Effective diagnosis of inflammation and cancer by molecular imaging is challenging because of interference from nonselective accumulation of the contrast agents in normal tissues. Here, we report a series of novel fluorescence imaging agents that efficiently target cyclooxygenase-2 (COX-2), which is normally absent from cells, but is found at high levels in inflammatory lesions and in many premalignant and malignant tumors. After either i.p. or i.v. injection, these reagents become highly enriched in inflamed or tumor tissue compared with normal tissue and this accumulation provides sufficient signal for in vivo fluorescence imaging. Further, we show that only the intact parent compound is found in the region of interest. COX-2–specific delivery was unambiguously confirmed using animals bearing targeted deletions of COX-2 and by blocking the COX-2 active site with high-affinity inhibitors in both in vitro and in vivo models. Because of their high specificity, contrast, and detectability, these fluorocoxibs are ideal candidates for detection of inflammatory lesions or early-stage COX-2–expressing human cancers, such as those in the esophagus, oropharynx, and colon. Cancer Res; 70(9); 3618–27. ©2010 AACR.

This article is featured in Breaking Advances, p. 3417

Molecular imaging presents exciting opportunities for the selective detection of specific cell populations, such as those bearing markers of disease (1, 2). Cyclooxygenase-2 (COX-2) is an attractive target for molecular imaging because it is expressed in only a few normal tissues and is greatly upregulated in inflamed tissues as well as many premalignant and malignant tumors (3, 4). COX-2 is an important contributor to the etiology of inflammation and cancer as illustrated by the efficacy of COX-2–selective inhibitors as anti-inflammatory agents, cancer preventive agents, and adjuvant cancer therapeutic agents (5). The importance of COX-2 in tumor progression has been thoroughly documented in the esophagus and colon where COX-2 is detected in premalignant lesions and its levels seem to increase during tumor progression (68). The importance of COX-2 in survival and response to therapy has been elegantly shown by Edelman and colleagues (9) who reported that non–small cell lung cancer patients expressing high levels of COX-2 in their tumors have reduced survival compared with patients expressing low levels of COX-2. Patients with high tumor expression of COX-2 benefit from the combination of carboplatin and gemcitabine plus the COX-2 inhibitor, celecoxib, whereas patients with low expression exhibit a poorer response to carboplatin/gemcitabine/celecoxib than to carboplatin/gemcitabine alone (9).

Positron emission tomography or single-photon emission computed tomography imaging agents (18F-, 11C-, or 123I-labeled COX-2 inhibitors) have been described for nuclear imaging (1017). These have all been based on the diarylheterocycle structural class analogous to celecoxib and rofecoxib. Although selective uptake into macrophages or tumor cells expressing COX-2 has been shown in vitro for some compounds, such selectivity has not been rigorously shown in vivo and significant nonspecific binding has been observed (18). Thus, despite recognition of the potential of COX-2–targeted imaging agents, in vivo proof-of-concept for this strategy is lacking.

Fluorescent COX-2 inhibitors are attractive candidates as targeted imaging agents. Such compounds have the advantage that each molecule bears the fluorescent tag and the compounds are nonradioactive and stable. Thus, they can be used conveniently for cellular imaging, animal imaging, and clinical imaging of tissues in which topical or endoluminal illumination is possible (e.g., esophagus, colon, and upper airway through endoscopy, colonoscopy, and bronchoscopy, respectively). Prior work from our laboratory showed that fluorescent COX-2 inhibitors can be useful biochemical probes of protein binding but these earlier compounds were neither potent inhibitors of COX-2 nor did they possess appropriate fluorescence properties to be useful for cellular or in vivo imaging (19). Thus, we initiated a program to design and synthesize a series of fluorescent COX-2 inhibitors that could be used for these applications. The design strategy for candidate development was based on our prior discovery that amide derivatives of the nonselective COX inhibitor, indomethacin, are selective COX-2 inhibitors. Many compounds were synthesized and screened for COX-2 inhibition in vitro and in intact cells. Then, the most promising compounds were evaluated as imaging agents in intact cells and in animal models of inflammation and cancer. We describe herein the optimized candidates, their selective uptake by COX-2–expressing cells and tumors, and genetic and pharmacologic validation that their in vivo target is COX-2.

Synthesis and characterization of all compounds is described in Supplementary Data.

Inhibition assay using purified COX-1 and COX-2

Cyclooxygenase activity of ovine COX-1 or human COX-2 was assayed by a method that quantifies the conversion of [1-14C]arachidonic acid to [1-14C]prostaglandin products. Reaction mixtures of 200 μL consisted of hematin-reconstituted protein in 100 mmol/L Tris-HCl (pH 8.0), 500 μmol/L phenol, and [1-14C]arachidonic acid (50 μmol/L, approximately 55–57 mCi/mmol, Perkin-Elmer). For the time-dependent inhibition assay, hematin-reconstituted COX-1 (44 nmol/L) or COX-2 (66 nmol/L) was preincubated at 25°C for 17 minutes and 37°C for 3 minutes with varying inhibitor concentrations in DMSO followed by the addition of [1-14C]arachidonic acid (50 μmol/L) for 30 seconds at 37°C. Reactions were terminated by solvent extraction in Et2O/CH3OH/1 mol/L citrate (pH 4.0; 30:4:1). The phases were separated by centrifugation at 2,000 g for 2 minutes and the organic phase was spotted on a TLC plate (EMD Kieselgel 60, VWR). The plate was developed in EtOAc/CH2Cl2/glacial AcOH (75:25:1) at 4°C. Radiolabeled products were quantified with a radioactivity scanner (Bioscan, Inc.). The percentage of total products observed at different inhibitor concentrations was divided by the percentage of products observed for protein samples preincubated for the same time with DMSO.

