Abstract
In recent years, numerous in vivo molecular imaging probes have been developed. As a consequence, much has been published on the design and synthesis of molecular imaging probes focusing on each modality, each type of material, or each target disease. More recently, second generation molecular imaging probes with unique, multi-functional, or multiplexed characteristics have been designed. This critical review focuses on (i) molecular imaging using combinations of modalities and signals that employ the full range of the electromagnetic spectra, (ii) optimized chemical design of molecular imaging probes for in vivo kinetics based on biology and physiology across a range of physical sizes, (iii) practical examples of second generation molecular imaging probes designed to extract complementary data from targets using multiple modalities, color, and comprehensive signals (277 references).
A. Introduction
Conventional diagnostic imaging methods employing contrast agents, such as angiography, computed tomography, and magnetic resonance (MR) imaging have had a profound impact on medicine.1 Yet, improvements are needed in sensitivity, resolution and specificity. Radionuclide imaging provides a good example of current limitations. While radionuclide imaging provides physiologic information (blood flow, perfusion, lymphatic flow) or metabolic information (phosphate, sugar, nucleic acid, and some amino acids), it does so with low spatial resolution. Only a few molecularly targeted radioactive probes (somatostatin analog and radiolabeled antibodies) have been approved for use in clinical practice. Molecular imaging, seen as one of the next important advances in imaging sciences, offers the possibility of obtaining in vivo target-specific information with high sensitivity and specificity.2–6 To achieve this goal it may be necessary to design probes that mix different modalities, probes, or signal processing.
Molecular imaging has had few clinical successes to date but there have been considerable preclinical advances in oncology,5,7–11 cardiology,2,12–16 and neuroscience.10,17–22 Thus, the field is widely considered still to be in its infancy. One factor limiting molecular imaging is that most currently used probes are monochromatic (yielding only one signal type per molecule) and emit signal continuously (so called “always on”). Monochromatic imaging, although quite useful in current clinical practice, yields linear, uniparametric data. This results in a “one test–one answer” paradigm that provides only a unidimensional piece of the puzzle resulting in “many tests–many answers”. The problem with “always on” probes is that they diminish target to background ratios secondary to non-specific background signal. This dramatically reduces sensitivity even while the probe may be highly specific. The ability to generate polychromatic, multi-parametric data, and activatable imaging probes will provide more rich and complex data sets with higher sensitivity and specificity.23–27 Innovations in chemistry and nanotechnology will lead to develop highly targeted probes28–30 that are optimized for the high affinity binding to specific molecular targets and their signaling properties, and eventually yield multiparametric data. Such probes may not only be more informative but may also result in improved efficiencies with regard to delivering medical care, thus lowering costs (one test–many answers)31,32
Each modality offers specific capabilities such as high spatial resolution, high temporal resolution or high sensitivity; and each utilizes electromagnetic waves from different parts of the spectrum for data acquisition followed by data reconstruction. With each technology, there are several approaches to obtaining multi-parametric data: (1) detect two or more photons from different parts of the electromagnetic spectrum using two or more different imaging modalities (multi-modality);33,34 (2) detect two or more photons from the same part of the spectrum, and use spectral separation techniques to differentiate the signals (multi-color) and (3) detect photons of the same energy but extract additional information (e.g. time domain information) by signal processing (multiple signaling) (Fig. 1).
Fig. 1.
Schematic explanation of multiplexed imaging technologies based on the electromagnetic waves of different wavelengths.
An important underlying principle of probe design is to optimize tumor to background ratio (TBR). This can be achieved either by maximizing signal from the target, minimizing signal from the background, or both. Improving TBR increases the sensitivity and specificity for detecting target molecules in vivo. Strategies for optimizing TBR can be considered at several scales, from the whole organism level to the atomic level. At the organism level, the size of the animal (e.g. mouse vs. human) should be considered as the depth of signal penetration varies between probe types, most notably for optical imaging, and therefore the depth of target tissue from the surface of the organism is an important component of detected signal strength. At the organ level, physiology should be considered, given that probe pharmacokinetics, including the uptake of the agent, its breakdown, clearance, and excretion all affect TBR. On a cellular level target expression, binding affinity, on and off rates, intracellular processing and catabolism can affect probe signal.9,35,36 On a molecular level, the physical interaction with the target and chemical or enzymatic processing alter probe signal and therefore TBR. Finally, at the atomic level, inter- or intra-molecular energy transfer, photon-induced electron transfer, and caging are significant means of achieving signal activation and/or signal amplification and therefore warrant consideration (Fig. 2).
Fig. 2.
A schema of the rational strategies for target specific imaging in all physical levels.
In this article, we discuss design strategies for molecular imaging probes emphasizing multi-modal, multi-color and multi-signal strategies.
B. Multiplexed data acquisition
Each modality has strengths and limitations. Therefore, the simultaneous use of two or more modalities, imaging reagents or signaling methods, to achieve “multiplexed imaging,” could overcome the limitations of each modality and improve the data obtained during a single imaging session. An excellent current example is positron emission tomography/computerized tomography (PET/CT) wherein the PET provides a high sensitivity metabolic image while the CT provides anatomic information on which the metabolic information can be superimposed. An example of multi-color imaging includes the simultaneous detection of different targets, using single-photon emission computerized tomography (SPECT) imaging cameras which can resolve multiple tracers based on different emission energies (i.e. Tc-99m (140 keV) and Tl-201 (71 keV)) using two different energy detection windows to gather multi-parametric information. A current example of multiple signaling is MR imaging in which T1-, T2-, and diffusion-weighted images are obtained in one session, each reflecting a different tissue signal. Thus, the template for the future is already in place. The design of the next generation of molecular imaging probes should take into account whether the agent will provide multiplexed imaging data. In this section, we review rational designs and practical examples of these different types of probes not yet in the clinic.
B.1 Multi-modality imaging probes
As shown in Fig. 1, nuclear imaging employs photons which emit gamma rays at wavelengths less than 1 nm. Among these photons, those with energies greater than 70 keV can generally penetrate through the human body without significant attenuation. From a practical perspective, radionuclides, which emit photons with energies ranging from 80 keV to 350 keV, are appropriate for molecular imaging but require extensive collimation for accurate reconstruction. This results in inefficient photon collection because many photons are absorbed by the collimator. Positron emitting radionuclides produce two 511 keV photons (in opposite directions) that can be detected with co-incident detectors and because the line of response can be determined, PET scanners do not require a collimator, resulting in improved resolution compared with gamma emitters. Radionuclide cameras are sensitive enough to detect pico-molar concentrations of radionuclides even deep in the body. Nuclear imaging is a well-developed area of medicine, which has evolved over the past fifty years. As a result, newly developed agents can be readily tested in humans using a micro-dosing processing. However, given that gamma- or X-ray exposes the subject to ionizing radiation, radionuclide imaging must always take into consideration radiation dose based on the radioactivity of the agent and its physical and biological half life. Thus, an imaging modality with comparable sensitivity but without ionizing radiation would be desirable.
Optical imaging relies on photons ranging in wavelength from the visible to the near infrared (NIR) (500 to 1500 nm wavelength). Charge Coupled Device (CCD) detectors, which are used for optical imaging, generally show better sensitivity for photons at shorter wavelengths. Within this range, NIR light, ranging from 650 to 800 nm, provides the best visualization of deeper structures, up to several centimeters, from the surface, as this range is less readily absorbed by living tissue (Fig. 3). Shorter wavelength photons are limited to imaging the surface or immediately subsurface phenomena because of near complete absorbance of light by oxy- and deoxyhemoglobin. Of course, depth of penetration is partly related to the strength of the light source, therefore, the signal from fluorophores with very strong emission can penetrate tissue more deeply. Numerous visible-to-NIR wavelength emitting agents are used for optical imaging and such fluorophores can be detected with pico-molar sensitivity without ionizing radiation. Optical imaging alone (without exogenous probes) has been used in clinical practice for detecting blood flow or blood oxygen concentration in relatively superficial organs including the breasts, brain, and eyes. Only two optical imaging reagents; fluorescein and indocyanine green (ICG), have been approved for use in clinical practice. Small numbers of target specific fluorescent probes have been introduced for use during surgery- or endoscopy, but none is in common clinical use.
Fig. 3.
A schema of the in vivo NIR window and the extinction coefficient value of water, oxy- and deoxy-hemoglobin are plotted ranging from visible to near infrared wavelength.
MR imaging relies on radio waves which have wavelengths on the order of 1 centimetre. Radiofrequency waves can penetrate deep into the body. MR imaging generally measures the magnetic relaxation properties of protons but this requires excitation by radiofrequency. Paramagnetic metal ions or nano-sized metal particles are used as contrast agents as they alter the relaxation rate of adjacent water protons and disrupt the magnetic field around particles. Detectable relaxation change can only be achieved when the paramagnetic concentration of the imaging agent is greater than 10 micro-molar concentration of metal ions or atoms, thus limiting sensitivity. Radio waves do not expose the subject to ionizing radiation. With regard to contrast agents, gadolinium chelates and iron oxide particles have been approved for use in the clinical practice. However, there is mounting concern regarding the use of gadolinium contrast agents in patients with renal impairment due to the risk of nephrogenic systemic sclerosis, a potentially fatal disorder that results from heavy metal toxicity of gadolinium ions that are released from chelates. With this said, MRI signal can be processed in different ways yielding methods that have a great potential for obtaining multiplexed information beyond anatomy and without the need for contrast material.
By keeping these characteristics in mind, appropriate combinations of two or more signaling moieties, also called “hybrid imaging”, can be effective in providing more complete information about a physiologic process. In the next sections, new molecular probe development, mostly presented as preclinical examples, is discussed.
B.1.1 Nuclear/optical multi-modality probes
Multimodal nuclear and optical imaging probes are promising agents for molecular imaging and drug (theranostic) development.37–42 Within this strategy, the role of the radioactive component is to provide quantitative, whole body data. The radioactive component provides a continuously emitting signal that can be utilized to determine probe biodistribution and clearance and provide physiologic data such as glomerular filtration rate and probe concentrations in specific sites (such as in a tumor or organ).43–45 The optical component of the probe provides the ability to qualitatively visualize target tissue in real time to assist with interventional, surgical, or endoscopic procedures, or for specific detection of molecular profiles using multi-color “always on” or tumor-cell specific “activatable” optical probes (discussed later). These properties make nuclear/optical probes useful for drug development, physiologic imaging,46 enzyme-specific imaging,37 cardiac imaging, and for effective tumor targeting.38,39,47,48
B.1.1.1 Nuclear/optical dual labeled probes
The use of a positron emitting isotope as the radioactive component, provides even better quantitative data for dual nuclear/optical agents.49 PET has high penetration and sensitivity and is more quantitative.50,51 With this in mind, several combinations of PET/optical agents have been developed. For example, an optical quantum dot (QD) with NIR fluorescence can be attached to a chelating agent to bind 64Cu for PET imaging, and to a vascular endothelial growth factor (VEGF) protein analog for site-specific targeting of the VEGF receptor. The pharmacokinetic properties and efficacy of this dual labeled probe were evaluated for tumor-specific imaging in an early clinical trial.50 The PET agent provided an overall view of VEGF receptor positive tumors whereas the optical imaging provided real time localization of the tumor for resection.
Another example of a dual radionuclide–optical probe is a trastuzumab (anti HER2 antibody) labeled with a positron-emitting radionuclide and a NIR fluorophore which was tested in a mouse model of breast cancer.52 Bimodal nuclear/optical agents possess promise for providing sensitive disease detection over the whole body (nuclear), quantitative data regarding pharmacokinetics (nuclear), as well as the ability to achieve real time, multi-color, highly specific molecular targeting and cellular-level imaging (optical). Practically speaking, the similar sensitivities of the two modalities make it easier to design and apply these probes together (Fig. 4).
Fig. 4.
Multi-color and quantitative lymphatic images using a nuclear and NIR optical dual labeled imaging agent are shown. As depicted in this schema, generation-6 PAMAM dendrimer-based dual labeled contrast agents of approximately 10 nm in diameter are employed. A gamma scintigram camera shows quantitative lymphatic drainage and a 5-color optical image shows distinct drainages from different injection sites of agents.
B.1.1.2 Cerenkov luminescence imaging (CLI)
A recent development in bi-modal optical/nuclear imaging is the use of Cerenkov Radiation (CR) for optical imaging.53–58 CR occurs when a charged particle travels faster than the speed of light through an insulating medium.53,59–61 The relevance to molecular and optical imaging is that positron-emitting radionuclides used in biomedical imaging have sufficient energy to result in CR that can be detected in vivo using optical imaging equipment.59,60 Currently used positron emitting radionuclides include: fluorine-18, oxygen-15, gallium-68 and iodine-124; and other possible beta-ray emitters include: iodine-131, strontium-89, yttrium-90 and copper-67.58,59 These agents are mostly used for radionuclide therapy but they may be used in the future for theranostic imaging as well. Unlike typical fluorescence, which results in a peaked spectra, CR spectra are continuous with higher frequencies related to higher energy conditions.57,61 The photons produced by this process have been detected in mice using CCD optical imaging systems.53 However, the light produced with CR is mostly in the ultraviolet and blue range where it is highly absorbed, thus limiting its usefulness for in vivo optical imaging. However, this limitation has been overcome using a multimodal strategy in which Cerenkov radiation serves as an energy donor for an adjacent fluorophore in a process termed Cerenkov radiation energy transfer (CRET).61 By coupling PET isotopes and quantum dots which can be excited by a broad range of wavelengths and produce fluorescent light that has a high Stokes-shift (i.e. is much higher in wavelength than the excitation light), Cerenkov radiation can induce light which is relatively easy to detect.61
B.1.2 MR/optical multi-modality probes
Another approach to multimodal imaging is to combine MR and optical imaging capabilities.62 In this combination, MR offers detailed anatomic imaging, while the optical component offers the option of real-time molecular targeting that can be used for image guided procedures. Potential clinical applications include preoperative MR imaging followed by optical image-guided surgical intervention during the treatment of cancer. One general challenge in the development of these probes is the >1000 fold discrepancy in detection sensitivity that exists between MR (low sensitivity) and optical imaging (high sensitivity). A much higher concentration of the MR agent is needed to achieve successful imaging compared to the optical agent. Another challenge is that MR is inherently a three dimensional imaging method whereas optical imaging is generally a projection imaging method. Therefore the optical image is often superimposed on the surface of the MR image. Creating a bimodal MR/optical agent that allows for superimposed images and that incorporates enough paramagnetic signal to generate an image yet does not overwhelm the optical signal can be challenging.
B.1.2.1 MR/optical dual-labeled probes
Numerous dual function MR/optical probes have been developed.63–68 Three primary approaches are currently used to synthesize such probes: (1) iron oxide nanoparticles functionalized with fluorescent molecules, (2) biocompatible molecules that are inherently paramagnetic yet also have optical properties, (3) nanomaterials specifically designed for both MR and optical imaging.
