jbo2.jpg

3.4.

Immunohistochemistry

Immunohistochemistry on tumor tissue slides with CD13 antibody H-300 [Figs. 3c and 3d ] exhibited membranous CD13/APN antigen expression on HT-1080 cells and only background staining in MCF-7 xenografts. Specific in vivo binding of NGR-Cy 5.5 in HT-1080 tumor tissue could be visualized in Fig. 3e, exhibiting an intense, membranous fluorescence in tumor tissue 24h after injection of the optical tracer, whereas MCF-7 tissue showed only background staining [Fig. 3f].

Fig. 3

In vivo expression of CD13/APN in tumor xenografts. Immunohistochemical staining of CD13/APN and fluorescence microscopy of tumor slides taken 24h after injection of NGR-Cy 5.5. Paraffin sections from HT-1080 and MCF-7 tumor xenografts were stained with: H&E [(a) and (b)] and CD13 [(c) and (d); H-300; 5μgml ]. Nuclei were counterstained with hematoxylin. HT-1080 tumors exhibited a strong, membranous CD13 expression (c), whereas MCF-7 tumors showed only background CD13 staining (d). Red arrows exemplify CD13 staining. HT-1080 tissue sections (e) exhibited a strong cellular fluorescence indicating a high binding of the optical tracer, while MCF-7 tissue showed only background staining. Objective magnification 20 and 40× .

011007_1_020801jbo3.jpg

3.5.

In Vivo Target Assessment and Optical Imaging

Figure 4a shows typical NIR fluorescence reflectance images of mice with HT-1080 (n=6) and MCF-7 (n=5) tumors, respectively, 24h after injection with NGR-Cy 5.5. After clearing of the unbound circulating fluorochromes, the HT-1080 tumor xenografts could be clearly delineated from the surrounding background tissue up to 24h with maximal fluorescence intensities of 2756.2±209.4AU , visible 3h post injection [Fig. 4b]. Compared to the targeted probe, the blocking experiment revealed lower fluorescence intensities (e.g., 1h post injection: 2339.3±269.9AU versus 1999.5±213.5AU ; n=6 ; p<0.05 ). Target-to-background ratios (TBR) after tracer injection ranged around 1.61±0.52 ( 1.54±0.56 for the blocking experiment). MCF-7 xenografts showed a weaker fluorescence, indicating a lower expression of the CD13/APN receptor [ 1407.5±122.0AU 3h p.i.; Fig. 4b]. In HT-1080 tumor xenografts, the nonmodified Cy 5.5 revealed a fluorescence intensity of 1404.4±243.8AU (n=7) 24h post injection (data not shown).

Fig. 4

In vivo fluorescence reflectance imaging of HT-1080 and MCF-7 xenografts. HT-1080 fibrosarcoma (n=6) and MCF-7 adenocarcinoma (n=5) bearing mice were i.v. injected with the NGR-Cy 5.5 probe and imaged the following 24h using a multichannel fluorescence reflectance imager (FRI). (a) FRI image 24h past NGR-Cy 5.5 injection. Tumors were marked with an arrow. (b) The fluorochrome showed a strong retention over the time into the tumor (solid line), revealing a significantly higher target to background ratio as compared to APN-negative MCF-7 xenografts (dashed line). Tracer uptake could be blocked by preinjection of the unlabeled peptide (not shown).

011007_1_020801jbo4.jpg

FMT confirmed the FRI results, exhibiting a strong fluorochrome distribution in the target tissue throughout the tumor (Fig. 5 ). Quantitative data analysis revealed a fluorochrome concentration 5h after injection of 306.7±54.3nM NGR-Cy 5.5 (Fig. 5), while blocking with the free peptide resulted in an average fluorochrome concentration of 195.3±21.9nM NGR-Cy 5.5 (Fig. 5). TBR values for this time-point ranged around 7.53±4.04 and 3.33±2.13 for the blocking experiment. Tumor fluorescence reached maximum intensities after 5h and showed a slow clearance over the time [Fig. 5g]. Fluorochrome concentrations in nontarget tissue were significantly lower and ranged around 116.0±18.3nM .