Cell culture and in vitro intact cell metabolism assay

HCT116, ATCC CCL-247 human colorectal carcinoma cells, passage 8 to 18, Mycoplasma negative by a PCR detection method (Sigma VenorGem) were grown in DMEM (Invitrogen/Life Technologies) + 10% fetal bovine serum (FBS; Atlas) to 70% confluence. RAW264.7, ATCC TIB-71 murine macrophage-like cells, passage number 8 to 15, Mycoplasma negative by a PCR detection method were grown in DMEM + 10% heat-inactivated FBS to 40% confluence (six-well plates, Sarstedt) and activated for 7 hours in 2 mL serum-free DMEM with 200 ng/mL lipopolysaccharide (LPS; Calbiochem) and 10 μ/mL IFN γ (Calbiochem). Human 1483 head and neck squamous cell carcinoma (HNSCC) cells (20), derived, characterized, and provided by Dr. Peter Sacks (New York University School of Dentistry, New York, NY), were grown at passage 8 to 18, Mycoplasma negative by a PCR detection method, in DMEM/F12 + 10% FBS + Antibiotic/Antimycotic in six-well plates to 60% confluence. Serum-free medium (2 mL) was added and the cells were treated with inhibitor dissolved in DMSO (0–5 μmol/L, final concentration) for 30 minutes at 37°C followed by the addition of [1-14C]-arachidonic acid (10 μmol/L, ∼55 mCi/mmol) for 20 minutes at 37°C. Reactions were terminated by solvent extraction in Et2O/CH3OH/1 mol/L citrate (pH 4.0; 30:4:1) and the organic phase was spotted on a 20 × 20 cm TLC plate (EMD Kieselgel 60, VWR). The plate was developed in EtOAc/CH2Cl2/glacial AcOH (75:25:1) and radiolabeled products were quantified with a radioactivity scanner (Bioscan, Inc.). The percentage of total products observed at different inhibitor concentrations was divided by the percentage of products observed for cells preincubated with DMSO.

Fluorescence microscopy of RAW264.7 cells or 1483 HNSCC cells

RAW264.7 cells were plated on 35-mm MatTek dishes (MatTek Corp.) such that the cells were 40% confluent and human 1483 HNSCC cells were 60% confluent on the day of the experiment. The RAW264.7 cells were activated for 6 hours in serum-free DMEM with 200 ng/mL LPS and 10 μ/mL IFN γ. Both cell lines were incubated in 2.0 mL HBSS/Tyrode's buffer with 200 nmol/L compound 1, 2, or 3 for 30 minutes at 37°C. To block the COX-2 active site, the cells were preincubated with 10 μmol/L indomethacin or 5 μmol/L celecoxib for 20 minutes before the addition of compound 1 or 2. The cells were then washed briefly thrice and incubated in HBSS/Tyrode's buffer for 30 minutes at 37°C. Following the required washout period, the cells were imaged in 2.0 mL fresh HBSS/Tyrode's buffer on a Zeiss Axiovert 25 Microscope with the propidium iodide filter (0.5–1.0 second exposure, gain of 2). All treatments were performed in duplicate dishes in at least three separate experiments.

Confocal microscopy of 1483 cells treated with compound 2/mitotrackerGR

1483 HNSCC were plated in MatTek dishes (MatTek Corp.) and grown to 60% to 70% confluence for 48 hours. DMSO or compound 2 (100 nmol/L) was added to each dish containing 2.0 mL HBSS/Tyrode's buffer for 30 minutes at 37°C. After four quick HBSS washes, cells were incubated for 30 minutes in 2.0 mL HBSS/Tyrode's buffer and imaged with a Zeiss LSM510 confocal microscope using a 63 × 1.4 NA plan-Apochromat objective lens. To visualize cellular mitochondria, 100 nmol/L Mitotracker GR was added for 15 minutes at 37°C followed by three quick washes before imaging. Four hundred eighty-eight nanometer excitation were used to image Mitotracker GR through a 500- to 530-nm bandpass filter and compound 2 was imaged using 532 nm excitation and collection through a 565- to 615-nm bandpass filter. The pinhole was set to 1 Airy unit and images were collected throughout the focus of the cells. To assure a full sampling of the perinuclear region, analysis was performed on the optical sections through the middle of the nucleus.

In vivo imaging of COX-2 in inflammation

Carrageenan (50 μL 1% in sterile saline) was injected in the rear left footpad of female C57BL/6 mice, followed by compound 1 or 2 (1 mg/kg, i.p.) at 24 hours postcarrageenan. Animals were imaged 3 hours later in a Xenogen IVIS 200 (DsRed filter, 1.5 cm depth, 1 s). For comparison, animals also were dosed with compound 3, which does not inhibit COX-2. To test further the molecular target for compound 2, parallel experiments were performed using COX-2 (−/−) mice. Experiments also were performed in which compound 1 was administered to the same animals by repetitive i.p. injection on days 1, 3, 5, and 7 to monitor the time course of compound uptake following carrageenan induction of inflammation.

Establishment of xenografts in nude mice

Female nude mice, NU-Fox1nu, were purchased at 6 to 7 weeks of age from Charles River Laboratories. Human 1483 HNSCC cells and HCT116 colorectal carcinoma cells were trypsinized and resuspended in cold PBS containing 30% Matrigel such that 1 × 106 cells in 100 μL were injected s.c. on the left flank. The HCT116 or 1483 xenografts required only 2 to 3 weeks of growth.

In vivo imaging of nude mice with xenografts

Female nude mice bearing medium-sized HCT116 or 1483 xenograft tumors on the left flank were dosed by i.p. injection with 2 mg/kg compound 2 or by retro-orbital injection with 1 mg/kg compound 2. The animals were lightly anesthetized with 2% isoflurane for fluorescence imaging in the Xenogen IVIS 200 with the DSRed filter at 1.5-cm depth and 1-second exposure (f2). For the COX-2 active site–blocking experiments, nude mice bearing 1483 xenografts were predosed by i.p. injection with 2 mg/kg indomethacin at 24 hours and 1 hour before dosing with compound 2 (2 mg/kg, i.p.).