The use of supraparamagnetic iron oxide (IO) nanoparticles such as small particle of IO, ultra-small particle of IO, magnetic IO nanoparticle and cross-linked IO is commonly employed as a platform for dual MR/optical probes. IO nanoparticles have the advantage of being clinically available, with favorable toxicity profiles and are metabolized using well known iron salvage pathways that have been conserved through evolution.69–71 Additionally, IO nanoparticles (NPs) demonstrate different pharmacokinetic behaviors in vivo based on their size, and the size can be tuned for specific applications.72–77 For instance, IOs which are over 100 nm in diameter are quickly recognized by the reticuloendothelial system (RES) and are quickly cleared from the plasma to the liver, spleen, and bone-marrow.72 These organs are therefore, logical targets for such agents. In contrast, macromolecules smaller than 30 nm in diameter are taken up less rapidly by the RES and therefore, have longer circulating half lives and accumulate with enhanced permeation and retention (EPR) within tumors.72,78 In addition, cell surface chemistry modification also affects biodistribution and specific alterations can be made as needed.73,79 Applications of bimodal MR/optical probes have been numerous and varied.62 In one paradigm, preoperative MR imaging is used to confirm the diagnosis of an implanted brain tumor in a rat and then intraoperative real time NIR fluorescence imaging is used to discriminate cancer tissue from normal brain tissue during resection.80
An important drawback of iron oxide nanoparticles is that they produce a negative signal on proton-based MR imaging and this is more difficult to detect than a positive signal. Another design strategy for MR/optical probes is to functionalize molecules or particles (through encapsulation or linking) with traditional small molecule gadolinium chelates, thus making the molecule paramagnetic, so that it produces a positive signal. Although little toxicity data are currently available, organic molecules may prove to be more biocompatible and therefore may be more relevant clinically. Organic molecules capable of linking to paramagnetic chelates include dendrimers,63,81–83 nanomicelles64,84–88 liposomes89–93 viral capsids,94–97 and even natural products such as functionalized low density lipoprotein particles.69,98 Chelating the gadolinium with a strong macrocyclic chelate greatly reduces the chance of toxicity due to the release of free gadolinium ion. This strategy has been the basis of a number of dual MR/optical agents thought to be potentially translatable to the clinic.69 For instance, dendrimers with multiple Gd–DTPA chelates have been proposed as viable MR agents. Substituting one of the amine bound Gd–chelates on the dendrimer for a fluorophore creates a dual MR/optical imaging agent, with the virtue that the sensitivity differences between MR and optical are compensated by the relative number of attached molecules (Gd-chelates > optical fluorophores by >100 : 1) Additionally, toxicity may be further reduced with the use of nano-sized agents, which are small enough to be excreted by the urinary system and thus, theoretically, have better safety profiles99–102 (Fig. 5).
Fig. 5.
MR and NIR optical sentinel lymph node imaging of breast cancer in a mouse are shown. As depicted in this schema, generation-6 PAMAM dendrimer-based dual labeled contrast agents of approximately 10 nm in diameter containing 172 Gd ions and 2 Cy5.5 fluorophores are employed. Both MRI and NIR optical images define neck (yellow arrow) and axillary (red arrow) lymph nodes as sentinel lymph node of this breast cancer.
Much work is being done in nanomaterial science regarding the development of organic and non-organic nanoparticles for in vivo molecular imaging.6,103 Among the various types of nanomaterials, dendrimers and fluorescent quantum dots have considerable promise due to the wide range of synthetic possibilities that can be achieved with these agents.87,88,101,104,105 The branched structure of dendrimers presents an ideal platform for conjugation and can be useful for overcoming the difference in sensitivities between MR and optical agents, as numerous MR chelates can be attached to a single dendrimer.106–108 Additionally, in vivo behavior of dendrimer-based MR agents is, in part, determined by dendrimer diameter and particles can be optimized for their desired pharmacokinetic parameters, such as their ability to leak from angiogenic vessels, their ability to be excreted either via the kidneys or the liver, or for their ability to be recognized by the RES.109–113 Moreover, the larger size of these molecules improves the magnetic relaxivity by factors of 2–3 resulting in a lower required concentration of gadolinium to achieve the same change in signal.
Quantum dots are also a potential platform for dual MR/optical imaging agents and numerous MR-QD applications have been explored.6,27,104,114–124 Quantum dots can be conjugated to MR active molecules or doped with these agents during synthesis. A standard conjugation approach is to attach amine-reactive chelators such as tetraazacyclododecane-1,4,7,10-tetraacetic acid (DOTA) or diethylenetriaminepentaacetic acid (DTPA) to the amine-rich surface of a QD and then transmetalate with MR-active ions.69 The use of dendrimers or QDs can overcome difference in sensitivity between optical and MR.69 One general limitation to the clinical use of quantum dots is that they contain heavy metals such as cadmium and selenium which in high doses are toxic.125–129 However, toxicity may be reduced with the adoption of size parameters that are specifically designed to permit renal excretion, thus dramatically reducing the exposure time in patients with normal renal function.99,100,102
B.1.2.2 Lanthanide metal MR/optical imaging
A challenge limiting TBR in optical imaging is differentiating target fluorescence from the naturally occurring background fluorescence of native fluorophores (so called autofluorescence). The standard strategy employed to overcome this challenge is to use probes that have emission spectra far from the expected range of typical autofluorescence, which is maximal in the green-red part of the visible spectrum. A relatively new strategy is the use of upconversion luminescence (UCL), a process whereby low-energy light of high wavelength, usually in the near-infrared (NIR) range, is converted to higher-energy (lower wavelength) visible spectrum light through sequential absorption of multiple photons or energy transfers.130,131 This differs from traditional fluorescence where higher energy excitation light is absorbed by the fluorophore but is emitted at lower energy (or higher wavelength resulting in the well known Stokes shift). UCL has been demonstrated in lanthanide metals using excitation wavelengths well above those responsible for autofluorescence.130,132,133 For example, rare-earth nanophosphors exhibit unique UCL properties under continuous wave excitation at 980 nm, an energy at which neither endogenous nor conventional fluorescent probes are excited.130
With this in mind, lanthanide doped nanocrystal complexes have unique properties for bi-modal MR/optical imaging.134–139 These nanocrystals exhibit paramagnetic properties thought to be due to the non-interacting localized nature of the magnetic moment of Gd3+.140 Additionally, unlike traditional gadolinium ion complexes, which relax protons non-specifically, recent reports suggest that lanthanides have potential as “smart” MR imaging contrast agents by demonstrating enhanced relaxation in the presence of specific biological molecules.140 One group demonstrated tumor-specific UCL multicolor imaging in vivo after targeting an integrin receptor.130 The probe was imaged without any background autofluorescence, yielding a high TBR.130 Lanthanide doped nanocrystals are potentially important as bimodal nanoprobes because they are capable of exciting fluorophores deep in the body yet, demonstrate excellent photostability, and minimal autofluorescence.132,134–138,141,142
B.1.3 Nuclear MR multi-modality probes
PET has emerged as an extremely useful tool for whole-body imaging with detection sensitivity below the picomolar range.143–145 A logical next step after PET-CT success146,147 is to combine nuclear imaging, both PET and SPECT with MR imaging.148–151 However, numerous hurdles remain.150,152–155 For instance, conventional PET detectors do not function well in strong magnetic fields required for MR imaging. Early prototypes have been developed using novel solid state PET detectors which are less susceptible to magnetic fields148 and this has prompted investigators to develop PET-MR imaging probes. One group developed a bimodal target-specific PET/MR imaging nanoprobe and demonstrated in vivo efficacy.149 Targeting the tumor/endothelial integrin, alpha-v beta-3, a superparamagnetic iron oxide core was functionalized with DOTA for PET isotope chelation enabling a probe with dual modality imaging capabilities.149 Another bimodal PET/MR imaging agent was developed using MnEIO as the MR imaging agent and 124I for PET.156 Of note, continued progress in the realm of multisignal MR, as discussed below, may reduce the need for bi-modal PET/MR probes.
B.2 Multi-color imaging probes
Multicolor imaging provides rapid discrimination of two different targets, enabling multiparametric imaging. The advantages of multicolor imaging are that it is more efficient than sequentially administering probes and that it allows for merged imaging of two different targets. For these reasons, multicolor imaging is a highly desirable strategy and broadly includes the use of multiple nuclear probes distinguished by energy, the use of optical agents, which use different parts of the optical spectra, and even multiparametric MR imaging.
B.2.1 Optical imaging with multi-color probes
Optical imaging is naturally adapted for multicolor imaging. This strategy is achieved using multicolor optical imaging with numerous different colored probes designed so that each binds a different receptor. Multiple optical probes can be used simultaneously, each targeting a different cell surface receptor.33,157–166 Multicolor strategies can be used to increase the specificity and sensitivity of imaging.158 For example, although cancers often express specific cell-surface receptors, these same receptors are often also expressed (to a lesser degree) on normal tissue. However, by imaging a combination of cell surface receptors, sensitivity and specificity of the target tissue detection may be increased. Tissues that bind multiple probes are then identified by their merged color (e.g. red and green probes binding the same cell may produce a yellow color).158,167 Multicolor optical imaging may also be utilized to image other physiologic processes, such as the tumor microenvironment or lymphatic drainage (see also Fig. 4).46 One goal in advancing the surgical removal of cancer is to better identify microscopic foci of disease in the vasculature and stromal cells that might otherwise be missed. Using multiple color optical probes it may be possible to detect residual tumor to assist in image-guided tissue removal.158,159,167,168 Multiple color optical agents can be combined with multiple multimodality imaging.33,169–172
B.2.2 Nuclear “multi-color” imaging
Using energy resolution features of gamma cameras and different types of gamma emitters, multiple nuclear probes can be detected simultaneously, each targeting a different process. Different photon energies emitted by different radioisotopes can then be used to assign “colors” to a nuclear image. Clinical applications include SPECT for thyroid and parathyroid imaging in which radioactive iodine and thallium are used together173,174 as well as in cardiac scintigraphy where radioactive technetium and thallium may be combined.169–171 For instance, in the assessment of acute myocardial ischemia 201Tl is used to identify areas with patent blood flow and 99mTc-labelled myosin antibody-fragment is used to identify areas of damaged myocardium by detection of exposed myosin fibers.170,171 Although multiplexed nuclear imaging provides useful insights, broader application has been limited by the poor spatial resolution of SPECT and the double dose of ionizing radiation required. Additionally, current strategies for imaging with multiple radiolabelled antibodies require several days from the time of administration for sufficient TBR. Moreover, each type of emitter has a distinct half-life and determining the right combination of isotopes such that the clearance pharmacokinetics and physical half-life of the radionuclides are optimized for a single imaging session is challenging.23 Therefore, nuclear multi-color imaging is used primarily in applications, in which images can be acquired a short time after injection of reagents to minimize the effects due to different half-lives of radioisotopes.
B.2.3 MR “multi-color” imaging
Contrast enhancement in MR imaging is determined by two factors: the proton density of the tissue under examination, and the relaxation properties (e.g. T1 and T2) of protons.175 Conventional MR contrast agents work by shortening the longitudinal and/or transverse relaxation times of nearby protons, mostly within water molecules.175 As a result, MR contrast has several important limitations: (1) pre-contrast images are generally needed to serve as a baseline for the identification of enhanced regions; (2) the measured effect is a function of agent microenvironment (pH, temperature) and agent concentration, which are unknown in vivo; (3) targeted MR imaging agents allow imaging of only one target per exam.175 Recently a new contrast strategy has been developed that does not rely on water protons but instead relies on chemical exchange saturation transfer (CEST).175–180 These contrast agents work by selectively reducing the magnetization of the water signal with the use of CEST agents with exchangeable protons, while minimally affecting longitudinal relaxation rates.175 This technology has been used to achieve multicolor MR imaging in which a series of peptide sequences are distinguished by the resonant frequencies of their exchangeable protons.181 In gliomas, CEST has been used to differentiate between radiation necrosis and malignant tissue by detecting higher than normal concentrations of amide protons of endogenous mobile proteins and peptides that exist in high concentrations in malignant gliomas compared to normal tissue.182 CEST and combination CEST/conventional MR imaging is a powerful new imaging technology with many potential applications.183,184
B.3 Multi-signal imaging
Multiplexed imaging utilizing multiple signal collection relies on capturing two or more distinct signals from the same acquisition. For example, clinical dual-energy CT relies on the different absorption properties of tissue at two separate X-ray energies185 to characterize processes such as iodine–calcium separation (e.g., tumors, arterial plaque), renal-stone composition,186,187 gout,188,189 and perfusion and local blood volume in organs190–192 and tumors.185 Multi-signal imaging can also be achieved with MR and optical imaging technologies.
B.3.1 Multi-signal MR imaging
MR imaging is intrinsically a multi-signal phenomenon. Multiparametric MR imaging, which combines two or more MR imaging techniques is an example of a multi-signal strategy. Given that this can be achieved with most modern MR imaging scanners, without ionizing radiation and the strategy does not require any contrast agent administration, it is in common clinical use. Diffusion-weighted imaging (DWI) relies on the diffusivity of water protons and can provide information about cell density.193 Whole-body DWI provides global functional information and is able to highlight both oncological and non-oncological lesions throughout the entire body in a time efficient manner.193–196 Multi-signal MR imaging is achieved by using whole-body DWI along with traditional MR imaging (T1 and T2 imaging) providing both functional and anatomical data.193 Clinical images acquired in this manner have been reported to compete with PET/CT in their ability to highlight tumor sites.196 Multi-parametric imaging combining DWI with anatomical MR imaging dramatically increased specificity and accuracy of bone metastases detection in patients with lung cancer compared to DWI alone.197 Similarly, the combination of DWI/T2-weighted imaging dramatically improved the sensitivity and specificity of DWI in the detection of abdominal malignancies including prostate cancer.193,198 Further, multi-parametric MR imaging, combining DWI with perfusion weighted imaging is useful for the identification of acute stroke, and its penumbra and has been used in the selection of patients for thrombolytic therapy treatment after stroke.199,200
B.3.2 Multi-signal optical imaging
Multi-signal molecular imaging relies on extracting multiple types of data from a single source. This same principle can be applied to optical imaging. Signal intensity can serve as a measure of local fluorophore concentration and therefore can provide semi-quantitative data regarding the distribution of the probe. Fluorescence lifetime measurements (the time constant describing the loss of fluorescence after initial emission) serve as a separate parameter which is dependent on the microenvironmental factors, such as temperature and pH.201,202 These two distinct parameters can be measured to ascertain probe concentration while simultaneously gaining insight into physical factors pertaining to the tumor microenvironment. (Fig. 6) Clinical applications include tumor-specific optical probes that efficiently localize to the cancer tissue. In such an application, tumor imaging can be achieved by measuring fluorescence intensity, while fluorescence lifetime provides an estimation of tissue oxygenation and degree of hypoxia within the tumor microenvironment.201
Fig. 6.
Fluorescence intensity and lifetime multi-signal images of a HER2-positive (3T3/HER2) and negative (Blab/3T3) tumors’ bearing mouse 2 days after injection with trastuzumab–Alexa680 fluorescent antibody are shown. The HER2-negative tumor still shows high fluorescence intensity due to superior EPR effect. In contrast, the HER2-positive tumor shows elongation of fluorescence lifetime due to specific binding of antibodies to antigens on the cell surface.