Fig. 5

Fluorescence-mediated tomography of HT-1080 and MCF-7 tumor bearing nude mice. FMT experiments were performed to visualize and quantify the three-dimensional (3-D) fluorochrome distribution in deeper tissue sections. The fluorochrome concentration is displayed color encoded (scale: 1to256nM ). FMT was carried out 24h after injection of 2nmol NGR-Cy 5.5 probe in HT-1080 (a) and MCF-7 (e) tumor–bearing nude mice. In order to assess probe specificity, 200nmol of the unlabeled peptide were i.v. injected 10min before application of the NGR-Cy 5.5 probe (c). FMT revealed intense fluorochrome retention 60min after probe injection in HT-1080 tumors (b), which could be significantly reduced after pretreatment with the nonmodified peptide (d). Compared to the fluorescence measured in HT-1080 tumors, the fluorochrome accumulation in MCF-7 tumors (f) was substantially lower. Tumor fluorescence washout over the time was slower in HT-1080 than in MCF-7 tumor tissue (g), black bar=native HT-1080; gray bar=blocked HT-1080; white bar=MCF-7 .

011007_1_020801jbo5.jpg

3.6.

Biodistribution

The distribution of Cy 5.5-labeled NGR-peptide was measured in tumor, muscle, heart, lung, spleen, kidneys, and liver from nude mice bearing HT-1080 fibrosarcoma (n=6) and MCF-7 adenocarcinoma (n=5) tumors. Data were obtained by FRI measuring 24h post injection of the tracer. A representative example of NGR-Cy 5.5 biodistribution in HT-1080 tumor–bearing mice is shown in Fig. 6 . The highest concentration of optical tracer 24h after injection was determined in kidneys ( 792.6±272.6AU for HT-1080 and 725.3±52.6AU for MCF-7), followed by the accumulation in liver (398.0±86.9AU) , tumor (331.9±151.7AU) , and lung (306.5±38.6AU) . Preinjection of the unlabeled peptide resulted in decreased uptake in all dissected tissues (data not shown).

Fig. 6

Representative overlay of white light and color-encoded FRI images of dissected organs of HT-1080 tumor–bearing nude mice. Animals (n=6) were sacrificed 24h after intravenous injection of 2nmol NGR-Cy 5.5. Enhanced fluorochrome accumulation was detected in kidneys, liver, and lung tissue [(1): HT-1080 tumor, (2) heart, (3) spleen, (4) lung, (5) liver, (6) kidneys], indicating a high tracer washout of tumor tissue and a renal excretion. The fluorochrome concentration is displayed color encoded (scale: 1to256nM ).

011007_1_020801jbo6.jpg

4.

Discussion

Targets that are located on the endothelial surface are ideal candidates for in vivo molecular imaging approaches because pharmacological barriers are virtually nonexistent. CD13/APN is an ectoenzyme located in the outer cell membranes of various cells like epithelial cells, macrophages, and endothelial cells. Therapeutic targeting of the CD13/APN receptor has successfully been performed using, e.g., NGR-fusion peptides with related anticancer prodrugs exhibiting selective cytotoxity toward CD13/APN positive tumor cells or angiogenetic endothelial cells.14, 15, 18, 24, 25, 26 More recently, fluorochrome labeling of NGR-peptides has been shown to be feasible.17

For our study, we chose a cyclic NGR-peptide with a cystin group and a gly-lys carboxy terminus as a spacer function. The use of a cystin moiety for cyclization was chosen because of the reported enhanced tumor targeting efficiency of such peptides.15 CD13/APN activity of positive HT-1080 cells16 and negative MCF-7 cells were demonstrated by FACS, Western blotting, and an APN activity assay using Ala-MCA as substrate. Blocking of CD13-specific aminopeptidase activity with the NGR-peptide and the peptide-dye conjugate was in line with the activity-blocking antibody WM15,23, 24 indicating the specific binding of the labeled NGR-peptide to the target structure.