Pharmacokinetics of candidate compounds

Female nude mice with medium-sized 1483 HNSCC xenograft tumors on the left flank were injected i.p. with 2 mg/kg compound 2. At 0, 0.5, 3, 12, and 24 hours, the mice (n = 4 for each time point, duplicate experiments) were anesthetized with isoflurane. Blood samples were taken by cardiac puncture into a heparinized syringe into a 1.5-mL heparinized tube on ice, followed by removal of the liver, kidney, contralateral leg muscle, and xenograft tumor. All organs/tissues were rinsed briefly in ice-cold PBS, blotted dry, weighed, and snap frozen in liquid nitrogen. The blood samples were centrifuged at 4°C at 6,000 rpm for 5 minutes and the plasma was transferred to clean tubes and frozen at −80°C. Compound 2 was extracted by homogenizing the tissue in 100 mmol/L Tris (pH 7.0) buffer and mixing an aliquot of the homogenate with 1.2× volume of acetonitrile. The acetonitrile was removed and the samples were dried, reconstituted, and analyzed through reversed-phase high-performance liquid chromatography (HPLC)-UV using a Phenomenex 10 × 0.2 cm C18 or a Phenomenex 7.5 × 0.2 cm Synergi Hydro-RP column held at 40°C. The samples were quantified against a standard curve prepared by adding compound 2 to tissue homogenates of undosed animals followed by the workup described. Cochromatography was performed with multiple columns and elution conditions as described in the Supplemental Data.

In vivo imaging of Min mice

C57BL/6 APC-Min mice maintained on a high-fat (11%) diet for 18 weeks developed 20 to 30 intestinal polyps per mouse. Before imaging, Min mice were anesthetized (2% inhaled isoflurane) for retro-orbital injection of compound 2 at 1 mg/kg. At 2 hours postinjection, the mice were euthanized and the intestines were resected, washed with PBS, and fixed in 10% formalin before ex vivo imaging by fluorescence dissecting microscopy (Zeiss M2Bio; n = 5).

COX-2 is a potentially ideal target for molecular imaging because its active site (and the active site of COX-1) is buried deep inside each subunit of the homodimeric protein (2123). Access to the active site is controlled by a constriction that separates it from a large opening in the membrane-binding domain that we have termed the lobby (Supplementary Fig. S1). All substrates or inhibitors bind in the lobby and then diffuse through the constriction into the active site (24). The constriction is composed of Tyr-355, Glu-524, and Arg-120 and serves as the binding site for the carboxylic acid group of substrates and certain inhibitors (25). We have reported that neutral derivatives (esters and amides) of certain carboxylic acid inhibitors (e.g., indomethacin) bind to COX-2 but not to COX-1 (26). A three-dimensional structure of COX-2 complexed to such a conjugate has not been solved but structures of related complexes suggest the indomethacin unit binds in the active site with the tethered amide, breeching the constriction and projecting into the lobby (22, 27). These structural and functional analyses provide the design principles for the construction of COX-2–targeted imaging agents.

Synthesis of candidate compounds and cellular imaging

Three carboxylic acid cores—i.e., indomethacin, a celecoxib carboxylic acid derivative, and an indolyl carboxamide analogue of indomethacin—were tethered through a series of alkylenediamines, piperazines, polyethylene glycol, or phenylenediamines to a diverse range of fluorophores. The fluorophores attached included dansyl, dabsyl, coumarin, fluorescein, rhodamine, alexa-fluor, nile blue, cy5, cy7, near IR, and IR dyes as well as lanthanide chelators. Nearly 200 compounds were synthesized and each conjugate was tested for its ability to selectively inhibit COX-2 in assays using purified proteins in vitro. Promising molecules were tested for their ability to inhibit COX-2 in LPS-treated RAW264.7 macrophages. Preliminary experiments indicated that indomethacin conjugates bound most tightly and selectively to COX-2; therefore, most of the compounds synthesized were derived from this core.

Indomethacin conjugates to dansyl, dabsyl, coumarin, fluorescein, and rhodamine-derived fluorophores exhibited promising COX-2 inhibition and selectivity both in vitro and in intact cells. The carboxy-X-rhodamine (6-ROX- and 5-ROX)–based conjugates, 1 and 2, displayed the best balance of cellular activity and optical properties (λex = 581 nm, λemit = 603 nm) and were used for all subsequent experiments (Table 1). A detailed kinetic analysis indicated that 1 and 2 require lengthy preincubations with COX-2 to achieve maximal inhibition but once bound, they dissociate very slowly (Supplementary Fig. S2). Thus, they are slow, tight-binding inhibitors with very low rates of association and dissociation. Compounds 1 and 2 were less potent than celecoxib or rofecoxib as inhibitors of COX-2 (Table 1). A negative control molecule (3) was synthesized that contained 6-ROX bound to indomethacin through a shorter ethylenediamine tether, which eliminated COX-2 inhibition (Table 1).

Table 1.

Biochemical properties of carboxy-X-rhodamine derivatives

Compound no.StructurePurified enzyme IC50 (μmol/L)Cell IC50 (μmol/L)
COX-1COX-2
graphic
 
>25 0.83 0.36 
graphic
 
>25 0.7 0.31 
graphic
 
>25 >4 ND 
graphic
 
>25 >4 ND 
graphic
 
>25 >4 ND 
graphic
 
0.92 0.21 NT 
graphic
 
>25 0.14 NT 
graphic
 
0.05 0.75 0.01 
Compound no.StructurePurified enzyme IC50 (μmol/L)Cell IC50 (μmol/L)
COX-1COX-2
graphic
 
>25 0.83 0.36 
graphic
 
>25 0.7 0.31 
graphic
 
>25 >4 ND 
graphic
 
>25 >4 ND 
graphic
 
>25 >4 ND 
graphic
 
0.92 0.21 NT 
graphic
 
>25 0.14 NT 
graphic
 
0.05 0.75 0.01 

NOTE: Assays were conducted as described in Materials and Methods. IC50s for inhibition of ovine COX-1 or human COX-2. Compounds also were tested in intact RAW264.7 macrophages (Cell IC50).

Abbreviations: ND, no inhibition detected up to 5 μmol/L; NT, not tested.

The human head and neck cancer cell line, 1483, which expresses high levels of COX-2 (28), exhibited strong labeling with compounds 1 or 2 (Fig. 1A). Preincubation of the cells with the COX-2–selective inhibitor, celecoxib, prevented the labeling of 1483 cells by either compound (Fig. 1B). In all of these in vitro experiments, the labeling seemed to be intracellular, so confocal microscopy was performed to verify the localization. Incubation of compound 2 with 1483 cells led to the perinuclear labeling of membraneous structures that appeared to be endoplasmic reticulum or Golgi (Fig. 1C). The perinuclear labeling correlated well to multiple previous reports of the intracellular localization of COX-2 (2932). Incubation of the same cells with Mitotracker showed that the mitochondria did not colocalize with compound 2 (Fig. 1C).