C. Target-specific molecular imaging probes: chemistry and biology
Thus far, we have discussed technological approaches to optimizing molecular imaging via multiplexing (combining multiple imaging strategies). Another approach to improving molecular imaging is to optimize the imaging probe to provide maximum target-to-background ratio (TBR). In order to maximize TBR, the imaging probe should accumulate signal at the target tissue/cells while clearing rapidly from the background resulting in minimal signal in non-target organs and tissues. However, this is difficult to achieve as the in vivo pharmacokinetics of many targeted macromolecular moieties, such as monoclonal antibodies, is dictated by the prolonged clearance from the vascular compartment, which leads to high background signal. On the other hand, small molecules, while less specific, are more rapidly cleared, thus improving the target-to-background ratio but reducing the absolute amount of accumulation at the target. Additionally, imaging of organs within the excretory route of the agent (liver, kidney, gastrointestinal tract) is challenging as clearance can obscure pathology in and around these organs. Conventional imaging probes used for CT, MR imaging and radioisotope imaging, are “always on”, and therefore, the signal emanating from the probes reflects their biodistribution but this results in high background signal. One method to reduce background noise is to employ signal activation mechanisms, which turn the probe “on” only at the target tissue.203,204 Signal activation strategies combine biology (target binding), pharmacology (in vivo pharmacokinetic and targeting delivery), and chemistry (signal activation)205 (Fig. 7).
Fig. 7.
A schema of the molecular design for target-specific activatable imaging probes.
C.1 Biodistribution of multi-signal agents
Probe pharmacokinetics and biodistribution play an important role in the TBR of a given probe. In general, imaging probes are delivered to target tissues from the vascular compartment and final tumor signal, depends primarily upon tissue perfusion, time spent in circulation, as well as endothelial permeability at the target; factors collectively referred to as the “input function.” On the other hand, unbound circulating probe and its catabolites produces non-specific background signal and, therefore, prolonged circulatory times decrease TBR. With this in mind, optimizing TBR with conventional “always on” signaling agents poses the dilemma of requiring high blood concentrations to maximize target tissue accumulation but low blood concentrations to minimize background signal. Briefly, macromolecular targeting moieties such as monoclonal antibodies are highly specific and can be delivered in high concentrations to the target. Unfortunately, these large molecules have the undesirable feature of prolonged circulatory retention times of unbound molecules, leading to high background signal. Yet, small molecules are rapidly cleared, which improves the target-to-background ratio, but reduces the absolute amount of accumulation within the target and increases accumulation in the excretion pathways—typically the hepato-biliary system or the urinary tract. One different pharmacokinetic approach to reduce background is to “chase” unbound probe from the circulation with an injection of an agent that binds and sequesters the unbound probe.206–210 However, this approach requires the injection of a second agent. Local activation was developed to address this.35,211–213 New generation “activatable” agents, which have recently been developed and tested in small animals, yield signal only after binding to specific molecules in the target tissue and therefore no signal is produced from the unbound circulating fraction.9,35,214–216 Optimizing TBR differs between conventional and activatable probes as the design of activatable imaging agents focuses on achieving a high input function primarily through high blood probe concentrations, while optimizing the bound versus unbound probe fraction is paramount for conventional agents. The differences in desired pharmacokinetic properties for conventional “always on” versus “activatable” probes, are discussed separately in the following section.
C.1.1 Pharmacokinetics and tissue distribution
Size is a primary determinant of molecular retention time within the vascular and extravascular compartments. Large molecules, such as antibodies, remain in circulation for days to weeks, unless they are recognized and trapped by the reticuloendothelial system in the liver or spleen. Small molecules, such as polysaccharides or amino acids/peptides, are generally cleared from the circulation over the course of minutes. Therefore, large molecules demonstrate high input function but also high background signal from prolonged circulatory retention. In contrast, small molecules demonstrate low input function and low background signal. As a result, for “always on” agents, there is little difference in TBR between large versus small molecular imaging agents. Interestingly, intermediate sized agents demonstrate the highest TBR with acceptable target accumulation for imaging (Fig. 8).217 For “activatable” probes large molecules are clearly desirable because these probes achieve the highest target accumulation but background signal is negligible. However, among macromolecules, larger molecules tend to have poorer penetration into deep tissues, resulting in slow target accumulation.218,219 Molecules with approximate sizes of an IgG immunoglobulin (150 kD; 12 × 9 nm) are optimal for target accumulation within a reasonable amount of time.
Fig. 8.
Schemas explanation of blood clearance and tumor accumulation of antibody and genetically modified antibody fragments of various sizes are shown.
C.1.2 Clearance/excretion
Injected molecules are mostly excreted into the urine or the bile. Kidneys quickly excrete small molecules through two separate processes: (1) glomerular filtration or (2) tubular secretion. Glomerular filtration is the ideal excretion route as it does not involve probe passage through host cells and is therefore felt to be the safest excretory route. Although small molecules such as amino acids and ions might be absorbed and then excreted by tubular secretion, most imaging agents are removed via glomerular filtration. Hard molecules/crystals smaller than 5.5 nm in diameter and slightly larger proteins/polymers with flexible shapes can undergo excretion via glomerular filtration.102,220,221 Both glomerular filtration and tubular excretion are rapid processes, therefore, imaging agents excreted by the kidneys are quickly cleared from the circulation leading to decreased background signal and improved TBR. Again, decreased circulatory retention times compromise the input function and therefore decrease target tissue probe accumulation. However, despite this, renal excretion is almost always considered advantageous for “always on” signaling agents. A disadvantage of renal excretion is that pathology within the genitourinary tract; the kidneys, ureters, and the bladder, may be more difficult to diagnose. In comparison, hepatic excretion is generally a slower process. Most imaging agents, which are not excreted through the kidney, will stay in the circulation for hours, days, and even weeks. Therefore, hepatic excretion is generally advantageous for “activatable” imaging agents as prolonged retention times lead to very high input functions. Of note, the excretory pathway signal is not an issue for “activatable” agents, because they are not activated during excretion.
C.2 Cell binding and activation
In most cases, in vivo imaging aims to visualize specific populations of cells or cellular functions. The outer layer comprised of the lipid bilayer provides a semi-permeable protective envelope around the cell. Most large molecules and hydrophilic small molecules cannot penetrate the cell membrane. However, certain non-permeable molecules can be transported across the lipid bilayer through transmembrane pumps, transporters or receptors. Some small and/or hydrophobic molecules, such as steroids, can directly penetrate the cell membrane and then bind specific receptors in the cytoplasm or in the nucleus. Although this strategy has had relatively limited clinical translation thus far, recently, targeted PET imaging of estrogen receptor positive-tissue in the hormone-therapy responsive breast cancer with radiolabeled estrogen derivatives was reported.222–224 Thus, probes can be designed with specific cell membrane targets in mind for molecular imaging. Further, upon binding, probes may undergo internalization and further enzymatic or lysozomal processing enabling activation of signal production.
C.2.1 Target accumulation
Millions of different molecules, and distinct combinations of these molecules, are expressed on the cell surface. However, few are truly specific for a particular population of cells. In order to achieve target-specific molecular imaging, TBR is an important consideration at the cellular level. To achieve this, target cells should either express unique molecules or overexpress a common molecule by at least 100-fold compared to normal cells. For example, in some cancer cells altered receptors such as the EGF receptors34,225–227 or glycosylation products such as sialyl-Tn,228,229 TAG72230,231 and Louis-Y232,233 antigens can be unique targets. Epidermal growth factor (EGF) receptors, are over expressed in a large proportion of lung, breast, gastrointestinal cancers, and can therefore be used as targets for tumor-specific imaging.227,234–239 In general, molecules such as growth factors or cytokine receptors are expressed during carcinogenesis and may contribute to the cancer cell growth and therefore tend to be reasonable targets for molecular imaging of cancers.
C.2.2 Biological processing and catabolism in the cell
After binding to a specific molecule or entering target cells through a specific channel, biological processing and/or catabolism can help increase probe accumulation, resulting in improved TBR. For example, 18F–fluorodeoxy glucose (18F–FDG) or 18F–fluorothymidine (18F-FLT) undergo phosphorylation by specific kinases (hexokinase and thymidine kinase, respectively) following transport into the cytoplasm and are subsequently trapped within the cell. Furthermore, 18F–FDG-6-phosphate is not hydrolyzed by the glucose-6-phosphatase because it is not recognized as a substrate and accumulates in the cytoplasm unable to go through the glycolytic cascade. Therefore, 18F–FDG accumulates within the cell. Target-specific ligands, which bind to cell surface molecules with high specificity, can provide similar trapping mechanisms for imaging probes. For example, monoclonal IgG-based imaging agents against EGF receptor families can bind to adjacent receptors on the membrane, leading to dimerization and subsequent phosphorylation at the intercellular domain, which then leads to endocytosis of the antibody–receptor complex and internalization into the endolysosome. Within the endolysosome, imaging probes are exposed to harsh chemical environments leading to catabolism, a process which can be utilized for retaining activated signaling as is described later in Section, 3.3.2. Since target-specificity is secured at the time of antigen–antibody binding, any biological processing mechanism that leads to probe retention or signal activation can improve TBR.
C.3 Molecular level
By taking into account pharmacological and biological in vivo processing of imaging probes, agents with superior specificity can be designed. In general, targets are molecules, e.g. antigens, receptors, or substrates, that alter the probe after binding, e.g. by enzymatic catalysis. Therefore, a central principle in the design of molecular imaging agents is that the probes should contain moieties, which bind the target or are recognized by target molecules. Furthermore, incorporating post-binding molecular processing into probe design can result in enhanced retention or signal activation, and can further improve TBR (see Fig. 7).
C.3.1 Target specificity
Targeting with agents such as antibodies or receptor-specific ligands, results in highly specific molecular imaging probes. In contrast, probes that target catabolic processes require additional mechanisms to achieve specificity at sites containing processing molecules. Most enzyme-targeting probes rely on reactions that occur outside of or on the surface of target cells. However, there are several notable exceptions, i.e., 18F–FDG, which binds to hexose-6-monophosphate enzymes in the cytoplasm. Therefore, without a localizing mechanism, processed imaging probes can leak out of target tissue, increasing the background signal and decreasing TBR. Thus, an important design criterion is to assure that probes remain at the desired; i.e., making the enzyme catabolite hydrophobic outside of the cell membrane so that it can readily permeate into the cell membrane of adjacent cells,35 and then, again making the enzyme catabolite hydrophilic inside the target cell so that it cannot leak out from the cell. This is similar to the probe cell capture strategy of 18F–FDG, which undergoes intracellular phosphorylation, effectively trapping the probe in the cell. Utilization of this mechanism results in efficient retention of probes.
C.3.2 Chemical or enzymatic processing after specific binding
After binding to specific molecules on the surface of target cells, large proportions of imaging probes are then internalized into endolysosomes, where specific chemical and biological conditions exist, including low pH (4–5), highly oxidative conditions, and highly reactive acid proteases. An example of enzymatic processing increasing probe retention is the release of radiometals from chelates such as DTPA derivatives in radiopharmaceuticals. Such probes release elemental radioisotopes in acidic conditions. The released radiometals then undergo transchelation by endogenous metal-containing proteins including metallothionein, resulting in prolonged retention in target cells.240,241 Some hydrophobic organic fluorophore-labeled imaging probes are catabolized in the lysosome to release fluorophore–amino acid catabolites. These small molecules can penetrate the lipid bilayers and migrate into intracellular organelles including mitochondria, leading to prolongation of probe retention.242–246 Such retention mechanisms are advantageous for increasing TBR of both “always on” and “activatable” imaging probes.
Intracellular chemical or enzymatic processing plays a significant role in the design of some “activatable” imaging probes which rely on specific activation mechanisms.24,47,247–249 For example, pH-activatable,9 oxidation-activatable, enzyme-activatable,248,250,251 unfolding or catabolism-activatable252 probes have recently been synthesized by employing inter-molecular, inter-fluorophores, or intra-molecular photochemical reactions as shown in the next section.
If specific probes demonstrate minimal internalization after binding to target molecules, “activatable” imaging strategies can be tailored towards pretargeting mechanisms. One strategy is to first inject target-specific binding reagents conjugated with “activating” moieties such as biotin or enzymes, and follow this with a second injection containing “activatable” imaging agents, which are activated upon reacting with the pretargeted moieties through binding214 or enzymatic cleavage.35
C.4 Signal activation via energy transfer and dequenching
In the design of “activatable” optical imaging probes, photon-induced mechanisms, including energy or electron transfer and uncaging, can activate fluorescence. The available “activatable” quenching mechanisms can be separated into two categories; (1) inter-fluorophore quenching mechanisms including Förster resonance energy transfer (FRET) and homo- or hetero-dimer formation between two fluorophores, or (2) intra-fluorophore quenching mechanisms including photon-induced electron transfer and so called “caged” fluorophores (Fig. 9).
Fig. 9.
Schematic explanation of activation mechanisms based on the photo-chemical reactions; a. FRET, b. H-dimer formation, c. photon-induced electron transfer (PeT), d. caged fluorophore.
Non-irradiating FRET for quenching fluorescence signal occurs when two fluorophores are in close proximity thereby exchanging energy between the excited donor and the acceptor within a single molecule or even between two different bound molecules.253,254 In addition, when two fluorophores are close enough to physically behave as a single molecule, some molecules form H- or J-type dimers, which are totally non-fluorescent but emit signal upon dissociation.255 The dissociation constant of H-type dimers was reported to be approximately 10−4 M in solution.256 However, when two molecules are conjugated to a single protein backbone, the H-dimer can be stabilized at a much lower concentration.255,256 In both mechanisms quenching and activation depends upon the distance between the two fluorophores.
Photon-induced electron transfer is based on the energy level difference between the highest occupied molecular orbital (HOMO) and the lowest unoccupied molecular orbital (LUMO) states, which can be rationally modified by attaching or detaching certain side residues on core xanthenes or benzene rings of the fluorophore.257,258 Photon- or enzyme-induced uncaging occurs when chemical changes induced by electron transfer after absorbance of a photon or enzymatic cleavage results in a conformational change releasing the fluorophores from close steric proximity.259 Given that quenching and activation can occur within a single fluorophore, small molecules employing these mechanisms can be incorporated within larger macromolecules with target-specific “activatable” imaging probe complexes.
C.4.1 Inter- and intra-molecular Förster resonance energy transfer
Fluorescent signal quenching based on FRET can be induced when the intermolecular distance equals the inverse of the square of half the distance (1/r2) between the energy donor and acceptor molecules. Thus, energy transfer generally operates within 10 nm. Therefore, this mechanism applies not only to a single macromolecule but also between two different molecules. Protein cleavage by enzymes leading to separation of a single molecule into several pieces is necessary to achieve complete dequenching of “activatable” imaging probes.35,215 However, when large numbers of fluorophores were bound to a single molecule, unfolding and linearization of the protein was all that was required to activate the quenched imaging probe, in one case leading to a 30-fold increase in signal compared to baseline.252 When using two of the same fluorophores to achieve Homo-FRET, baseline quenching states are incomplete and will demonstrate low level fluorescence, however, the probes yield strong signal once they are dequenched. In contrast, fluorophore–quencher pairs, have near complete baseline quenching states, yet, the probes cannot yield as strong signal as Homo-FRET “activatable” probes once completely separated from each other (Fig. 9a).