In in vitro binding studies evaluated by fluorescence microscopy, the peptide-dye conjugate showed distinct binding affinity to HT-1080 cells, while MCF-7 cells and incubation of cells with Cy 5.5 revealed little to no fluorescence. Initially, the fluorescence was membrane associated – a phenomenon also observable in fluorescence microscopic analysis of the tumor xenografts; prolonged incubation times resulted in endocytosis of the tracer and nuclear staining. The specificity of the probe could be confirmed by a competition assay with a tenfold excess of unlabeled NGR-peptide, which resulted in a reduction of cellular fluorescence. Thus, in vitro tracer binding results closely correlate with FACS and APN activity assays and suggest a high binding affinity of the peptide-dye conjugate to the target.

In vivo, HT-1080 xenografts were clearly visualized by FRI up to 24h after i.v. injection of NGR-Cy 5.5, showing a maximum TBR of 1.61±0.52 after 1h . Blocking with free peptide resulted in a decreased uptake of the optical tracer (TBR 1.54±0.56 ). MCF-7 tumors showed a TBR of 0.96±0.23 . To overcome FRI-related quantification problems, FMT was performed in a group of animals, allowing the three-dimensional (3-D) reconstruction and quantification of fluorochrome distribution in vivo. At maximum tumor tracer uptake after 5h , FMT data correlates nicely with in vitro and FRI data, reflecting the different APN cellular expression level for HT-1080 and MCF-7 tumors. Blocking with the unlabeled peptide led to a decreased uptake of optical tracer (e.g., for HT-1080 after 24h 112.8±25.7nM ). Besides target-specific fluorochrome accumulation, nonspecific accumulation of the tracer occurs due to perfusion and vascular permeability effects as well as endocytosis of the peptides over time. This effect was also observed here by imaging the nonmodified fluorophore and has been described previously for other non-target-specific NIR-fluorochromes.27 These nonspecific, contrasting mechanisms can also explain the relatively high remaining fluorescence signal after blocking with the free peptide. Necrotic areas within the tumor also lead to a heterogeneous tracer uptake and will influence the correlation between NGR-Cy 5.5 and APN expression. TBR measured by FMT for HT-1080 xenografts (e.g., 5h post injection 7.53±4.04 ) was significantly higher as compared to corresponding FRI data (1.61±0.52) . This may be on one hand attributed to a lower amount of bleed-through between the excitation and emission spectrum in the FMT. On the other hand, this demonstrates the capability of resolving and differentiating deeply located tissue fluorescence (e.g., deriving from the liver) from more superficially located tumor-associated fluorochrome accumulation. While FRI cannot differentiate between deeply located, strongly fluorescent organs (e.g., liver) and superficial fluorochrome tumoral accumulation, cross-sectional imaging with FMT can. Thus, scattered fluorescence from deep-seated structures can significantly spoil the TBR in FRI compared to FMT.

Direct imaging of excised organs 24h p.i. exhibited a high accumulation of the optical tracer not in the tumor but in the kidneys and liver of all tested mice. That has a lot to commend a rapid blood clearance and a renal excretion of the NGR-Cy 5.5 peptide, a feature that is preferable for further clinical applications. Nevertheless, CD13 expression by the reabsorptive epithelium of the kidney was also reported,28, 29 but NGR drug conjugates like NGR-TNF failed to bind to CD13 expressed in normal kidney,24 indicating a selective tumor-homing property to a specific CD13 isoform. Further studies are necessary to clarify the binding properties of NGR-conjugated optical imaging probes to different APN isoforms. However, the immunohistochemical data suggest that the NGR-Cy 5.5 probe binds to both the membrane-associated protein of the tumor cells as well as endothelial targets.