Figure 1.

Labeling of COX-2–expressing cells by compound 2. The experimental protocols are described in Materials and Methods. A, 1483 HNSCC cells treated with 200 nmol/L compound 2 for 30 min. B, 1483 HNSCC cells pretreated with 5 μmol/L celecoxib for 20 min before compound 2 treatment. C, confocal microscopy of 1483 HNSCC cells treated with both mitotrackerGR (blue; mitochondria) and compound 2 (red; perinuclear).

Figure 1.

Labeling of COX-2–expressing cells by compound 2. The experimental protocols are described in Materials and Methods. A, 1483 HNSCC cells treated with 200 nmol/L compound 2 for 30 min. B, 1483 HNSCC cells pretreated with 5 μmol/L celecoxib for 20 min before compound 2 treatment. C, confocal microscopy of 1483 HNSCC cells treated with both mitotrackerGR (blue; mitochondria) and compound 2 (red; perinuclear).

Close modal

The mouse macrophage–like cell line, RAW 264.7, does not express COX-2 and exhibited very weak labeling with 1 or 2 (e.g., Supplementary Fig. S3A), whereas LPS-pretreated cells labeled more strongly (Supplementary Fig. S3B). The labeling of the COX-2–expressing RAW cells by 1 or 2 was prevented by pretreatment of the cells with indomethacin (Supplementary Fig. S3C) or celecoxib, which block the COX-2 active site. Importantly, no labeling was observed when either control or LPS-pretreated RAW cells were incubated with compound 3, which does not inhibit COX-2 (Supplementary Fig. S3D). The extent of compound 2 uptake increased at 4 hours with the appearance of COX-2 protein. A further increase in uptake was not observed at 7 hours although there was higher COX-2 protein as detected by Western blotting (Supplementary Fig. S4). Comparison of the amount of compound 2 taken up at 7 hours to the amount of COX-2 estimated by Western blotting in the LPS-treated cells suggested a stoichiometry of binding of 0.90 (Supplementary Data).

Imaging carrageenan–induced inflammation

Compounds 1 and 2 seemed promising based on these in vitro imaging experiments, so their potential for in vivo imaging was evaluated using carrageenan-induced inflammation in the mouse footpad, human tumor xenografts in nude mice, and spontaneous tumors arising in mouse models. The mouse footpad model is well documented for the role of COX-2–derived prostaglandins as a major driving force for the acute edema that results 24 hours after carrageenan injection into the paw (33). One of the significant advantages of this animal model of inflammation is the ability to image the inflamed footpad compared with the noninflamed contralateral footpad, which does not express COX-2. We injected female C57BL/6 mice with 50 μL 1% carrageenan in the rear left footpad, followed by compound 1 or 2 (1 mg/kg, i.p.) at 24 hours postcarrageenan. Animals were imaged 3 hours later in a Xenogen IVIS 200 (DsRed filter, 1.5 cm depth, 1 s). Both compounds 1 and 2 targeted the swollen footpad with an average 4.5-fold increase in fluorescence over that of the contralateral, uninjected footpad (Fig. 2). For comparison, animals also were dosed with compound 3, which does not inhibit COX-2. In Fig. 2A, the left mouse was dosed with compound 3 and the right mouse with compound 1. Compound 3 yielded minimal fluorescence in the inflamed paw compared with the contralateral paw, whereas compound 1 yielded a strong signal in the inflamed paw. To test further the molecular target of compounds 1 or 2, parallel experiments were performed using mice bearing targeted deletions in COX-2. Figure 2B depicts the fold difference in the compound 2–derived fluorescence signal in the 24-hour carrageenan-injected footpad over the control footpad for wild-type versus COX-2 (−/−) mice. The COX-2 null mice consistently showed approximately a 40% increase in signal in the swollen footpad apparently due to nonspecific binding. This contrasts with a 400% to 600% increase in the swollen footpad in wild-type mice. Finally, experiments were conducted to evaluate the uptake of compound 1 during the resolution of inflammation. Following carrageenan injection, compound 1 was administered i.p. 1, 3, 5, and 7 days later and the animals were imaged. Uptake of compound 1 was maximal at 24 hours but declined thereafter (Fig. 2C). Attempts to estimate active COX-2 protein by the quantification of prostaglandins in paw extracts were unsuccessful because of poor recovery.

Figure 2.

In vivo labeling of COX-2–expression in inflammation by compound 1, 2, or 3. A, C57BL/6 mouse with carrageenan-induced inflammation in the left foot pad. The left mouse was dosed with the negative control molecule 3 (1 mg/kg, i.p.) and the right mouse was dosed with compound 1 (1 mg/kg, i.p.). Both mice were imaged at 3 h postinjection. B, fold increase of fluorescence in inflamed versus contralateral paw of wild-type (WT) and COX-2 (−/−) mice at 3 h postinjection of compound 2 (1 mg/kg, i.p.; n = 6). RFU, relative fluorescence unit. C, carrageenan was injected in the rear left footpads of female C57BL/6 mice, followed by dosing compound 1 (1 mg/kg i.p.) 24 h later. Animals were reinjected with compound 1 at 3, 5, and 7 d postcarrageenan (n = 9). Mice were imaged at 3 h after compound injection. The plot shows the fold increase of fluorescence in swollen versus contralateral foot (n = 6).

Figure 2.