C.4.2 H-dimer formation
H-dimer formation can also occur when two fluorophores bind sufficiently close to each other on another molecule. The distance between the fluorophores must be within a few angstroms to form an H-dimer and, therefore, H-dimer formation can occur on a single macromolecular imaging probe, such as monoclonal antibodies or receptor ligands.256 Additionally, hydrophobic pockets are especially conducive to H-dimer formation. Once two fluorophores form an H-dimer, fluorescent signal is totally quenched. However, given the often random configuration of fluorophores on probes, not all fluorophores form H-dimers. Therefore, baseline states of “activatable” H-dimer imaging probes are not completely quenched, reducing the difference between the signal intensity in the quenched and activated states.255,256 The proportion of fluorophores within the probe that form H-dimers depends on the chemical structure of the fluorophore and the molecule to which it is conjugated (Fig. 9b).
C.4.3 Photon-induced electron transfer
Photon-induced electron transfer (PeT) is a widely accepted mechanism for “activating” fluorescence quenching, whereby electron transfer from the PeT donor to the excited fluorophore quenches the fluorescence signal. When the PeT donor is cleaved from the fluorophore or inactivated by changing HOMO or LUMO energy status, full activation of fluorescent signal is achieved.260 This switching can operate within a single small fluorophore by utilizing environmental queues including pH (proton density), oxidation by specific reactive oxygen species, specific metals, or enzymative cleavage to cleave the PeT donor.255,261–265 Therefore, activatable fluorophores based on the PeT mechanism can be conjugated with almost any target-specific macromolecular probe including monoclonal antibodies and receptor ligands (Fig. 9c).
C.4.4 Photon- or enzyme-induced uncaging
Conventional caged fluorophores, which have internal photo-breakable bonds after exposure to UV, form chemically unique structures, and are another widely used activating mechanism, most often for fluorescence microscopy. When irradiated with UV light, this additional bond referred to as a “cage” is broken and the probe then emits a fluorescent signal.266–270 Caged fluorophores, which can uncage with irradiation of visible light, have recently been reported and are more applicable to bio-imaging as they do not rely on UV which not only has potential toxicities but also does not penetrate tissue to any degree.. Using a similar mechanism, a spirocyclic cage has recently been reported in the design of activatable fluorophores. The spirocyclic cage is composed of additional bonds formed in the fluorophore that can be broken through enzymatic cleavage of molecules on the side chain of the fluorophore.271,272 Similar to the PeT mechanism, this switching mechanism can operate within a single fluorophore. Therefore, activatable fluorophores based on the PeT mechanism can be conjugated with any target-specific macromolecular probe (Fig. 9d).
C.4.5 Combined approaches using more than one mechanism
Inter-fluorophore quenching, FRET and H-dimer, and intra-fluorophore quenching mechanisms, PeT and caging, operate independently. Therefore, by combining mechanisms, synergistic quenching and activation effects can be achieved. For example, a pH-activatable fluorophore based on PeT can also form H-dimers upon conjugation with target-specific macromolecules, resulting in improved activation ratios compared to the baseline quenched state.255,256
D. Practical preclinical applications of the next generation of molecular imaging probes
Effective molecular imaging detects and characterizes tissue. This requires consideration of the desired imaging objectives (real-time image guided intervention, pharmacokinetic assessment, etc.), a targeting strategy, and a strategy to obtain optimal TBR. To achieve optimization of these endpoints, multiplexed imaging is often warranted. In this section we demonstrate the thought processes involved in the design of a multiplexed imaging strategy. Creation of an optimal probe and ultimately the most effective clinical application requires thoughtful consideration of the numerous and often interconnected aspects of each modality, molecule and application.
D.1 Highly specific cancer detection
One desired medical application for multimodal molecular imaging is to be able to specifically identify tumors with a low false positive rate. For this medical application there are several important considerations (1) finding a highly specific tumor target and (2) optimizing TBR. For this application, monoclonal antibodies were selected as they offer highly specific binding to tumor cell surface molecules. Additionally, numerous humanized antibodies targeting cancer related epitopes have been approved by the FDA for clinical use. The same sub-class of humanized monoclonal antibodies, which are grafted specific binding site, complimentarily determining regions (CDRs), into a human immunoglobulin framework such as IgG1, show >98% homology of protein sequence, therefore, the biodistribution of different humanized antibodies against distinct antigens is similar. However, targeting with humanized antibodies poses an important dilemma for the overall strategy. Due to slow clearance from the vascular compartment, antibody based probes have high background signal and therefore, have a low TBR. With this in mind, a multimodal strategy is warranted. Two ways of approaching the limitations posed by conventional antibody-based imaging using multimodal imaging are (1) multicolor imaging and (2) activatable imaging; either strategy will improve the TBR for the proposed clinical application.
Multicolor optical imaging uses multiple, differently colored optical probes. This improves TBR since the additional, non specific colors serve as an internal control for unbound antibody within the blood pool (Fig. 10a).34,168 Spectral unmixing is used to accurately differentiate multiple probes and this facilitates highly specific cancer imaging without waiting for clearance of unbound antibody.168 Additionally, multi-excitation spectral fluorescence imaging allows greater flexibility in color selection and permits the simultaneous use of numerous targeting agents. TBR is ultimately increased as the number of independent signals increase, yielding more specific data. The use of multiple colors can depict single lesions with higher TBR and can also detect molecularly heterogeneous lesions, as may occur in naturally occurring tumors over time (Fig. 10b). The combination of multi-excitation and unmixing techniques employing multiple, humanized antibodies allows the performance of multi-target imaging with relatively high TBR.
Fig. 10.
Two practical examples for cancer-cell specific imaging using combinations of proposed technologies; multi-color imaging (a, 2-color; b, 3-color), and reversible pH-activatable imaging using a target-cell activatable imaging probe. (a and b) Multi-color imaging targeting HER1, HER2, and CD25 using Cy5-, Cy7-, and Alexa700-labeled respective specific antibodies; cetuximab (anti-HER1), trastuzumab (anti-HER2), and daclizumab (anti-CD25) clearly shows different specific target molecules, which were expressed by distinct cancers, in different colors shown in pink, yellow, and blue, respectively. (c) A reversible activatable imaging probe of pH-sensitive trastuzumab–dimethyl-phenyl-BODIPY conjugate cannot only depict receptor-positive tiny target tumor metastasis (green) but also monitor the therapeutic effect of anti-cancer therapy in real time.
The other approach for target specific cancer imaging, which overcomes limitations of low TBR of conventional antibody based imaging is the use of “activatable” probes. As discussed above, these probes are activated by cancer-specific factors such as enzymatic reactions or cancer-cell mediated processing initiated by binding specific molecules. In the implementation and design of these probes, special consideration is given to the design of signaling molecules that harness unique aspects of the cancer milieu for probe activation. For instance, we utilized a cancer-cell specific targeting probe to design an activatable fluorescent probe.9 Our probe emitted fluorescence only after target binding and subsequent cellular internalization. We designed the probe such that the low pH in the lysosome resulted in the addition of a proton to the fluorophore, which inhibited electron transfer and resulted in dequenching or activation of the fluorescence signal. This strategy uses an activatable approach to overcome the low TBR normally expected with labeled antibodies (Fig. 10c). Furthermore, the design strategy can be further refined to achieve signal activation that is reversible. Relying on the premise that damaged or dead cells cannot effectively acidify the lysosome because the ATP-consuming proton pumps are not functioning, activated fluorescent signal disappears upon cancer cell death thus enabling real-time monitoring of cellular viability (Fig. 10c).
D.2 Molecular binding specific imaging
An ideal application for molecular imaging is drug development.273,274 Molecular imaging of the drug provides critical information about agent pharmacokinetics and multi-plexed imaging can provide the added benefit of real-time imaging of probe binding. Thus far, imaging of drug–target binding has not been feasible with conventional strategies, but the implementation of a thoughtful multimodal approach may make this possible in the near future. In order to achieve real-time binding we must know: the absolute amount of probe in the local area and the component of the signal that arises from the bound fraction. Two different approaches are proposed. 1. Bi-modal probes with pharmacokinetic imaging capabilities using “always on” labeling and probe-binding imaging capabilities using “activatable” labeling. 2. Dual-signaling for probe density and binding activation from a single labeled probe.
The creation of an effective bi-modal probe with pharmacokinetic imaging capabilities using “always on” labeling and imaging using “activatable” labeling requires a multifaceted design strategy. If we select cancer-specific humanized monoclonal antibodies for target tissue binding (due to high binding affinities) and utilize an imaging modality that is easy to quantitate this method could be used to monitor a drug. To achieve this, the radiometal (111In) can be conjugated to the targeting antibody via a chelate. To achieve the other endpoint of detecting the binding of the probe, an “activatable” ICG is an ideal choice as it has the unique property of losing fluorescence when it is covalently bound to an antibody. However, the signal is then activated by cellular processing after target-specific binding and internalization as described above. This quality is useful in designing an antibody-based activatable probe which is irreversible.275 Therefore, in vivo multimodality imaging can be used to achieve target-specific imaging with optical probes while also investigating the probe pharmacokinetics and biodistribution with nuclear imaging (Fig. 11a). This bi-modal strategy provides data regarding both the absolute local amount of probe and the bound fraction.276
Fig. 11.
Two practical examples for visualizing target-binding of imaging probes using combinations of proposed technologies; dual-modality imaging using an always on 111In and activatable ICG optical dual-labeling trastuzumab antibody (a), and Dual-signaling tumor imaging of trastuzumab–Alexa750 conjugate employing fluorescence intensity and lifetime technologies (b). Both methods simultaneously show distribution of imaging probes with always on gamma-ray (111In; a) or Alexa750 signal (b) as well as binding specific-image with activatable ICG (a) or elongated fluorescence lifetime of Alexa750 (b).
The other multiplexed approach for imaging probe concentration and molecular specific binding is multi-signaling fluorescence. This strategy relies on targeting with antibodies but utilizes two different aspects of fluorescence signal—intensity and lifetime—to derive imaging information. For instance, when AlexaFluor750 is conjugated to an anti-HER2 antibody, it will yield signal related to the fluorophore concentration. Independently, fluorescence lifetime measurements will depend on microenvironmental conditions of the fluorophore: such as solvent, temperature, pH, and oxygen content. Bearing in mind that target-specific binding and subsequent cellular processing change the microenvironment of the fluorophore, it is not unexpected that there are changes in the fluorescence lifetime signal; thus, this parameter can be used to determine real-time probe binding. Using an AlexaFluor750–anti-HER2 antibody conjugate, fluorescence lifetime measurements could distinguish the bound and unbound fraction in vitro.277 Since fluorescence lifetime is unrelated to fluorophore concentration and light scattering, this dual-signal detection imaging technique provides information regarding probe concentration and cell specific binding (Fig. 11b).
E. Conclusion
Conventional imaging methods have revolutionized medicine, yet the next advance will involve overcoming inherent limitations of individual modalities, such as poor spatial resolution or low sensitivity. Multiplexed imaging aims to advance imaging by strategically combining existing technologies with uniquely designed molecular probes. The ways in which imaging can be multiplexed are plentiful. Additionally, there are many aspects and approaches to designing an optimal multimodal imaging probe. Achieving effective molecular imaging, in which tissue is both detected and molecularly characterized, requires consideration of the desired objectives (real-time image guided intervention, pharmacokinetic assessment, etc.) as well as a strategy for achieving effective targeting and optimal TBR. To optimally achieve these endpoints, a multi-disciplinary approach that takes into account physics and engineering as well as target-specific delivery and signal production mechanisms that integrate pharmacology, biology, and chemistry is needed. Multiplexed imaging aims to provide greater sensitivity and specificity for earlier and more accurate disease detection. We hope that this review has provided useful insights into achieving these endpoints.
Acknowledgements
This research was supported by the Intramural Research Program of the National Institutes of Health, National Cancer Institute, Center for Cancer Research.
Abbreviations
- MR
Magnetic Resonance
- TBR
Tumor to Background Ratio
- PET
Positron Emission Tomography
- CT
Computerized Tomography
- NIR
Near InfraRed
- CCD
Charge Coupled Device
- QD
Quantum Dot
- VEGF
Vascular Endothelial Growth Factor
- IO
Iron Oxide
- RES
ReticuloEndothelial System
- EPR
Enhanced Permeation and Retention
- CLI
Cerenkov Luminescence Imaging
- CR
Cerenkov Radiation
- NP
NanoParticles
- UCL
UpConversion Luminescence
- SPECT
Single-Photon Emission Computerized Tomography
- DOTA
1,4,7,10-tetraazacycloDOdecane-1,4,7,10-Tetraacetic Acid
- DTPA
DiethyleneTriaminePentaacetic Acid
- CEST
Chemical Exchange Saturation Transfer
- DWI
Diffusion-Weighted Imaging
- FDG
FluoroDeoxyGlucose
- FLT
FLuoroThymidine
- FRET
Förster Resonance Energy Transfer
- EGF
Epidermal Growth Factor
- HOMO
Highest Occupied Molecular Orbital
- LUMO
Lowest Occupied Molecular Orbital
- PeT
Photon-induced electron Transfer
- CDR
Complimentarily Determining Regions
- ICG
IndoCyanine Green
- HER2
Human Epidermal growth factor Receptor type-2
Biographies
Hisataka Kobayashi
Dr Hisataka Kobayashi is the Chief scientist in the Molecular Imaging Program at the National Cancer Institute of the National Institutes of Health in Bethesda, MD. Dr Kobayashi is awarded MD and PhD (Immunology/Medicine) from the Kyoto University in Japan. He moved to the current position in the Molecular Imaging Program at NCI in 2004. His interest is in developing the novel molecular imaging agents and technologies especially for targeting cancers. Dr Kobayashi has published over 200 peer reviewed articles and 30 invited reviews and book chapters covering various imaging modalities from basic chemistry, preclinical development, to the clinical studies.
Michelle R. Longmire
Dr Michelle R Longmire was a medical student in the Molecular Imaging Program, NCI/NIH funded by the Howard Hugh Medical Institute from 2008 to 2010. She is a graduate of the University of New Mexico, School of Medicine. Her interest is in the development of novel molecular imaging probes and nanomedicines that can identify and treat cancers in the earliest phases of disease. She plans to focus on imaging and radiologic sciences during her career.
Mikako Ogawa
Dr Mikako Ogawa was a postdoctoral fellow in the Molecular Imaging Program at the National Cancer Institute in Bethesda, MD by May 2009. Now Dr Ogawa is an associate professor in the Photon Medical Research Center at the Hamamatsu Medical University. Dr Ogawa is awarded PhD (Pharmaceutical Sciences) from the Kyoto University in Japan. She joined to the Molecular Imaging Program at NCI in 2007. Her interest is in developing the novel molecular imaging agents.