For in vivo applications, specifically for tomographic imaging in deep tissue sections, a maximum TBR is desirable. The affinity of the currently available NGR-NIR-dye conjugate is moderate. Different strategies have been proposed for other peptidic tracers, especially the RGD-peptide directed against the integrin αvβ3 -receptor, to increase the target affinity of the imaging agent and decrease a high accumulation in other organs such as liver and kidneys. Possible methods are glycosylation of the tracer, peptide carbohydrate conjugates, and peptidomimetics based on the NGR-structure as well as heterodimeric, homodimeric, and homotetrameric ligand systems as tested for the RGD-peptide.30, 31, 32 These modifications have already been carried out with radio-labeled30, 33 and Cy 5.5-labeled RGD probes.34 The results could be a useful source for the development of NGR optical imaging probes.

A clinical translation of this optical imaging technology is conceivable for certain organs such as breast or head and neck tumors. The feasibility of contrast-enhanced diffuse optical mammography in humans has been shown by Ntziachristos 35 Combining this imaging technique with target-specific fluorochromes as described in this work should greatly enhance the diagnostic accuracy for detecting breast cancer at an early tumor stage.36 Besides tomographic imaging, planar imaging techniques such as FRI can easily be translated for clinical use. A tumor binding fluorochrome could for instance be applied to preoperatively “tag” the tissue to be resected. The NIR signal deriving from the tumor could then easily be detected with planar optical imaging techniques in the operating room, and thus tumor margins could be visualized for the surgeon.37, 38

In conclusion, our study shows that CD13/APN expression can be imaged in vivo using fluorescence reflectance imaging and fluorescence-mediated tomography approaches. Since FMT allowed 3-D reconstruction of fluorochrome distribution, the tumor-to-background ratio could be significantly enhanced and furthermore quantified noninvasively. This imaging technique is therefore a versatile tool not only for experimental small animal molecular imaging but potentially also for noninvasive target detection, e.g., in clinical breast cancer imaging.36

Acknowledgments

We thank Wiebke Gottschlich, Ingrid Otto-Valk, Ursula Römer, and Janine Ring for technical assistance. For critical reading and fruitful discussion, we gratefully thank Prof. Dr. Werner Schlegel (University of Münster, Germany). This work was supported in part by the Deutsche Forschungsgemeinschaft (DFG) (BR 1653/2-1, SFB 656 Project A4) and the European Community (IP 6th framework; LSHG-CT-2003-503259).

References

1. 

C. Bremer, V. Ntziachristos, and R. Weissleder, “Optical-based molecular imaging: contrast agents and potential medical applications,” Eur. Radiol., 13 (2), 231 –243 (2003). 0938-7994 Google Scholar

2. 

J. Grimm, D. G. Kirsch, S. D. Windsor, C. F. Kim, P. M. Santiago, V. Ntziachristos, T. Jacks, and R. Weissleder, “Use of gene expression profiling to direct in vivo molecular imaging of lung cancer,” Proc. Natl. Acad. Sci. U.S.A., 102 (40), 14404 –14409 (2005). https://doi.org/10.1073/pnas.0503920102 0027-8424 Google Scholar

3. 

U. Mahmood and R. Weissleder, “Near-infrared optical imaging of proteases in cancer,” Mol. Cancer Ther., 2 (5), 489 –496 (2003). Google Scholar

4. 

V. Ntziachristos, C. Bremer, and R. Weissleder, “Fluorescence imaging with near-infrared light: new technological advances that enable in vivo molecular imaging,” Eur. Radiol., 13 (1), 195 –208 (2003). 0938-7994 Google Scholar

5. 

E. E. Graves, D. Yessayan, G. Turner, R. Weissleder, and V. Ntziachristos, “Validation of in vivo fluorochrome concentrations measured using fluorescence molecular tomography,” J. Biomed. Opt., 10 (4), 44019 (2005). 1083-3668 Google Scholar

6. 

X. Montet, V. Ntziachristos, J. Grimm, and R. Weissleder, “Tomographic fluorescence mapping of tumor targets,” Cancer Res., 65 (14), 6330 –6336 (2005). https://doi.org/10.1158/0008-5472.CAN-05-0382 0008-5472 Google Scholar

7. 