In vivo labeling of COX-2–expression in inflammation by compound 1, 2, or 3. A, C57BL/6 mouse with carrageenan-induced inflammation in the left foot pad. The left mouse was dosed with the negative control molecule 3 (1 mg/kg, i.p.) and the right mouse was dosed with compound 1 (1 mg/kg, i.p.). Both mice were imaged at 3 h postinjection. B, fold increase of fluorescence in inflamed versus contralateral paw of wild-type (WT) and COX-2 (−/−) mice at 3 h postinjection of compound 2 (1 mg/kg, i.p.; n = 6). RFU, relative fluorescence unit. C, carrageenan was injected in the rear left footpads of female C57BL/6 mice, followed by dosing compound 1 (1 mg/kg i.p.) 24 h later. Animals were reinjected with compound 1 at 3, 5, and 7 d postcarrageenan (n = 9). Mice were imaged at 3 h after compound injection. The plot shows the fold increase of fluorescence in swollen versus contralateral foot (n = 6).

Close modal

Imaging COX-2–expressing tumors

The results in the footpad inflammation model show that COX-2–targeted fluorescent conjugates are taken up in inflamed paws of COX-2–expressing mice but not in COX-2 null animals. We next evaluated the ability of these compounds to target COX-2 in human tumor xenografts. Female nude mice were injected in the left flank with HCT-116 or 1483 cells and the xenografts were allowed to grow to approximately 750 to 1,000 mm3. Animals were dosed by retro-orbital injection with compound 2 (1 mg/kg) then lightly anesthetized with 2% isoflurane in preparation for imaging. No fluorescence was observed during the first 60 minutes postinjection, but signal was reproducibly detected in the COX-2–expressing 1483 tumors starting at 3 to 5 hours and persisted as long as 26 hours postinjection. At 3.5 hours postinjection, the HCT116 tumor, which does not express COX-2 (34), showed minimal fluorescence (Fig. 3A) whereas the 1483 tumor exhibited bright fluorescence (Fig. 3B). In another control experiment, nude mice bearing 1483 xenografts were treated with the fluorophore alone, 5-ROX (2 mg/kg, i.p.), which is neither an inhibitor of COX-2 nor COX-1. No signal from 5-ROX alone accumulated in the tumors at any time point. This result showed that the fluorophore moiety was not responsible for the tumor uptake of compound 2, supportingthe conclusion that the difference in labeling of 1483 and HCT116 xenografts is due their differential in COX-2 expression.

Figure 3.

In vivo labeling of COX-2–expressing xenografts by compound 2. A, nude mice with HCT116 xenograft (COX-2 negative) or 1483 xenograft (B; COX-2 positive) were dosed (retro-orbital) with 1 mg/kg compound 2 and imaged at 3.5 h postinjection. C, nude mice with 1483 xenografts were predosed with DMSO before injection of compound 2 (2 mg/kg, i.p.), or predosed with indomethacin (D; 2 mg/kg, i.p.) 24 and 1 h before compound 2 and imaged at 3 h postinjection (Xenogen IVIS, DsRed filter, 1 s, f2, 1.5 cm depth). The emission observed around the peritoneal cavity in C and D is due to residual compound 2 at the site of injection.

Figure 3.

In vivo labeling of COX-2–expressing xenografts by compound 2. A, nude mice with HCT116 xenograft (COX-2 negative) or 1483 xenograft (B; COX-2 positive) were dosed (retro-orbital) with 1 mg/kg compound 2 and imaged at 3.5 h postinjection. C, nude mice with 1483 xenografts were predosed with DMSO before injection of compound 2 (2 mg/kg, i.p.), or predosed with indomethacin (D; 2 mg/kg, i.p.) 24 and 1 h before compound 2 and imaged at 3 h postinjection (Xenogen IVIS, DsRed filter, 1 s, f2, 1.5 cm depth). The emission observed around the peritoneal cavity in C and D is due to residual compound 2 at the site of injection.

Close modal

Nude mice with 1483 xenografts were pretreated with either DMSO or indomethacin in DMSO (2 mg/kg, i.p.) before compound 2 dosing (2 mg/kg, i.p.). At 3 hours postinjection, the DMSO-pretreated mice showed strong fluorescence in their tumors (Fig. 3C) compared with weak signals in the tumors of the indomethacin-pretreated mice (Fig. 3D). In the mouse xenograft model, indomethacin was able to block 92 ± 6% (n = 8) of the COX-2–expressing tumor uptake of compound 2.

We next investigated whether the COX-2 inhibitory activity of our imaging probes correlated with their in vivo efficacy in targeting COX-2–expressing tumors. Nude mice bearing 1483 xenografts were dosed (2 mg/kg, i.p.) with compound 4 (no COX inhibition at 3 μmol/L), compound 5 (30% COX-2 inhibition at 3 μmol/L), and compound 2 (90% COX-2 inhibition at 3 μmol/L; Table 1). At 3.5 hours postinjection, fluorescence from the tumor region was directly proportional to the compound potency as a COX-2 inhibitor (Supplementary Fig. S5).

Experiments were conducted to determine the identity of the fluorescent material(s) detected in vivo and to monitor the time course of its distribution and tissue uptake following injection of compound 2 into nude mice bearing 1483 human tumor xenografts. Extracts of plasma, liver, kidney, tumor, and adjacent muscle were quantitatively analyzed by HPLC at different times after i.p. administration of the compound. A single fluorescent compound was detected in all the extracts, which coeluted with a standard of compound 2 in multiple HPLC systems. This compound displayed an identical mass spectrum to the unmetabolized parent molecule, compound 2 (Fig. 4A). The time courses of uptake and distribution of compound 2 in plasma and various tissues are displayed in Fig. 4B. Compound 2 was rapidly distributed following i.p. administration and reached nearly maximal levels in plasma, liver, and kidney 30 minutes after injection. Compound 2 levels declined substantially over the course of 12 to 24 hours to a small fraction of its initial levels in all three of these compartments. In contrast, the time course for uptake of compound 2 into the 1483 tumors lagged substantially and required ∼3 hours to reach near maximal levels. The levels of compound 2 remained relatively high in the tumor so by 24 hours, the tumor levels were as high as the levels in liver or kidney. This indicates both slow uptake and release of compound 2 into and out of the tumor.

Figure 4.

Analysis of fluorescent material in xenografts and several mouse tissues. A, representative HPLC-UV chromatogram (detection, 581 nm) of 1483 tumor extract (4 h postadministration) revealed a single major fluorescent compound that coeluted with compound 2 (15.3 min). The peak eluting at 13.3 min integrates for <5% of the peak at 15.3 min. Nonspecific fluorescent peaks were eluted at or near the void volume of the column. Inset, the Q1 mass spectrum of the extracted fluorescent material was identical to that of an authentic standard of compound 2. B, time course and distribution of compound 2 in various mouse tissues in vivo.