Peter L. Choyke
Dr Peter Choyke is the Chief of the Molecular Imaging Program at the Center for Cancer Research of the National Cancer Institute in Bethesda, Maryland. Dr Choyke is a graduate of Jefferson Medical School and received training in Diagnostic Radiology at Yale University and the University of Pennsylvania. He joined the Diagnostic Radiology Department at the Clinical Center of the National Institutes of Health in 1988 and formed the Molecular Imaging Program at NCI in 2004. His interest is in accelerating the treatment of cancer by using novel molecular imaging agents which target specific features of cancers.
Note and references
- 1.Runge VM. Advances in Diagnostic Radiology. Invest. Radiol. 2010 Nov 5; doi: 10.1097/RLI.0b013e31820244fa. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- 2.Osborn EA, Jaffer FA. The year in molecular imaging. JACC: Cardiovasc. Imaging. 2010;3:1181–1195. doi: 10.1016/j.jcmg.2010.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Brower V. Beyond FDG: Many Molecular Imaging Agents Are in Development. J. Natl. Cancer Inst. 2011;103:13–15. doi: 10.1093/jnci/djq537. [DOI] [PubMed] [Google Scholar]
- 4.Massoud TF, Gambhir SS. Molecular imaging in living subjects: seeing fundamental biological processes in a new light. Genes Dev. 2003;17:545–580. doi: 10.1101/gad.1047403. [DOI] [PubMed] [Google Scholar]
- 5.Weissleder R. Molecular imaging in cancer. Science. 2006;312:1168–1171. doi: 10.1126/science.1125949. [DOI] [PubMed] [Google Scholar]
- 6.Rosenblum LT, Kosaka N, Mitsunaga M, Choyke PL, Kobayashi H. In vivo molecular imaging using nanomaterials: general in vivo characteristics of nano-sized reagents and applications for cancer diagnosis. Mol. Membr. Biol. 2010;27:274–285. doi: 10.3109/09687688.2010.481640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Zhang L, Lee KC, Bhojani MS, Khan AP, Shilman A, Holland EC, Ross BD, Rehemtulla A. Molecular imaging of Akt kinase activity. Nat. Med. (N. Y.) 2007;13:1114–1119. doi: 10.1038/nm1608. [DOI] [PubMed] [Google Scholar]
- 8.Seaman ME, Contino G, Bardeesy N, Kelly KA. Molecular imaging agents: impact on diagnosis and therapeutics in oncology. Expert Rev. Mol. Med. 2010;12:e20. doi: 10.1017/S1462399410001511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Urano Y, Asanuma D, Hama Y, Koyama Y, Barrett T, Kamiya M, Nagano T, Watanabe T, Hasegawa A, Choyke PL, Kobayashi H. Selective molecular imaging of viable cancer cells with pH-activatable fluorescence probes. Nat. Med. 2009;15:104–109. doi: 10.1038/nm.1854. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Basu S, Alavi A. Molecular imaging (PET) of brain tumors. Neuroimaging Clin. North Am. 2009;19:625–646. doi: 10.1016/j.nic.2009.08.012. [DOI] [PubMed] [Google Scholar]
- 11.Weissleder R, Pittet MJ. Imaging in the era of molecular oncology. Nature. 2008;452:580–589. doi: 10.1038/nature06917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Strauss HW, Grewal RK, Pandit-Taskar N. Molecular imaging in nuclear cardiology. Semin. Nucl. Med. 2004;34:47–55. doi: 10.1053/j.semnuclmed.2003.09.006. [DOI] [PubMed] [Google Scholar]
- 13.Schindler TH, Schelbert HR, Quercioli A, Dilsizian V. Cardiac PET imaging for the detection and monitoring of coronary artery disease and microvascular health. JACC: Cardiovasc. Imaging. 2010;3:623–640. doi: 10.1016/j.jcmg.2010.04.007. [DOI] [PubMed] [Google Scholar]
- 14.Al-Mallah MH, Sitek A, Moore SC, Di Carli M, Dorbala S. Assessment of myocardial perfusion and function with PET and PET/CT. J. Nucl. Cardiol. 2010;17:498–513. doi: 10.1007/s12350-010-9223-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Ohshima S, Petrov A, Fujimoto S, Zhou J, Azure M, Edwards DS, Murohara T, Narula N, Tsimikas S, Narula J. Molecular imaging of matrix metalloproteinase expression in atherosclerotic plaques of mice deficient in apolipoprotein e or low-density-lipoprotein receptor. J. Nucl. Med. 2009;50:612–617. doi: 10.2967/jnumed.108.055889. [DOI] [PubMed] [Google Scholar]
- 16.Jaffer FA, Nahrendorf M, Sosnovik D, Kelly KA, Aikawa E, Weissleder R. Cellular imaging of inflammation in atherosclerosis using magnetofluorescent nanomaterials. Mol. Imaging. 2006;5:85–92. [PubMed] [Google Scholar]
- 17.Erritzoe D, Holst K, Frokjaer VG, Licht CL, Kalbitzer J, Nielsen FA, Svarer C, Madsen J, Knudsen G. A nonlinear relationship between cerebral serotonin transporter and 5-HT(2A) receptor binding: an in vivo molecular imaging study in humans. J. Neurosci. 2010;30:3391–3397. doi: 10.1523/JNEUROSCI.2852-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Song PJ, Barc C, Arlicot N, Guilloteau D, Bernard S, Sarradin P, Chalon S, Garreau L, Kung HF, Lantier F, Vergote J. Evaluation of prion deposits and microglial activation in scrapie-infected mice using molecular imaging probes. Mol. Imaging Biol. 2010;12:576–582. doi: 10.1007/s11307-010-0321-1. [DOI] [PubMed] [Google Scholar]
- 19.Peterson EC, Kim LJ. Magnetic resonance imaging at the molecular level. World Neurosurg. 2010;73:604–605. doi: 10.1016/j.wneu.2010.06.044. [DOI] [PubMed] [Google Scholar]
- 20.Sandu N, Popperl G, Toubert ME, Spiriev T, Arasho B, Orabi M, Schaller BJ. Current Molecular Imaging of Spinal Tumors in Clinical Practice. Mol. Med. 2011;17:308–316. doi: 10.2119/molmed.2010.00218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Taghva A, Khalessi AA, Kim PE, Liu CY, Apuzzo ML. From atom to brain: applications of molecular imaging to neurosurgery. World Neurosurg. 2010;73:477–485. doi: 10.1016/j.wneu.2010.02.058. [DOI] [PubMed] [Google Scholar]
- 22.VanBrocklin H, Herscovitch P. Molecular imaging update: advances in MI of the brain. J. Nucl. Med. 2007;48:24N–29N. [PubMed] [Google Scholar]
- 23.Kobayashi H, Longmire MR, Ogawa M, Choyke PL, Kawamoto S. Multiplexed imaging in cancer diagnosis: applications and future advances. Lancet Oncol. 2010;11:589–595. doi: 10.1016/S1470-2045(10)70009-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Lee S, Xie J, Chen X. Activatable molecular probes for cancer imaging. Curr. Top. Med. Chem. 2010;10:1135–1144. doi: 10.2174/156802610791384270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kuil J, Velders AH, van Leeuwen FW. Multimodal tumor-targeting peptides functionalized with both a radio- and a fluorescent label. Bioconjugate Chem. 2010;21:1709–1719. doi: 10.1021/bc100276j. [DOI] [PubMed] [Google Scholar]
- 26.Zeng W, Miao W. Development of small molecular probes for the molecular imaging of apoptosis. Anticancer Agents Med. Chem. 2009;9:986–995. doi: 10.2174/187152009789377763. [DOI] [PubMed] [Google Scholar]
- 27.Jennings LE, Long NJ. ‘Two is better than one’—probes for dual-modality molecular imaging. Chem. Commun. 2009:3511–3524. doi: 10.1039/b821903f. [DOI] [PubMed] [Google Scholar]
- 28.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;5:3745–3757. doi: 10.1039/b711244k. [DOI] [PubMed] [Google Scholar]
- 29.Anderson CJ, Bulte JW, Chen K, Chen X, Khaw BA, Shokeen M, Wooley KL, VanBrocklin HF. Design of targeted cardiovascular molecular imaging probes. J. Nucl. Med. 2010;51(suppl 1):3S–17S. doi: 10.2967/jnumed.109.068130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Chen K, Chen X. Design and development of molecular imaging probes. Curr. Top. Med. Chem. 2010;10:1227–1236. doi: 10.2174/156802610791384225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Sogaard R, Fischer BM, Mortensen J, Hojgaard L, Lassen U. Preoperative staging of lung cancer with PET/CT: cost-effectiveness evaluation alongside a randomized controlled trial. Eur. J. Nucl. Med. Mol. Imaging. 2011;38:802–809. doi: 10.1007/s00259-010-1703-y. [DOI] [PubMed] [Google Scholar]
- 32.von Schulthess GK, Burger C. Integrating imaging modalities: what makes sense from a workflow perspective? Eur. J. Nucl. Med. Mol. Imaging. 2010;37:980–990. doi: 10.1007/s00259-009-1378-4. [DOI] [PubMed] [Google Scholar]
- 33.Kobayashi H, Ogawa M, Kosaka N, Choyke PL, Urano Y. Multicolor imaging of lymphatic function with two nanomaterials: quantum dot-labeled cancer cells and dendrimer-based optical agents. Nanomedicine (London, U. K.) 2009;4:411–419. doi: 10.2217/nnm.09.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Barrett T, Koyama Y, Hama Y, Ravizzini G, Shin IS, Jang BS, Paik CH, Urano Y, Choyke PL, Kobayashi H. In vivo diagnosis of epidermal growth factor receptor expression using molecular imaging with a cocktail of optically labeled monoclonal antibodies. Clin. Cancer Res. 2007;13:6639–6648. doi: 10.1158/1078-0432.CCR-07-1119. [DOI] [PubMed] [Google Scholar]
- 35.Kamiya M, Kobayashi H, Hama Y, Koyama Y, Bernardo M, Nagano T, Choyke PL, Urano Y. An enzymatically activated fluorescence probe for targeted tumor imaging. J. Am. Chem. Soc. 2007;129:3918–3929. doi: 10.1021/ja067710a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hama Y, Urano Y, Koyama Y, Bernardo M, Choyke PL, Kobayashi H. A comparison of the emission efficiency of four common green fluorescence dyes after internalization into cancer cells. Bioconjugate Chem. 2006;17:1426–1431. doi: 10.1021/bc0601626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Lee H, Akers WJ, Cheney PP, Edwards WB, Liang K, Culver JP, Achilefu S. Complementary optical and nuclear imaging of caspase-3 activity using combined activatable and radio-labeled multimodality molecular probe. J. Biomed. Opt. 2009;14 doi: 10.1117/1.3207156. 040507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Wang W, Ke S, Kwon S, Yallampalli S, Cameron AG, Adams KE, Mawad ME, Sevick-Muraca EM. A new optical and nuclear dual-labeled imaging agent targeting interleukin 11 receptor alpha-chain. Bioconjugate Chem. 2007;18:397–402. doi: 10.1021/bc0602679. [DOI] [PubMed] [Google Scholar]
- 39.Li C, Wang W, Wu Q, Ke S, Houston J, Sevick-Muraca E, Dong L, Chow D, Charnsangavej C, Gelovani JG. Dual optical and nuclear imaging in human melanoma xenografts using a single targeted imaging probe. Nucl. Med. Biol. 2006;33:349–358. doi: 10.1016/j.nucmedbio.2006.01.001. [DOI] [PubMed] [Google Scholar]
- 40.Blasberg RG. In vivo molecular-genetic imaging: multi-modality nuclear and optical combinations. Nucl. Med. Biol. 2003;30:879–888. doi: 10.1016/s0969-8051(03)00115-x. [DOI] [PubMed] [Google Scholar]
- 41.Culver J, Akers W, Achilefu S. Multimodality molecular imaging with combined optical and SPECT/PET modalities. J. Nucl. Med. 2008;49:169–172. doi: 10.2967/jnumed.107.043331. [DOI] [PubMed] [Google Scholar]
- 42.Thorp-Greenwood FL, Coogan MP. Multimodal radio- (PET/SPECT) and fluorescence imaging agents based on metallo-radioisotopes: current applications and prospects for development of new agents. Dalton Trans. 2011 doi: 10.1039/c0dt01398f. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- 43.Merkel OM, Librizzi D, Pfestroff A, Schurrat T, Behe M, Kissel T. In vivo SPECT and real-time gamma camera imaging of biodistribution and pharmacokinetics of siRNA delivery using an optimized radiolabeling and purification procedure. Bioconjugate Chem. 2009;20:174–182. doi: 10.1021/bc800408g. [DOI] [PubMed] [Google Scholar]
- 44.Gainkam LO, Huang L, Caveliers V, Keyaerts M, Hernot S, Vaneycken I, Vanhove C, Revets H, De Baetselier P, Lahoutte T. Comparison of the biodistribution and tumor targeting of two 99mTc-labeled anti-EGFR nanobodies in mice, using pinhole SPECT/micro-CT. J. Nucl. Med. 2008;49:788–795. doi: 10.2967/jnumed.107.048538. [DOI] [PubMed] [Google Scholar]
- 45.Ho CL, Chen LC, Lee WC, Chiu SP, Hsu WC, Wu YH, Yeh CH, Stabin MG, Jan ML, Lin WJ, Lee TW, Chang CH. Receptor-binding, biodistribution, dosimetry, and micro-SPECT/CT imaging of 111In-[DTPA(1), Lys(3), TyR(4)]-bombesin analog in human prostate tumorbearing mice. Cancer Biother. Radiopharm. 2009;24:435–443. doi: 10.1089/cbr.2008.0616. [DOI] [PubMed] [Google Scholar]
- 46.Kobayashi H, Koyama Y, Barrett T, Hama Y, Regino CA, Shin IS, Jang BS, Le N, Paik CH, Choyke PL, Urano Y. Multimodal nanoprobes for radionuclide and five-color near-infrared optical lymphatic imaging. ACS Nano. 2007;1:258–264. doi: 10.1021/nn700062z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Yang Y, Hong H, Zhang Y, Cai W. Molecular Imaging of Proteases in Cancer. Cancer Growth Metastasis. 2009;2:13–27. doi: 10.4137/cgm.s2814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Sampath L, Kwon S, Ke S, Wang W, Schiff R, Mawad ME, Sevick-Muraca EM. Dual-labeled trastuzumab-based imaging agent for the detection of human epidermal growth factor receptor 2 overexpression in breast cancer. J. Nucl. Med. 2007;48:1501–1510. doi: 10.2967/jnumed.107.042234. [DOI] [PubMed] [Google Scholar]