V. Ntziachristos, C. Bremer, E. E. Graves, J. Ripoll, and R. Weissleder, “In vivo tomographic imaging of near-infrared fluorescent probes,” Mol. Imaging, 1 (2), 82 –88 (2002). https://doi.org/10.1162/153535002320162732 1535-3508 Google Scholar

8. 

V. Ntziachristos, C. Bremer, C. Tung, and R. Weissleder, “Imaging cathepsin B up-regulation in HT-1080 tumor models using fluorescence-mediated molecular tomography (FMT),” Acad. Radiol., 9 S323 –325 (2002). 1076-6332 Google Scholar

9. 

V. Ntziachristos, C. H. Tung, C. Bremer, and R. Weissleder, “Fluorescence molecular tomography resolves protease activity in vivo,” Nat. Med., 8 (7), 757 –760 (2002). 1078-8956 Google Scholar

10. 

D. Riemann, A. Kehlen, and J. Langner, “CD13—not just a marker in leukemia typing,” Immunol. Today, 20 (2), 83 –88 (1999). https://doi.org/10.1016/S0167-5699(98)01398-X 0167-4919 Google Scholar

11. 

S. V. Bhagwat, J. Lahdenranta, R. Giordano, W. Arap, R. Pasqualini, and L. H. Shapiro, “CD13/APN is activated by angiogenic signals and is essential for capillary tube formation,” Blood, 97 (3), 652 –659 (2001). https://doi.org/10.1182/blood.V97.3.652 0006-4971 Google Scholar

12. 

R. Pasqualini, E. Koivunen, R. Kain, J. Lahdenranta, M. Sakamoto, A. Stryhn, R. A. Ashmun, L. H. Shapiro, W. Arap, and E. Ruoslahti, “Aminopeptidase N is a receptor for tumor-homing peptides and a target for inhibiting angiogenesis,” Cancer Res., 60 (3), 722 –727 (2000). 0008-5472 Google Scholar

13. 

Y. Sato, “Role of aminopeptidase in angiogenesis,” Biol. Pharm. Bull., 27 (6), 772 –776 (2004). https://doi.org/10.1248/bpb.27.772 0918-6158 Google Scholar

14. 

W. Arap, R. Pasqualini, and E. Ruoslahti, “Cancer treatment by targeted drug delivery to tumor vasculature in a mouse model,” Science, 279 (5349), 377 –380 (1998). https://doi.org/10.1126/science.279.5349.377 0036-8075 Google Scholar

15. 

G. Colombo, F. Curnis, G. M. De Mori, A. Gasparri, C. Longoni, A. Sacchi, R. Longhi, and A. Corti, “Structure-activity relationships of linear and cyclic peptides containing the NGR tumor-homing motif,” J. Biol. Chem., 277 (49), 47891 –47897 (2002). https://doi.org/10.1074/jbc.M207500200 0021-9258 Google Scholar

16. 

Y. van Hensbergen, H. J. Broxterman, Y. W. Elderkamp, J. Lankelma, J. C. Beers, M. Heijn, E. Boven, K. Hoekman, and H. M. Pinedo, “A doxorubicin-CNGRC-peptide conjugate with prodrug properties,” Biochem. Pharmacol., 63 (5), 897 –908 (2002). 0006-2952 Google Scholar

17. 

Z. Zhang, H. Harada, K. Tanabe, H. Hatta, M. Hiraoka, and S. I. Nishimoto, “Aminopeptidase N/CD13 targeting fluorescent probes: synthesis and application to tumor cell imaging,” Peptides, 26 (11), 2182 –2187 (2005). 0196-9781 Google Scholar

18. 

Z. Zhang, H. Hatta, K. Tanabe, and S. Nishimoto, “A new class of 5-fluoro-2-deoxyuridine prodrugs conjugated with a tumor-homing cyclic peptide CNGRC by ester linkers: synthesis, reactivity, and tumor-cell-selective cytotoxicity,” Pharm. Res., 22 (3), 381 –389 (2005). https://doi.org/10.1007/s11095-004-1875-x 0724-8741 Google Scholar

19. 