Figure 4.

Analysis of fluorescent material in xenografts and several mouse tissues. A, representative HPLC-UV chromatogram (detection, 581 nm) of 1483 tumor extract (4 h postadministration) revealed a single major fluorescent compound that coeluted with compound 2 (15.3 min). The peak eluting at 13.3 min integrates for <5% of the peak at 15.3 min. Nonspecific fluorescent peaks were eluted at or near the void volume of the column. Inset, the Q1 mass spectrum of the extracted fluorescent material was identical to that of an authentic standard of compound 2. B, time course and distribution of compound 2 in various mouse tissues in vivo.

Close modal

APCmin mice bear the same mutation (Apc−) that is causative for familial adenomatous polyposis in human beings and these mice primarily develop small intestinal tumors that express COX-2 (35, 36). Crossing APCmin mice with COX-2 (−/−) mice reduces intestinal tumor development by 85% and treatment of APCmin mice with COX-2 inhibitors also reduces tumorigenesis (37, 38). APCmin mice (ages 18–20 wk, fed a high-fat diet) were injected retro-orbitally with compound 2 (1 mg/kg), and after 2 hours, animals were sacrificed and their intestines were removed. The tissue was washed thoroughly with PBS, opened longitudinally, and imaged. Figure 5A shows the low background fluorescence of a section of small intestine without polyps. A single polyp (Fig. 5B) and a five-polyp cluster (Fig. 5C) displayed high fluorescence, with greatly increased detection compared with bright-field visualization. The signal enrichment of compound 2 in the polyps was estimated to be >50:1. COX-2 expression in the polyps seems to be required for this selective uptake although other factors beside the level of COX-2 protein may contribute to the relative enrichment over surrounding normal tissue.

Figure 5.

In vivo labeling of COX-2–expression in intestinal polyps by compound 2. C57BL/6J-Min/+ mice bearing small intestinal polyps were euthanized at 2 h after retro-orbital injection of compound 2 (1 mg/kg) and small intestines were washed, opened, and examined by dissecting fluorescence microscopy. A, section of small intestine with no polyp, 90-millisecond exposure. B, single polyp, 90-millisecond exposure. C, polyp cluster, 90-millisecond exposure.

Figure 5.

In vivo labeling of COX-2–expression in intestinal polyps by compound 2. C57BL/6J-Min/+ mice bearing small intestinal polyps were euthanized at 2 h after retro-orbital injection of compound 2 (1 mg/kg) and small intestines were washed, opened, and examined by dissecting fluorescence microscopy. A, section of small intestine with no polyp, 90-millisecond exposure. B, single polyp, 90-millisecond exposure. C, polyp cluster, 90-millisecond exposure.

Close modal

These studies show the feasibility of specific in vivo targeting of COX-2 in inflammatory lesions and tumors using organic fluorophores tethered to indomethacin through an amide linkage. Compounds 1 and 2 display a very high degree of selectivity of uptake by inflammatory tissue and tumors in live animals relative to surrounding normal tissue or muscle as determined by either imaging or mass spectrometry. This selectivity seems greater than that reported in previous literature reports of fluorescent tumor imaging in which the ratio of tumor fluourescence was compared with muscle fluorescence (39). Uptake of our compounds requires the expression of COX-2 at the target site and declines as the level of COX-2 decreases. Although uptake into inflamed or tumor tissue seems to be slower than expected from simple distribution in the body, the kinetics of compound release seem to be extremely slow, thus leading to a detectable buildup of the label. Similar results are observed by both imaging compounds 1 or 2 (Figs. 2 and 3) or by direct quantitative analysis of compound 2 (Supplementary Fig. S4; Fig. 4).

To achieve this success, ~200 compounds were evaluated as candidate COX-2–targeted imaging agents. Although a significant percentage showed COX-2 inhibitory activity against purified protein, only a fraction of these compounds inhibited COX-2 activity in intact cells, and of those, most did not possess fluorescence properties suitable for in vivo imaging. Among the compounds that emerged from our development pathway, only compounds 1 and 2 exhibited sufficient metabolic stability to survive long enough to distribute to inflammatory lesions or xenograft tumors. The low overall success rate (∼1%) likely underscores why COX-2–targeted imaging agents have proven difficult to develop.

The specificity for COX-2 binding of these compounds was illustrated by multiple observations: (a) only cultured cells that express COX-2 took up fluorocoxibs and uptake was inhibited by the COX inhibitors indomethacin and celecoxib. The intracellular localization of the probes matches that of COX-2 protein and the stoichiometry of uptake was ∼0.9 molecule of beacon per subunit; (b) uptake into inflamed over noninflamed tissue was blocked by indomethacin pretreatment of the animals and was not observed in COX-2 (−/−) animals. No uptake was observed with a close structural analogue of compound 1 that does not inhibit COX-2; (c) uptake into COX-2–expressing tumors was blocked by indomethacin pretreatment of the animals and a correlation was found between the amount of light emission from the tumor and the COX-2 inhibitory potency of the beacon. The nontargeted fluorophores 5-ROX and 6-ROX did not accumulate in COX-2–expressing xenografts; and (d) >95% of the fluorescent material present in the tumors is the unmetabolized parent compound. Thus, in vitro and in vivo studies provide strong support for the conclusion that binding to COX-2 is the major determinant of uptake into inflamed, premalignant, or malignant tissue. Although the stoichiometry of compound 2 binding to COX-2 protein was estimated to be 0.9 in activated RAW cells, such high stoichiometry cannot be assumed in all situations. The extent of uptake in cells, inflamed tissue, or tumors will depend upon several factors such as the permeability of COX-2–expressing cells to the probe, kinetics of binding and release from the COX-2 active site, vascularization of the tissue, and possible expulsion of the probes by transporters. Further studies will be needed to explore in greater detail the quantitative aspects of the use of these compounds in vivo.