- 49.Nahrendorf M, Keliher E, Marinelli B, Waterman P, Feruglio PF, Fexon L, Pivovarov M, Swirski FK, Pittet MJ, Vinegoni C, Weissleder R. Hybrid PET-optical imaging using targeted probes. Proc. Natl. Acad. Sci. U. S. A. 2010;107:7910–7915. doi: 10.1073/pnas.0915163107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Chen K, Li ZB, Wang H, Cai W, Chen X. Dual-modality optical and positron emission tomography imaging of vascular endothelial growth factor receptor on tumor vasculature using quantum dots. Eur. J. Nucl. Med. Mol. Imaging. 2008;35:2235–2244. doi: 10.1007/s00259-008-0860-8. [DOI] [PubMed] [Google Scholar]
- 51.Gambhir SS. Molecular imaging of cancer with positron emission tomography. Nat. Rev. Cancer. 2002;2:683–693. doi: 10.1038/nrc882. [DOI] [PubMed] [Google Scholar]
- 52.Sampath L, Kwon S, Hall MA, Price RE, Sevick-Muraca EM. Detection of Cancer Metastases with a Dual-labeled Near-Infrared/Positron Emission Tomography Imaging Agent. Transl. Oncol. 2010;3:307–217. doi: 10.1593/tlo.10139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Robertson R, Germanos MS, Li C, Mitchell GS, Cherry SR, Silva MD. Optical imaging of Cerenkov light generation from positron-emitting radiotracers. Phys. Med. Biol. 2009;54:N355–N365. doi: 10.1088/0031-9155/54/16/N01. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lewis MA, Kodibagkar VD, Öz OK, Mason RP. On the potential for molecular imaging with Cerenkov luminescence. Opt. Lett. 2010;35:3889–3891. doi: 10.1364/OL.35.003889. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Li C, Mitchell GS, Cherry SR. Cerenkov luminescence tomography for small-animal imaging. Opt. Lett. 2010;35:1109–1111. doi: 10.1364/OL.35.001109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Holland JP, Normand G, Ruggiero A, Lewis JS, Grimm J. Intraoperative imaging of positron emission tomographic radiotracers using Cerenkov luminescence emissions. Mol. Imaging. 2011;10:177–186. [PMC free article] [PubMed] [Google Scholar]
- 57.Ruggiero A, Holland JP, Lewis JS, Grimm J. Cerenkov luminescence imaging of medical isotopes. J. Nucl. Med. 2010;51:1123–1130. doi: 10.2967/jnumed.110.076521. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Liu H, Ren G, Miao Z, Zhang X, Tang X, Han P, Gambhir SS, Cheng Z. Molecular optical imaging with radioactive probes. PLoS One. 2010;5:e9470. doi: 10.1371/journal.pone.0009470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Lucignani G. Cerenkov radioactive optical imaging: a promising new strategy. Eur. J. Nucl. Med. Mol. Imaging. 2011;38:592–595. doi: 10.1007/s00259-010-1708-6. [DOI] [PubMed] [Google Scholar]
- 60.Spinelli AE, D’Ambrosio D, Calderan L, Marengo M, Sbarbati A, Boschi F. Cerenkov radiation allows in vivo optical imaging of positron emitting radiotracers. Phys. Med. Biol. 2010;55:483–495. doi: 10.1088/0031-9155/55/2/010. [DOI] [PubMed] [Google Scholar]
- 61.Dothager RS, Goiffon RJ, Jackson E, Harpstrite S, Piwnica-Worms D. Cerenkov radiation energy transfer (CRET) imaging: a novel method for optical imaging of PET isotopes in biological systems. PLoS One. 2010;5:e13300. doi: 10.1371/journal.pone.0013300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Penet MF, Mikhaylova M, Li C, Krishnamachary B, Glunde K, Pathak AP, Bhujwalla ZM. Applications of molecular MRI and optical imaging in cancer. Future Med. Chem. 2010;2:975–988. doi: 10.4155/fmc.10.25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Koyama Y, Talanov VS, Bernardo M, Hama Y, Regino CA, Brechbiel MW, Choyke PL, Kobayashi H. A dendrimer-based nanosized contrast agent dual-labeled for magnetic resonance and optical fluorescence imaging to localize the sentinel lymph node in mice. J. Magn. Reson. Imaging. 2007;25:866–871. doi: 10.1002/jmri.20852. [DOI] [PubMed] [Google Scholar]
- 64.Kumar R, Ohulchanskyy TY, Turowski SG, Thompson ME, Seshadri M, Prasad PN. Combined magnetic resonance and optical imaging of head and neck tumor xenografts using Gadolinium-labelled phosphorescent polymeric nanomicelles. Head Neck Oncol. 2010;2:35. doi: 10.1186/1758-3284-2-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sung CK, Hong KA, Lin S, Lee Y, Cha J, Lee JK, Hong CP, Han BS, Jung SI, Kim SH, Yoon KS. Dual-modal nanoprobes for imaging of mesenchymal stem cell transplant by MRI and fluorescence imaging. Korean J. Radiol. 2009;10:613–622. doi: 10.3348/kjr.2009.10.6.613. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lisy MR, Hartung A, Lang C, Schuler D, Richter W, Reichenbach JR, Kaiser WA, Hilger I. Fluorescent bacterial magnetic nanoparticles as bimodal contrast agents. Invest. Radiol. 2007;42:235–241. doi: 10.1097/01.rli.0000255832.44443.e7. [DOI] [PubMed] [Google Scholar]
- 67.Mishra A, Pfeuffer J, Mishra R, Engelmann J, Mishra AK, Ugurbil K, Logothetis NK. A new class of Gd-based DO3A-ethylamine-derived targeted contrast agents for MR and optical imaging. Bioconjugate Chem. 2006;17:773–780. doi: 10.1021/bc050295b. [DOI] [PubMed] [Google Scholar]
- 68.Pan D, Caruthers SD, Chen J, Winter PM, Senpan A, Schmieder AH, Wickline SA, Lanza GM. Nanomedicine strategies for molecular targets with MRI and optical imaging. Future Med. Chem. 2010;2:471–490. doi: 10.4155/fmc.10.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Louie A. Multimodality imaging probes: design and challenges. Chem. Rev. 2010;110:3146–3195. doi: 10.1021/cr9003538. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Soenen SJ, De Cuyper M. Assessing iron oxide nanoparticle toxicity in vitro: current status and future prospects. Nanomedicine (London, U. K.) 2010;5:1261–1275. doi: 10.2217/nnm.10.106. [DOI] [PubMed] [Google Scholar]
- 71.Jain TK, Reddy MK, Morales MA, Leslie-Pelecky DL, Labhasetwar V. Biodistribution, clearance, and biocompatibility of iron oxide magnetic nanoparticles in rats. Mol. Pharmacol. 2008;5:316–327. doi: 10.1021/mp7001285. [DOI] [PubMed] [Google Scholar]
- 72.Mandeville JB, Moore J, Chesler DA, Garrido L, Weissleder R, Weisskoff RM. Dynamic liver imaging with iron oxide agents: effects of size and biodistribution on contrast. Magn. Reson. Med. 1997;37:885–890. doi: 10.1002/mrm.1910370613. [DOI] [PubMed] [Google Scholar]
- 73.Thorek DL, Tsourkas A. Size, charge and concentration dependent uptake of iron oxide particles by non-phagocytic cells. Biomaterials. 2008;29:3583–3590. doi: 10.1016/j.biomaterials.2008.05.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Huang J, Bu L, Xie J, Chen K, Cheng Z, Li X, Chen X. Effects of nanoparticle size on cellular uptake and liver MRI with polyvinylpyrrolidone-coated iron oxide nanoparticles. ACS Nano. 2010;4:7151–7160. doi: 10.1021/nn101643u. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Guardia P, Perez N, Labarta A, Batlle X. Controlled synthesis of iron oxide nanoparticles over a wide size range. Langmuir. 2010;26:5843–5847. doi: 10.1021/la903767e. [DOI] [PubMed] [Google Scholar]
- 76.Stjerndahl M, Andersson M, Hall HE, Pajerowski DM, Meisel MW, Duran RS. Superparamagnetic Fe3O4SiO2 nanocomposites: enabling the tuning of both the iron oxide load and the size of the nanoparticles. Langmuir. 2008;24:3532–3536. doi: 10.1021/la7035604. [DOI] [PubMed] [Google Scholar]
- 77.Jarrett BR, Frendo M, Vogan J, Louie AY. Size-controlled synthesis of dextran sulfate coated iron oxide nanoparticles for magnetic resonance imaging. Nanotechnology. 2007;18 doi: 10.1088/0957-4484/18/3/035603. 035603. [DOI] [PubMed] [Google Scholar]
- 78.Larsen EK, Nielsen T, Wittenborn T, Birkedal H, Vorup-Jensen T, Jakobsen MH, Ostergaard L, Horsman MR, Besenbacher F, Howard KA, Kjems J. Size-Dependent Accumulation of PEGylated Silane-Coated Magnetic Iron Oxide Nanoparticles in Murine Tumors. ACS Nano. 2009;3:1947–1951. doi: 10.1021/nn900330m. [DOI] [PubMed] [Google Scholar]
- 79.Briley-Saebo KC, Johansson LO, Hustvedt SO, Haldorsen AG, Bjornerud A, Fayad ZA, Ahlstrom HK. Clearance of iron oxide particles in rat liver: effect of hydrated particle size and coating material on liver metabolism. Invest. Radiol. 2006;41:560–571. doi: 10.1097/01.rli.0000221321.90261.09. [DOI] [PubMed] [Google Scholar]
- 80.Veiseh O, Sun C, Gunn J, Kohler N, Gabikian P, Lee D, Bhattarai N, Ellenbogen R, Sze R, Hallahan A, Olson J, Zhang M. Optical and MRI multifunctional nanoprobe for targeting gliomas. Nano Lett. 2005;5:1003–1008. doi: 10.1021/nl0502569. [DOI] [PubMed] [Google Scholar]
- 81.Xu H, Regino CA, Koyama Y, Hama Y, Gunn AJ, Bernardo M, Kobayashi H, Choyke PL, Brechbiel MW. Preparation and preliminary evaluation of a biotin-targeted, lectin-targeted dendrimer-based probe for dual-modality magnetic resonance and fluorescence imaging. Bioconjugate Chem. 2007;18:1474–1482. doi: 10.1021/bc0701085. [DOI] [PubMed] [Google Scholar]
- 82.Mounzer R, Shkarin P, Papademetris X, Constable T, Ruddle NH, Fahmy TM. Dynamic imaging of lymphatic vessels and lymph nodes using a bimodal nanoparticulate contrast agent. Lymphatic Res. Biol. 2007;5:151–158. doi: 10.1089/lrb.2007.5302. [DOI] [PubMed] [Google Scholar]
- 83.Barrett T, Ravizzini G, Choyke PL, Kobayashi H. Dendrimers in medical nanotechnology. IEEE Eng. Med. Biol. Mag. 2009;28:12–22. doi: 10.1109/MEMB.2008.931012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Kaida S, Cabral H, Kumagai M, Kishimura A, Terada Y, Sekino M, Aoki I, Nishiyama N, Tani T, Kataoka K. Visible drug delivery by supramolecular nanocarriers directing to single-platformed diagnosis and therapy of pancreatic tumor model. Cancer Res. 2010;70:7031–7041. doi: 10.1158/0008-5472.CAN-10-0303. [DOI] [PubMed] [Google Scholar]
- 85.Kumagai M, Kano MR, Morishita Y, Ota M, Imai Y, Nishiyama N, Sekino M, Ueno S, Miyazono K, Kataoka K. Enhanced magnetic resonance imaging of experimental pancreatic tumor in vivo by block copolymer-coated magnetite nanoparticles with TGF-beta inhibitor. J. Controlled Release. 2009;140:306–311. doi: 10.1016/j.jconrel.2009.06.002. [DOI] [PubMed] [Google Scholar]
- 86.Imai Y, Kaneko E, Asano T, Kumagai M, Ai M, Kawakami A, Kataoka K, Shimokado K. A novel contrast medium detects increased permeability of rat injured carotid arteries in magnetic resonance T2 mapping imaging. J. Atheroscler. Thromb. 2007;14:65–71. doi: 10.5551/jat.14.65. [DOI] [PubMed] [Google Scholar]
- 87.Mulder WJ, Strijkers GJ, Briley-Saboe KC, Frias JC, Aguinaldo JG, Vucic E, Amirbekian V, Tang C, Chin PT, Nicolay K, Fayad ZA. Molecular imaging of macrophages in atherosclerotic plaques using bimodal PEG-micelles. Magn. Reson. Med. 2007;58:1164–1170. doi: 10.1002/mrm.21315. [DOI] [PubMed] [Google Scholar]
- 88.van Tilborg GA, Mulder WJ, Chin PT, Storm G, Reutelingsperger CP, Nicolay K, Strijkers GJ. Annexin A5-conjugated quantum dots with a paramagnetic lipidic coating for the multimodal detection of apoptotic cells. Bioconjugate Chem. 2006;17:865–868. doi: 10.1021/bc0600463. [DOI] [PubMed] [Google Scholar]
- 89.Strijkers GJ, Kluza E, Van Tilborg GA, van der Schaft DW, Griffioen AW, Mulder WJ, Nicolay K. Paramagnetic and fluorescent liposomes for target-specific imaging and therapy of tumor angiogenesis. Angiogenesis. 2010;13:161–173. doi: 10.1007/s10456-010-9165-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Erten A, Wrasidlo W, Scadeng M, Esener S, Hoffman RM, Bouvet M, Makale M. Magnetic resonance and fluorescence imaging of doxorubicin-loaded nanoparticles using a novel in vivo model. Nanomedicine. 2010;6:797–807. doi: 10.1016/j.nano.2010.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Shan L, Wang S, Sridhar R, Bhujwalla ZM, Wang PC. Dual probe with fluorescent and magnetic properties for imaging solid tumor xenografts. Mol. Imaging. 2007;6:85–95. [PubMed] [Google Scholar]
- 92.Freedman M, Chang EH, Zhou Q, Pirollo KF. Nanodelivery of MRI contrast agent enhances sensitivity of detection of lung cancer metastases. Acad. Radiol. 2009;16:627–637. doi: 10.1016/j.acra.2008.12.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Pirollo KF, Dagata J, Wang P, Freedman M, Vladar A, Fricke S, Ileva L, Zhou Q, Chang EH. A tumor-targeted nanodelivery system to improve early MRI detection of cancer. Mol. Imaging. 2006;5:41–52. [PubMed] [Google Scholar]
- 94.Liepold L, Anderson S, Willits D, Oltrogge L, Frank JA, Douglas T, Young M. Viral capsids as MRI contrast agents. Magn. Reson. Med. 2007;58:871–879. doi: 10.1002/mrm.21307. [DOI] [PubMed] [Google Scholar]