F. Pastorino, C. Brignole, D. Marimpietri, M. Cilli, C. Gambini, D. Ribatti, R. Longhi, T. M. Allen, A. Corti, and M. Ponzoni, “Vascular damage and anti-angiogenic effects of tumor vessel-targeted liposomal chemotherapy,” Cancer Res., 63 (21), 7400 –7409 (2003). 0008-5472 Google Scholar

20. 

A. Corti and M. Ponzoni, “Tumor vascular targeting with tumor necrosis factor alpha and chemotherapeutic drugs,” Ann. N.Y. Acad. Sci., 1028 104 –112 (2004). 0077-8923 Google Scholar

21. 

Y. van Hensbergen, H. J. Broxterman, R. Hanemaaijer, A. S. Jorna, N. A. van Lent, H. M. Verheul, H. M. Pinedo, and K. Hoekman, “Soluble aminopeptidase N/CD13 in malignant and nonmalignant effusions and intratumoral fluid,” Clin. Cancer Res., 8 (12), 3747 –3754 (2002). 1078-0432 Google Scholar

22. 

V. Ntziachristos, J. Ripoll, L. V. Wang, and R. Weissleder, “Looking and listening to light: the evolution of whole-body photonic imaging,” Nat. Biotechnol., 23 (3), 313 –320 (2005). https://doi.org/10.1038/nbt1074 1087-0156 Google Scholar

23. 

Y. van Hensbergen, H. J. Broxterman, S. Rana, P. J. van Diest, M. C. Duyndam, K. Hoekman, H. M. Pinedo, and E. Boven, “Reduced growth, increased vascular area, and reduced response to cisplatin in CD13-overexpressing human ovarian cancer xenografts,” Clin. Cancer Res., 10 (3), 1180 –1191 (2004). 1078-0432 Google Scholar

24. 

F. Curnis, G. Arrigoni, A. Sacchi, L. Fischetti, W. Arap, R. Pasqualini, and A. Corti, “Differential binding of drugs containing the NGR motif to CD13 isoforms in tumor vessels, epithelia, and myeloid cells,” Cancer Res., 62 (3), 867 –874 (2002). 0008-5472 Google Scholar

25. 

Y. Yokoyama and S. Ramakrishnan, “Addition of an aminopeptidase N-binding sequence to human endostatin improves inhibition of ovarian carcinoma growth,” Cancer, 104 (2), 321 –331 (2005). 0008-543X Google Scholar

26. 

A. Dirksen, S. Langereis, B. F. de Waal, M. H. van Genderen, E. W. Meijer, Q. G. de Lussanet, and T. M. Hackeng, “Design and synthesis of a bimodal target-specific contrast agent for angiogenesis,” Org. Lett., 6 (26), 4857 –4860 (2004). https://doi.org/10.1021/ol048084u 1523-7060 Google Scholar

27. 

K. Licha, B. Riefke, V. Ntziachristos, A. Becker, B. Chance, and W. Semmler, “Hydrophilic cyanine dyes as contrast agents for near-infrared tumor imaging: synthesis, photophysical properties, and spectroscopic in vivo characterization,” Photochem. Photobiol., 72 (3), 392 –398 (2000). https://doi.org/10.1562/0031-8655(2000)072<0392:HCDACA>2.0.CO;2 0031-8655 Google Scholar

28. 

D. Bordessoule, M. Jones, K. C. Gatter, and D. Y. Mason, “Immunohistological patterns of myeloid antigens: tissue distribution of CD13, CD14, CD16, CD31, CD36, CD65, CD66, and CD67,” Br. J. Haematol., 83 (3), 370 –383 (1993). 0007-1048 Google Scholar

29. 

J. Dixon, L. Kaklamanis, H. Turley, I. D. Hickson, R. D. Leek, A. L. Harris, and K. C. Gatter, “Expression of aminopeptidase-n (CD 13) in normal tissues and malignant neoplasms of epithelial and lymphoid origin,” J. Clin. Pathol., 47 (1), 43 –47 (1994). https://doi.org/10.1136/jcp.47.1.43 0021-9746 Google Scholar

30. 