Compounds 1 and 2 represent the first feasible reagents for clinical detection of tissues containing high levels of COX-2 in settings amenable to fluorescent excitation and analysis by surface measurement or endoscopy (e.g., skin, esophagus, intestine, and bladder). Although such fluorocoxibs will not be useful for applications in internal organs that are not accessible for optical imaging, their development provides rigorous proof of concept for the feasibility of molecular targeting of COX-2 in inflammatory lesions, premalignant lesions, and tumors.

L.J. Marnett: sponsored research and consulting, XL Tech Group. The other authors disclosed no potential conflicts of interest.

We thank S.K. Dey for the COX-2 null animals and Melissa Turman for the assistance with molecular graphics.

Grant Support: Research and Center grants from the NIH (CA86283, CA89450, CA105296, CA68485, CA60867, CA126588, CA111469, and GM72048), the Medical Free-Electron Laser Program of the US Department of Defense, XL TechGroup, and New York Crohn's Foundation.

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

1
Weissleder
R
,
Ntziachristos
V
. 
Shedding light onto live molecular targets
.
Nat Med
2003
;
9
:
123
8
.
2
Tanabe
K
,
Zhang
Z
,
Ito
T
,
Hatta
H
,
Nishimoto
S
. 
Current molecular design of intelligent drugs and imaging probes targeting tumor-specific microenvironments
.
Org Biomol Chem
2007
;
5
:
3745
57
.
3
Crofford
LJ
. 
COX-1 and COX-2 tissue expression: implications and predictions
.
The J Rheumatol
1997
;
49
:
15
9
.
4
Dannenberg
AJ
,
Lippman
SM
,
Mann
JR
,
Subbaramaiah
K
,
DuBois
RN
. 
Cyclooxygenase-2 and epidermal growth factor receptor: pharmacologic targets for chemoprevention
.
J Clin Oncol
2005
;
23
:
254
66
.
5
Marnett
LJ
. 
The COXIB experience: a look in the rear-view mirror
.
Annu Rev Pharmacol Toxicol
2009
;
49
:
265
90
.
6
Kandil
HM
,
Tanner
G
,
Smalley
W
,
Halter
S
,
Radhika
A
,
Dubois
RN
. 
Cyclooxygenase-2 expression in Barrett's esophagus
.
Dig Dis Sci
2001
;
46
:
785
9
.
7
Eberhart
CE
,
Coffey
RJ
,
Radhika
A
,
Giardiello
FM
,
Ferrenbach
S
,
DuBois
RN
. 
Up-regulation of cyclooxygenase 2 gene expression in human colorectal adenomas and adenocarcinomas
.
Gastroenterology
1994
;
107
:
1183
8
.
8
Kargman
SL
,
O'Neill
GP
,
Vickers
PJ
,
Evans
JF
,
Mancini
JA
,
Jothy
S
. 
Expression of prostaglandin G/H synthase-1 and -2 protein in human colon cancer
.
Cancer Res
1995
;
55
:
2556
9
.
9
Edelman
MJ
,
Watson
D
,
Wang
X
, et al
. 
Eicosanoid modulation in advanced lung cancer: cyclooxygenase-2 expression is a positive predictive factor for celecoxib + chemotherapy-Cancer and Leukemia Group B Trial 30203
.
J Clin Oncol
2008
;
26
:
848
55
.
10
Toyokuni
T
,
Kumar
JS
,
Walsh
JC
, et al
. 
Synthesis of 4-(5-[18F]fluoromethyl-3-phenylisoxazol-4-yl)benzenesulfonamide, a new [18F]fluorinated analogue of valdecoxib, as a potential radiotracer for imaging cyclooxygenase-2 with positron emission tomography
.
Bioorg Med Chem Lett
2005
;
15
:
4699
702
.
11
Tanaka
M
,
Fujisaki
Y
,
Kawamura
K
, et al
. 
Radiosynthesis and evaluation of 11C-labeled diaryl-substituted imidazole and indole derivatives for mapping cyclooxygenase-2
.
Biol Pharm Bull
2006
;
29
:
2087
94
.
12
Majo
VJ
,
Prabhakaran
J
,
Simpson
NR
,
Van Heertum
RL
,
Mann
JJ
,
Kumar
JS
. 
A general method for the synthesis of aryl [11C]methylsulfones: potential PET probes for imaging cyclooxygenase-2 expression
.
Bioorg Med Chem Lett
2005
;
15
:
4268
71
.
13
Prabhakaran
J
,
Underwood
MD
,
Parsey
RV
, et al
. 
Synthesis and in vivo evaluation of [18F]-4-[5-(4-methylphenyl)-3-(trifluoromethyl)-1H-pyrazol-1-yl]benzenesulfonamide as a PET imaging probe for COX-2 expression
.
Bioorg Med Chem
2007
;
15
:
1802
7
.
14
Wuest
F
,
Kniess
T
,
Bergmann
R
,
Pietzsch
J
. 
Synthesis and evaluation in vitro and in vivo of a 11C-labeled cyclooxygenase-2 (COX-2) inhibitor
.
Bioorg Med Chem
2008
;
16
:
7662
70
.
15
Schuller
HM
,
Kabalka
G
,
Smith
G
,
Mereddy
A
,
Akula
M
,
Cekanova
M
. 
Detection of overexpressed COX-2 in precancerous lesions of hamster pancreas and lungs by molecular imaging: implications for early diagnosis and prevention
.
Chem Med Chem
2006
;
1
:
603
10
.
16
de Vries
EF
,
van Waarde
A
,
Buursma
AR
,
Vaalburg
W
. 
Synthesis and in vivo evaluation of 18F-desbromo-DuP-697 as a PET tracer for cyclooxygenase-2 expression
.
J Nucl Med
2003
;
44
:
1700
6
.
17
de Vries
EF
,
Doorduin
J
,
Dierckx
RA
,
van Waarde
A
. 
Evaluation of [11C]rofecoxib as PET tracer for cyclooxygenase 2 overexpression in rat models of inflammation