- 95.Szerlip NJ, Walbridge S, Yang L, Morrison PF, Degen JW, Jarrell ST, Kouri J, Kerr PB, Kotin R, Oldfield EH, Lonser RR. Real-time imaging of convection-enhanced delivery of viruses and virus-sized particles. J. Neurosurg. 2007;107:560–567. doi: 10.3171/JNS-07/09/0560. [DOI] [PubMed] [Google Scholar]
- 96.Anderson EA, Isaacman S, Peabody DS, Wang EY, Canary JW, Kirshenbaum K. Viral nanoparticles donning a paramagnetic coat: conjugation of MRI contrast agents to the MS2 capsid. Nano Lett. 2006;6:1160–1164. doi: 10.1021/nl060378g. [DOI] [PubMed] [Google Scholar]
- 97.Manchester M, Singh P. Virus-based nanoparticles (VNPs): platform technologies for diagnostic imaging. Adv. Drug Delivery Rev. 2006;58:1505–1522. doi: 10.1016/j.addr.2006.09.014. [DOI] [PubMed] [Google Scholar]
- 98.Glickson JD, Lund-Katz S, Zhou R, Choi H, Chen IW, Li H, Corbin I, Popov AV, Cao W, Song L, Qi C, Marotta D, et al. Lipoprotein nanoplatform for targeted delivery of diagnostic and therapeutic agents. Mol. Imaging. 2008;7:101–110. [PubMed] [Google Scholar]
- 99.Kobayashi H, Brechbiel MW. Nano-sized MRI contrast agents with dendrimer cores. Adv. Drug Delivery Rev. 2005;57:2271–2286. doi: 10.1016/j.addr.2005.09.016. [DOI] [PubMed] [Google Scholar]
- 100.Choi HS, Liu W, Misra P, Tanaka E, Zimmer JP, Itty Ipe B, Bawendi MG, Frangioni JV. Renal clearance of quantum dots. Nat. Biotechnol. 2007;25:1165–1170. doi: 10.1038/nbt1340. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Longmire M, Choyke PL, Kobayashi H. Dendrimer-based contrast agents for molecular imaging. Curr. Top. Med. Chem. 2008;8:1180–1186. doi: 10.2174/156802608785849021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Longmire M, Choyke PL, Kobayashi H. Clearance properties of nano-sized particles and molecules as imaging agents: considerations and caveats. Nanomedicine (London, U. K.) 2008;3:703–717. doi: 10.2217/17435889.3.5.703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Srinivas M, Cruz LJ, Bonetto F, Heerschap A, Figdor CG, de Vries IJ. Customizable, multi-functional fluorocarbon nanoparticles for quantitative in vivo imaging using 19F MRI and optical imaging. Biomaterials. 2010;31:7070–7077. doi: 10.1016/j.biomaterials.2010.05.069. [DOI] [PubMed] [Google Scholar]
- 104.Mulder WJ, Castermans K, van Beijnum JR, Oude Egbrink MG, Chin PT, Fayad ZA, Lowik CW, Kaijzel EL, Que I, Storm G, Strijkers GJ, Griffioen AW, et al. Molecular imaging of tumor angiogenesis using alphavbeta3-integrin targeted multimodal quantum dots. Angiogenesis. 2009;12:17–24. doi: 10.1007/s10456-008-9124-2. [DOI] [PubMed] [Google Scholar]
- 105.Prinzen L, Miserus RJ, Dirksen A, Hackeng TM, Deckers N, Bitsch NJ, Megens RT, Douma K, Heemskerk JW, Kooi ME, Frederik PM, Slaaf DW, et al. Optical and magnetic resonance imaging of cell death and platelet activation using annexin a5-functionalized quantum dots. Nano Lett. 2007;7:93–100. doi: 10.1021/nl062226r. [DOI] [PubMed] [Google Scholar]
- 106.Talanov VS, Regino CA, Kobayashi H, Bernardo M, Choyke PL, Brechbiel MW. Dendrimer-based nanoprobe for dual modality magnetic resonance and fluorescence imaging. Nano Lett. 2006;6:1459–1463. doi: 10.1021/nl060765q. [DOI] [PubMed] [Google Scholar]
- 107.Wiener EC, Brechbiel MW, Brothers H, Magin RL, Gansow OA, Tomalia DA, Lauterbur PC. Dendrimer-based metal chelates: a new class of magnetic resonance imaging contrast agents. Magn. Reson. Med. 1994;31:1–8. doi: 10.1002/mrm.1910310102. [DOI] [PubMed] [Google Scholar]
- 108.Menjoge AR, Kannan RM, Tomalia DA. Dendrimer-based drug and imaging conjugates: design considerations for nanomedical applications. Drug Discovery Today. 2010;15:171–185. doi: 10.1016/j.drudis.2010.01.009. [DOI] [PubMed] [Google Scholar]
- 109.McCann TE, Kosaka N, Turkbey B, Mitsunaga M, Choyke PL, Kobayashi H. Molecular imaging of tumor invasion and metastases: the role of MRI. NMR Biomed. 2010 Dec 12; doi: 10.1002/nbm.1590. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kobayashi H, Brechbiel MW. Dendrimer-based macromolecular MRI contrast agents: characteristics and application. Mol. Imaging. 2003;2:1–10. doi: 10.1162/15353500200303100. [DOI] [PubMed] [Google Scholar]
- 111.Menjoge AR, Navath RS, Asad A, Kannan S, Kim CJ, Romero R, Kannan RM. Transport and biodistribution of dendrimers across human fetal membranes: implications for intravaginal administration of dendrimer–drug conjugates. Biomaterials. 2010;31:5007–5021. doi: 10.1016/j.biomaterials.2010.02.075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kojima C, Regino C, Umeda Y, Kobayashi H, Kono K. Influence of dendrimer generation and polyethylene glycol length on the biodistribution of PEGylated dendrimers. Int. J. Pharm. 2010;383:293–296. doi: 10.1016/j.ijpharm.2009.09.015. [DOI] [PubMed] [Google Scholar]
- 113.Khan MK, Nigavekar SS, Minc LD, Kariapper MS, Nair BM, Lesniak WG, Balogh LP. In vivo biodistribution of dendrimers and dendrimer nanocomposites—implications for cancer imaging and therapy. Technol. Cancer Res. Treat. 2005;4:603–613. doi: 10.1177/153303460500400604. [DOI] [PubMed] [Google Scholar]
- 114.Mackay PS, Kremers GJ, Kobukai S, Cobb JG, Kuley A, Rosenthal SJ, Koktysh DS, Gore JC, Pham W. Multimodal imaging of dendritic cells using a novel hybrid magnetooptical nanoprobe. Nanomedicine. 2011 Jan 5; doi: 10.1016/j.nano.2010.12.004. [Epub ahead of print]. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Jain K. Advances in the field of nanooncology. BMC Med. 2010;8:83. doi: 10.1186/1741-7015-8-83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Janczewski D, Zhang Y, Das GK, Yi DK, Padmanabhan P, Bhakoo KK, Tan TT, Selvan ST. Bimodal magnetic-fluorescent probes for bioimaging. Microsc. Res. Tech. 2010 Aug 23; doi: 10.1002/jemt.20912. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- 117.Liu Z, Peng R. Inorganic nanomaterials for tumor angiogenesis imaging. Eur. J. Nucl. Med. Mol. Imaging. 2010;37(S1):S147–S163. doi: 10.1007/s00259-010-1452-y. [DOI] [PubMed] [Google Scholar]
- 118.Lim YT, Cho MY, Kang JH, Noh YW, Cho JH, Hong KS, Chung JW, Chung BH. Perfluorodecalin/[InGaP/ZnS quantum dots] nanoemulsions as 19F MR/optical imaging nanoprobes for the labeling of phagocytic and nonphagocytic immune cells. Biomaterials. 2010;31:4964–4971. doi: 10.1016/j.biomaterials.2010.02.065. [DOI] [PubMed] [Google Scholar]
- 119.Sharma P, Singh A, Brown SC, Bengtsson N, Walter GA, Grobmyer SR, Iwakuma N, Santra S, Scott EW, Moudgil BM. Multimodal nanoparticulate bioimaging contrast agents. Methods Mol. Biol. 2010;624:67–81. doi: 10.1007/978-1-60761-609-2_5. [DOI] [PubMed] [Google Scholar]
- 120.Ravizzini G, Turkbey B, Barrett T, Kobayashi H, Choyke PL. Nanoparticles in sentinel lymph node mapping. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2009;1:610–623. doi: 10.1002/wnan.48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Setua S, Menon D, Asok A, Nair S, Koyakutty M. Folate receptor targeted, rare-earth oxide nanocrystals for bi-modal fluorescence and magnetic imaging of cancer cells. Biomaterials. 2010;31:714–729. doi: 10.1016/j.biomaterials.2009.09.090. [DOI] [PubMed] [Google Scholar]
- 122.Gao J, Gu H, Xu B. Multifunctional magnetic nanoparticles: design, synthesis, and biomedical applications. Acc. Chem. Res. 2009;42:1097–1107. doi: 10.1021/ar9000026. [DOI] [PubMed] [Google Scholar]
- 123.Debbage P, Jaschke W. Molecular imaging with nanoparticles: giant roles for dwarf actors. Histochem. Cell Biol. 2008;130:845–875. doi: 10.1007/s00418-008-0511-y. [DOI] [PubMed] [Google Scholar]
- 124.Medintz IL, Mattoussi H, Clapp AR. Potential clinical applications of quantum dots. Int. J. Nanomedicine. 2008;3:151–167. doi: 10.2147/ijn.s614. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Gauden AJ, Phal PM, Drummond KJ. MRI safety; nephrogenic systemic fibrosis and other risks. J. Clin. Neurosci. 2010;17:1097–1104. doi: 10.1016/j.jocn.2010.01.016. [DOI] [PubMed] [Google Scholar]
- 126.Rzigalinski BA, Strobl JS. Cadmium-containing nanoparticles: perspectives on pharmacology and toxicology of quantum dots. Toxicol. Appl. Pharmacol. 2009;238:280–288. doi: 10.1016/j.taap.2009.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Il’yasova D, Schwartz GG. Cadmium and renal cancer. Toxicol. Appl. Pharmacol. 2005;207:179–186. doi: 10.1016/j.taap.2004.12.005. [DOI] [PubMed] [Google Scholar]
- 128.Jain R, Levine M. Correction to the relative risk calculation for gadolinium-enhanced MR imaging and nephrogenic systemic fibrosis. Radiology. 2010;255:307–308. doi: 10.1148/radiol.091726. author reply 308. [DOI] [PubMed] [Google Scholar]
- 129.Edward M, Quinn JA, Burden AD, Newton BB, Jardine AG. Effect of different classes of gadolinium-based contrast agents on control and nephrogenic systemic fibrosis-derived fibroblast proliferation. Radiology. 2010;256:735–743. doi: 10.1148/radiol.10091131. [DOI] [PubMed] [Google Scholar]
- 130.Xiong L, Chen Z, Tian Q, Cao T, Xu C, Li F. High contrast upconversion luminescence targeted imaging in vivo using peptide-labeled nanophosphors. Anal. Chem. 2009;81:8687–8694. doi: 10.1021/ac901960d. [DOI] [PubMed] [Google Scholar]
- 131.Auzel F. Upconversion and anti-Stokes processes with f and d ions in solids. Chem. Rev. 2004;104:139–173. doi: 10.1021/cr020357g. [DOI] [PubMed] [Google Scholar]
- 132.Yang LW, Zhang YY, Li JJ, Li Y, Zhong JX, Chu PK. Magnetic and upconverted luminescent properties of multifunctional lanthanide doped cubic KGdF4 nanocrystals. Nanoscale. 2010;2:2805–2810. doi: 10.1039/c0nr00326c. [DOI] [PubMed] [Google Scholar]
- 133.Li Z, Zhang Y. Facile synthesis of lanthanide nanoparticles with paramagnetic, down- and up-conversion properties. Nanoscale. 2010;2:1240–1243. doi: 10.1039/c0nr00073f. [DOI] [PubMed] [Google Scholar]
- 134.Conn CE, Panchagnula V, Weerawardena A, Waddington LJ, Kennedy DF, Drummond CJ. Lanthanide phytanates: liquid-crystalline phase behavior, colloidal particle dispersions, and potential as medical imaging agents. Langmuir. 2010;26:6240–6249. doi: 10.1021/la904006q. [DOI] [PubMed] [Google Scholar]
- 135.Laurent S, Vander Elst L, Muller RN. Lanthanide complexes for magnetic resonance and optical molecular imaging. Q. J. Nucl. Med. Mol. Imaging. 2009;53:586–603. [PubMed] [Google Scholar]
- 136.Liu G, Conn CE, Drummond CJ. Lanthanide oleates: chelation, self-assembly, and exemplification of ordered nanostructured colloidal contrast agents for medical imaging. J. Phys. Chem. B. 2009;113:15949–15959. doi: 10.1021/jp906344u. [DOI] [PubMed] [Google Scholar]
- 137.Rojas-Quijano FA, Benyo ET, Tircso G, Kalman FK, Baranyai Z, Aime S, Sherry AD, Kovacs Z. Lanthanide(III) complexes of tris(amide) PCTA derivatives as potential bimodal magnetic resonance and optical imaging agents. Chem.–Eur. J. 2009;15:13188–13200. doi: 10.1002/chem.200901095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Picard C, Geum N, Nasso I, Mestre B, Tisnes P, Laurent S, Muller RN, Elst LV. A dual lanthanide probe suitable for optical (Tb3+ luminescence) and magnetic resonance imaging (Gd3+ relaxometry) Bioorg. Med. Chem. Lett. 2006;16:5309–5312. doi: 10.1016/j.bmcl.2006.07.091. [DOI] [PubMed] [Google Scholar]
- 139.Meade TJ, Taylor AK, Bull SR. New magnetic resonance contrast agents as biochemical reporters. Curr. Opin. Neurobiol. 2003;13:597–602. doi: 10.1016/j.conb.2003.09.009. [DOI] [PubMed] [Google Scholar]
- 140.Hanaoka K. Development of responsive lanthanide-based magnetic resonance imaging and luminescent probes for biological applications. Chem. Pharm. Bull. 2010;58:1283–1294. doi: 10.1248/cpb.58.1283. [DOI] [PubMed] [Google Scholar]
- 141.Surman AJ, Bonnet CS, Lowe MP, Kenny GD, Bell JD, Toth E, Vilar R. A Pyrophosphate-Responsive Gadolinium(iii) MRI Contrast Agent. Chem.–Eur. J. 2011;17:223–230. doi: 10.1002/chem.201001397. [DOI] [PubMed] [Google Scholar]
- 142.Bunzli JC. Benefiting from the unique properties of lanthanide ions. Acc. Chem. Res. 2006;39:53–61. doi: 10.1021/ar0400894. [DOI] [PubMed] [Google Scholar]
- 143.Cherry SR. Multimodality imaging: beyond PET/CT and SPECT/CT. Semin. Nucl. Med. 2009;39:348–353. doi: 10.1053/j.semnuclmed.2009.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Zhou J, Yu M, Sun Y, Zhang X, Zhu X, Wu Z, Wu D, Li F. Fluorine-18-labeled Gd3+/Yb3+/Er3+ co-doped NaYF4 nanophosphors for multimodality PET/MR/UCL imaging. Biomaterials. 2011;32:1148–1156. doi: 10.1016/j.biomaterials.2010.09.071. [DOI] [PubMed] [Google Scholar]