R. Haubner and H. J. Wester, “Radiolabeled tracers for imaging of tumor angiogenesis and evaluation of anti-angiogenic therapies,” Curr. Pharm. Des., 10 (13), 1439 –1455 (2004). Google Scholar

31. 

R. Haubner, H. J. Wester, F. Burkhart, R. Senekowitsch-Schmidtke, W. Weber, S. L. Goodman, H. Kessler, and M. Schwaiger, “Glycosylated RGD-containing peptides: tracer for tumor targeting and angiogenesis imaging with improved biokinetics,” J. Nucl. Med., 42 (2), 326 –336 (2001). 0161-5505 Google Scholar

32. 

R. Haubner, H. J. Wester, U. Reuning, R. Senekowitsch-Schmidtke, B. Diefenbach, H. Kessler, G. Stocklin, and M. Schwaiger, “Radiolabeled alpha(v)beta3 integrin antagonists: a new class of tracers for tumor targeting,” J. Nucl. Med., 40 (6), 1061 –1071 (1999). 0161-5505 Google Scholar

33. 

X. Chen, M. Tohme, R. Park, Y. Hou, J. R. Bading, and P. S. Conti, “Micro-PET imaging of alphavbeta3-integrin expression with 18F-labeled dimeric RGD peptide,” Mol. Imaging, 3 (2), 96 –104 (2004). https://doi.org/10.1162/1535350041464892 1535-3508 Google Scholar

34. 

Z. Cheng, Y. Wu, Z. Xiong, S. S. Gambhir, and X. Chen, “Near-infrared fluorescent RGD peptides for optical imaging of integrin alphavbeta3 expression in living mice,” Bioconjugate Chem., 16 (6), 1433 –1441 (2005). https://doi.org/10.1021/bc0501698 1043-1802 Google Scholar

35. 

V. Ntziachristos, A. G. Yodh, M. Schnall, and B. Chance, “Concurrent MRI and diffuse optical tomography of breast after indocyanine green enhancement,” Proc. Natl. Acad. Sci. U.S.A., 97 (6), 2767 –2772 (2000). https://doi.org/10.1073/pnas.040570597 0027-8424 Google Scholar

36. 

J. M. Martinez, I. Prieto, M. J. Ramirez, C. Cueva, F. Alba, and M. Ramirez, “Aminopeptidase activities in breast cancer tissue,” Clin. Chem., 45 (10), 1797 –1802 (1999). 0009-9147 Google Scholar

37. 

M. F. Kircher, U. Mahmood, R. S. King, R. Weissleder, and L. Josephson, “A multimodal nanoparticle for preoperative magnetic resonance imaging and intraoperative optical brain tumor delineation,” Cancer Res., 63 (23), 8122 –8125 (2003). 0008-5472 Google Scholar

38. 

A. M. De Grand and J. V. Frangioni, “An operational near-infrared fluorescence imaging system prototype for large animal surgery,” Technol. Cancer Res. Treat., 2 (6), 553 –562 (2003). 1533-0346 Google Scholar
©(2008) Society of Photo-Optical Instrumentation Engineers (SPIE)
Angelika von Wallbrunn, Jens Waldeck, Carsten Hoeltke, Michael Zühlsdorf, Rolf Mesters, Walter L. Heindel M.D., Michael Schäfers, and Christoph Bremer "In vivo optical imaging of CD13/APN-expression in tumor xenografts," Journal of Biomedical Optics 13(1), 011007 (1 January 2008). https://doi.org/10.1117/1.2839046
Published: 1 January 2008
Lens.org Logo
CITATIONS
Cited by 60 scholarly publications and 1 patent.
Advertisement
Advertisement
RIGHTS & PERMISSIONS
Get copyright permission  Get copyright permission on Copyright Marketplace
KEYWORDS
Tumors

Luminescence

In vivo imaging

Optical imaging

Tissues

Tomography

Imaging systems

Back to Top