.
Nucl Med Biol
2008
;
35
:
35
42
.
18
de Vries
EF
. 
Imaging of cyclooxygenase-2 (COX-2) expression: potential use in diagnosis and drug evaluation
.
Curr Pharm Des
2006
;
12
:
3847
56
.
19
Timofeevski
SL
,
Prusakiewicz
JJ
,
Rouzer
CA
,
Marnett
LJ
. 
Isoform-selective interaction of cyclooxygenase-2 with indomethacin amides studied by real-time fluorescence, inhibition kinetics, and site-directed mutagenesis
.
Biochemistry
2002
;
41
:
9654
62
.
20
Sacks
PG
,
Parnes
SM
,
Gallick
GE
, et al
. 
Establishment and characterization of two new squamous cell carcinoma cell lines derived from tumors of the head and neck
.
Cancer Res
1988
;
48
:
2858
66
.
21
Picot
D
,
Loll
PJ
,
Garavito
RM
. 
The X-ray crystal structure of the membrane protein prostaglandin H2 synthase-1
.
Nature
1994
;
367
:
243
9
.
22
Luong
C
,
Miller
A
,
Barnett
J
,
Chow
J
,
Ramesha
C
,
Browner
MF
. 
Flexibility of the NSAID binding site in the structure of human cyclooxygenase-2
.
Nature Struct Biol
1996
;
3
:
927
33
.
23
Kurumbail
RG
,
Stevens
AM
,
Gierse
JK
, et al
. 
Structural basis for selective inhibition of cyclooxygenase-2 by anti-inflammatory agents
.
Nature
1996
;
384
:
644
8
.
24
Kurumbail
RG
,
Kiefer
JR
,
Marnett
LJ
. 
Cyclooxygenase enzymes: catalysis and inhibition
.
Curr Opin Struct Biol
2001
;
11
:
752
60
.
25
Bhattacharyya
DK
,
Lecomte
M
,
Rieke
CJ
,
Garavito
RM
,
Smith
WL
. 
Involvement of arginine 120, glutamate 524, and tyrosine 355 in the binding of arachidonate and 2-phenylpropionic acid inhibitors to the cyclooxygenase active site of ovine prostaglandin endoperoxide H synthase-1
.
J Biol Chem
1996
;
271
:
2179
84
.
26
Kalgutkar
AS
,
Crews
BC
,
Rowlinson
SW
, et al
. 
Biochemically based design of cyclooxygenase-2 (COX-2) inhibitors: facile conversion of nonsteroidal antiiflammatory drugs to potent and highly selective COX-2 inhibitors
.
Proc Natl Acad Sci USA
2000
;
97
:
925
30
.
27
Harman
CA
,
Turman
MV
,
Kozak
KR
,
Marnett
LJ
,
Smith
WL
,
Garavito
RM
. 
Structural basis of enantioselective inhibition of cyclooxygenase-1 by S-α-substituted indomethacin ethanolamides
.
J Biol Chem
2007
;
282
:
28096
105
.
28
Zweifel
BS
,
Davis
TW
,
Ornberg
RL
,
Masferrer
JL
. 
Direct evidence for a role of cyclooxygenase 2-derived prostaglandin E2 in human head and neck xenograft tumors
.
Cancer Res
2002
;
62
:
6706
11
.
29
Regier
MK
,
DeWitt
DL
,
Schindler
MS
,
Smith
WL
. 
Subcellular localization of prostaglandin endoperoxide synthase-2 in murine 3T3 cells
.
Arch Biochem Biophys
1993
;
301
:
439
44
.
30
Coffey
RJ
,
Hawkey
CJ
,
Damstrup
L
, et al
. 
Epidermal growth factor receptor activation induces nuclear targeting of cyclooxygenase-2, basolaterial release of prostaglandins, and mitogenesis in polarizing colon cancer cells
.
Proc Natl Acad Sci USA
1997
;
94
:
657
62
.
31
Spencer
AG
,
Woods
JW
,
Arakawa
T
,
Singer
II
,
Smith
WL
. 
Subcellular localization of prostaglandin endoperoxide H synthases-1 and -2 by immunoelectron microscopy
.
J Biol Chem
1998
;
273
:
9886
93
.
32
Mbonye
UR
,
Wada
M
,
Rieke
CJ
,
Tang
HY
,
Dewitt
DL
,
Smith
WL
. 
The 19-amino acid cassette of cyclooxygenase-2 mediates entry of the protein into the endoplasmic reticulum-associated degradation system
.
J Biol Chem
2006
;
281
:
35770
8
.
33
di Meglio
P
,
Ianaro
A
,
Ghosh
S
. 
Amelioration of acute inflammation by systemic administration of a cell-permeable peptide inhibitor of NF-κB activation
.
Arthritis and rheumatism
2005
;
52
:
951
8
.
34
Sheng
HM
,
Shao
JY
,
Kirkland
SC
, et al
. 
Inhibition of human colon cancer cell growth by selective inhibition of cyclooxygenase-2
.
J Clin Invest
1997
;
99
:
2254
9
.
35
Moser
AR
,
Pitot
HC
,
Dove
WF
. 
A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse
.
Science
1990
;
247
:
322
4
.
36
Williams
CS
,
Luongo
C
,
Radhika
A
, et al
. 
Elevated cyclooxygenase-2 levels in Min mouse adenomas
.
Gastroenterology
1996
;
111
:
1134
40
.
37
Oshima
M
,
Dinchuk
JE
,
Kargman
S
, et al
. 
Suppression of intestinal polyposis in ApcΔ716 knockout mice by inhibition of cyclooxygenase 2 (COX-2)
.
Cell
1996
;
87
:
803
9
.
38
Jacoby
RF
,
Seibert
K
,
Cole
CE
,
Kelloff
G
,
Lubet
RA
. 
The cyclooxygenase-2 inhibitor celecoxib is a potent preventive and therapeutic agent in the min mouse model of adenomatous polyposis
.
Cancer Res
2000
;
60
:
5040
4
.
39
Bugaj
JE
,
Achilefu
S
,
Dorshow
RB
,
Rajagopalan
R
. 
Novel fluorescent contrast agents for optical imaging of in vivo tumors based on a receptor-targeted dye-peptide conjugate platform
.
J Biomed Opt
2001
;
6
:
122
33
.

Supplementary data