- 145.Berti V, Osorio RS, Mosconi L, Li Y, De Santi S, de Leon MJ. Early detection of Alzheimer’s disease with PET imaging. Neurodegener. Dis. 2010;7:131–135. doi: 10.1159/000289222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Kim EE. PET-CT Hybrid Imaging. J. Nucl. Med. 2011 Jan 13; [Epub ahead of print]. [Google Scholar]
- 147.De Wever W, Coolen J, Verschakelen JA. Integrated PET/CT and cancer imaging. JBR-BTR. 2009;92:13–19. [PubMed] [Google Scholar]
- 148.Pichler BJ, Judenhofer MS, Wehrl HF. PET/MRI hybrid imaging: devices and initial results. Eur. Radiol. 2008;18:1077–1086. doi: 10.1007/s00330-008-0857-5. [DOI] [PubMed] [Google Scholar]
- 149.Lee HY, Li Z, Chen K, Hsu AR, Xu C, Xie J, Sun S, Chen X. PET/MRI dual-modality tumor imaging using arginine-glycine-aspartic (RGD)-conjugated radiolabeled iron oxide nanoparticles. J. Nucl. Med. 2008;49:1371–1379. doi: 10.2967/jnumed.108.051243. [DOI] [PubMed] [Google Scholar]
- 150.Heiss WD. The potential of PET/MR for brain imaging. Eur. J. Nucl. Med. Mol. Imaging. 2009;36(S1):105–S112. doi: 10.1007/s00259-008-0962-3. [DOI] [PubMed] [Google Scholar]
- 151.Pichler BJ, Judenhofer MS, Pfannenberg C. Multimodal imaging approaches: PET/CT and PET/MRI. Handb. Exp. Pharmacol. 2008;185/1:109–132. doi: 10.1007/978-3-540-72718-7_6. [DOI] [PubMed] [Google Scholar]
- 152.Wehrl HF, Sauter AW, Judenhofer MS, Pichler BJ. Combined PET/MR imaging—technology and applications. Technol. Cancer Res. Treat. 2010;9:5–20. doi: 10.1177/153303461000900102. [DOI] [PubMed] [Google Scholar]
- 153.Beyer T, Weigert M, Quick HH, Pietrzyk U, Vogt F, Palm C, Antoch G, Muller SP, Bockisch A. MR-based attenuation correction for torso-PET/MR imaging: pitfalls in mapping MR to CT data. Eur. J. Nucl. Med. Mol. Imaging. 2008;35:1142–1146. doi: 10.1007/s00259-008-0734-0. [DOI] [PubMed] [Google Scholar]
- 154.Zaidi H. Is MR-guided attenuation correction a viable option for dual-modality PET/MR imaging? Radiology. 2007;244:639–642. doi: 10.1148/radiol.2443070092. [DOI] [PubMed] [Google Scholar]
- 155.Zaidi H, Mawlawi O, Orton CG. Point/counterpoint. Simultaneous PET/MR will replace PET/CT as the molecular multimodality imaging platform of choice. Med. Phys. 2007;34:1525–1528. doi: 10.1118/1.2732493. [DOI] [PubMed] [Google Scholar]
- 156.Choi JS, Park JC, Nah H, Woo S, Oh J, Kim KM, Cheon GJ, Chang Y, Yoo J, Cheon J. A hybrid nanoparticle probe for dual-modality positron emission tomography and magnetic resonance imaging. Angew. Chem., Int. Ed. 2008;47:6259–6262. doi: 10.1002/anie.200801369. [DOI] [PubMed] [Google Scholar]
- 157.Chan WC, Maxwell DJ, Gao X, Bailey RE, Han M, Nie S. Luminescent quantum dots for multiplexed biological detection and imaging. Curr. Opin. Biotechnol. 2002;13:40–46. doi: 10.1016/s0958-1669(02)00282-3. [DOI] [PubMed] [Google Scholar]
- 158.Kosaka N, Ogawa M, Longmire MR, Choyke PL, Kobayashi H. Multi-targeted multi-color in vivo optical imaging in a model of disseminated peritoneal ovarian cancer. J. Biomed. Opt. 2009;14 doi: 10.1117/1.3083449. 014023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Erogbogbo F, Yong KT, Roy I, Hu R, Law WC, Zhao W, Ding H, Wu F, Kumar R, Swihart MT, Prasad PN. In vivo Targeted Cancer Imaging, Sentinel Lymph Node Mapping and Multi-Channel Imaging with Biocompatible Silicon Nanocrystals. ACS Nano. 2011;5:413–423. doi: 10.1021/nn1018945. [DOI] [PubMed] [Google Scholar]
- 160.Fan HM, Olivo M, Shuter B, Yi JB, Bhuvaneswari R, Tan HR, Xing GC, Ng CT, Liu L, Lucky SS, Bay BH, Ding J. Quantum dot capped magnetite nanorings as high performance nanoprobe for multiphoton fluorescence and magnetic resonance imaging. J. Am. Chem. Soc. 2010;132:14803–14811. doi: 10.1021/ja103738t. [DOI] [PubMed] [Google Scholar]
- 161.Bulte JW. Science to practice: can CT be performed for multicolor molecular imaging? Radiology. 2010;256:675–676. doi: 10.1148/radiol.101127. [DOI] [PubMed] [Google Scholar]
- 162.Le Devedec SE, Lalai R, Pont C, de Bont H, van de Water B. Two-Photon Intravital Multicolor Imaging Combined with Inducible Gene Expression to Distinguish Metastatic Behavior of Breast Cancer Cells in vivo. Mol. Imaging Biol. 2011;13:67–77. doi: 10.1007/s11307-010-0307-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Kosaka N, Ogawa M, Sato N, Choyke PL, Kobayashi H. In vivo real-time, multicolor, quantum dot lymphatic imaging. J. Invest. Dermatol. 2009;129:2818–2822. doi: 10.1038/jid.2009.161. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Tian Z, Chen G, Li X, Liang H, Li Y, Zhang Z, Tian Y. Autofluorescence-free in vivo multicolor imaging using upconversion fluoride nanocrystals. Lasers Med. Sci. 2010;25:479–484. doi: 10.1007/s10103-009-0663-6. [DOI] [PubMed] [Google Scholar]
- 165.Stauffer JK, Khan T, Salcedo R, Hixon JA, Lincoln E, Back TC, Wigginton JM. Multicolor fluorescence-based approaches for imaging cytokine-induced alterations in the neo-vascularization, growth, metastasis, and apoptosis of murine neuroblastoma tumors. J. Immunother. 2006;29:151–164. doi: 10.1097/01.cji.0000190167.76663.c7. [DOI] [PubMed] [Google Scholar]
- 166.Larson DR, Zipfel WR, Williams RM, Clark SW, Bruchez MP, Wise FW, Webb WW. Water-soluble quantum dots for multiphoton fluorescence imaging in vivo. Science. 2003;300:1434–1436. doi: 10.1126/science.1083780. [DOI] [PubMed] [Google Scholar]
- 167.Longmire M, Kosaka N, Ogawa M, Choyke PL, Kobayashi H. Multicolor in vivo targeted imaging to guide real-time surgery of HER2-positive micrometastases in a two-tumor coincident model of ovarian cancer. Cancer Sci. 2009;100:1099–1104. doi: 10.1111/j.1349-7006.2009.01133.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Hama Y, Koyama Y, Urano Y, Choyke PL, Kobayashi H. Two-color lymphatic mapping using Ig-conjugated near infrared optical probes. J. Invest. Dermatol. 2007;127:2351–2356. doi: 10.1038/sj.jid.5700892. [DOI] [PubMed] [Google Scholar]
- 169.Johnson LL, Seldin DW, Keller AM, Wall RM, Bhatia K, Bingham CO, Tresgallo ME. Dual isotope thallium and indium antimyosin SPECT imaging to identify acute infarct patients at further ischemic risk. Circulation. 1990;81:37–45. doi: 10.1161/01.cir.81.1.37. [DOI] [PubMed] [Google Scholar]
- 170.Johnson LL, Lerrick KS, Coromilas J, Seldin DW, Esser PD, Zimmerman JM, Keller AM, Alderson PO, Bigger JT, Cannon PJ. Measurement of infarct size and percentage myocardium infarcted in a dog preparation with single photon-emission computed tomography, thallium-201, and indium 111-monoclonal antimyosin Fab. Circulation. 1987;76:181–190. doi: 10.1161/01.cir.76.1.181. [DOI] [PubMed] [Google Scholar]
- 171.Antunes ML, Johnson LL, Seldin DW, Bhatia K, Tresgallo ME, Greenspan RL, Vaccarino RA, Rodney RA. Diagnosis of right ventricular acute myocardial infarction by dual isotope thallium-201 and indium-111 antimyosin SPECT imaging. Am. J. Cardiol. 1992;70:426–431. doi: 10.1016/0002-9149(92)91184-6. [DOI] [PubMed] [Google Scholar]
- 172.Schoeder H, Topp H, Friedrich M, Jatzkewitz A, Roser M. Thallium and indium antimyosin dual-isotope single-photon emission tomography in acute myocardial infarction to identify patients at further ischaemic risk. Eur. J. Nucl. Med. 1994;21:415–422. doi: 10.1007/BF00171416. [DOI] [PubMed] [Google Scholar]
- 173.McBiles M, Lambert AT, Cote MG, Kim SY. Sestamibi parathyroid imaging. Semin. Nucl. Med. 1995;25:221–234. doi: 10.1016/s0001-2998(95)80012-3. [DOI] [PubMed] [Google Scholar]
- 174.Shih WJ, Stipp V, Magoun S, Ain KB, Pulmano C. Medullary thyroid carcinoma imaged by Tc-99m MIBI SPECT and Tl-201 chloride/Tc-99m pertechnetate subtraction SPECT. Clin. Nucl. Med. 1996;21:213–217. doi: 10.1097/00003072-199603000-00006. [DOI] [PubMed] [Google Scholar]
- 175.Hancu I, Dixon WT, Woods M, Vinogradov E, Sherry AD, Lenkinski RE. CEST and PARACEST MR contrast agents. Acta Radiol. 2010;51:910–923. doi: 10.3109/02841851.2010.502126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Sun PZ. Simplified and scalable numerical solution for describing multi-pool chemical exchange saturation transfer (CEST) MRI contrast. J. Magn. Reson. 2010;205:235–241. doi: 10.1016/j.jmr.2010.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Ward KM, Aletras AH, Balaban RS. A new class of contrast agents for MRI based on proton chemical exchange dependent saturation transfer (CEST) J. Magn. Reson. 2000;143:79–87. doi: 10.1006/jmre.1999.1956. [DOI] [PubMed] [Google Scholar]
- 178.Ward KM, Balaban RS. Determination of pH using water protons and chemical exchange dependent saturation transfer (CEST) Magn. Reson. Med. 2000;44:799–802. doi: 10.1002/1522-2594(200011)44:5<799::aid-mrm18>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
- 179.Terreno E, Stancanello J, Longo D, Castelli DD, Milone L, Sanders HM, Kok MB, Uggeri F, Aime S. Methods for an improved detection of the MRI-CEST effect. Contrast Media Mol. Imaging. 2009;4:237–247. doi: 10.1002/cmmi.290. [DOI] [PubMed] [Google Scholar]
- 180.Terreno E, Castelli DD, Aime S. Encoding the frequency dependence in MRI contrast media: the emerging class of CEST agents. Contrast Media Mol. Imaging. 2010;5:78–98. doi: 10.1002/cmmi.369. [DOI] [PubMed] [Google Scholar]
- 181.McMahon MT, Gilad AA, DeLiso MA, Berman SM, Bulte JW, van Zijl PC. New “multicolor” polypeptide diamagnetic chemical exchange saturation transfer (DIACEST) contrast agents for MRI. Magn. Reson. Med. 2008;60:803–812. doi: 10.1002/mrm.21683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Zhou J, Tryggestad E, Wen Z, Lal B, Zhou T, Grossman R, Wang S, Yan K, Fu DX, Ford E, Tyler B, Blakeley J, et al. Differentiation between glioma and radiation necrosis using molecular magnetic resonance imaging of endogenous proteins and peptides. Nat. Med. 2011;17:130–134. doi: 10.1038/nm.2268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Shah T, Lu L, Dell KM, Pagel MD, Griswold MA, Flask CA. CEST-FISP: a novel technique for rapid chemical exchange saturation transfer MRI at 7 T. Magn. Reson. Med. 2011;65:432–437. doi: 10.1002/mrm.22637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184.Gilad AA, van Laarhoven HW, McMahon MT, Walczak P, Heerschap A, Neeman M, van Zijl PC, Bulte JW. Feasibility of concurrent dual contrast enhancement using CEST contrast agents and superparamagnetic iron oxide particles. Magn. Reson. Med. 2009;61:970–974. doi: 10.1002/mrm.21928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Karcaaltincaba M, Aktas A. Dual-energy CT revisited with multidetector CT: review of principles and clinical applications. Diagn. Interv. Radiol. 2010 doi: 10.4261/1305-3825.DIR.3860-10.0. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- 186.Thomas C, Heuschmid M, Schilling D, Ketelsen D, Tsiflikas I, Stenzl A, Claussen CD, Schlemmer HP. Urinary calculi composed of uric acid, cystine, and mineral salts: differentiation with dual-energy CT at a radiation dose comparable to that of intravenous pyelography. Radiology. 2010;257:402–409. doi: 10.1148/radiol.10100526. [DOI] [PubMed] [Google Scholar]
- 187.Primak AN, Fletcher JG, Vrtiska TJ, Dzyubak OP, Lieske JC, Jackson ME, Williams JC, McCollough CH. Noninvasive differentiation of uric acid versus non-uric acid kidney stones using dual-energy CT. Acad. Radiol. 2007;14:1441–1447. doi: 10.1016/j.acra.2007.09.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Choi HK, Al-Arfaj AM, Eftekhari A, Munk PL, Shojania K, Reid G, Nicolaou S. Dual energy computed tomography in tophaceous gout. Ann. Rheum. Dis. 2009;68:1609–1612. doi: 10.1136/ard.2008.099713. [DOI] [PubMed] [Google Scholar]
- 189.Nicolaou S, Yong-Hing CJ, Galea-Soler S, Hou DJ, Louis L, Munk P. Dual-energy CT as a potential new diagnostic tool in the management of gout in the acute setting. AJR, Am. J. Roentgenol. 2010;194:1072–1078. doi: 10.2214/AJR.09.2428. [DOI] [PubMed] [Google Scholar]
- 190.Brockmann C, Scharf J, Nölte IS, Seiz M, Groden C, Brockmann MA. Dual-energy CT after peri-interventional subarachnoid haemorrhage: a feasibility study. Clin. Neuroradiol. 2010;20:231–235. doi: 10.1007/s00062-010-0036-3. [DOI] [PubMed] [Google Scholar]
- 191.Vlahos I, Godoy MC, Naidich DP. Dual-energy computed tomography imaging of the aorta. J. Thorac. Imaging. 2010;25:289–300. doi: 10.1097/RTI.0b013e3181dc2b4c. [DOI] [PubMed] [Google Scholar]
- 192.Thieme SF, Graute V, Nikolaou K, Maxien D, Reiser MF, Hacker M, Johnson TR. Dual Energy CT lung perfusion imaging-Correlation with SPECT/CT. Eur. J. Radiol. 2010 Dec 22; doi: 10.1016/j.ejrad.2010.11.037. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- 193.Kwee TC, Takahara T, Ochiai R, Katahira K, Van Cauteren M, Imai Y, Nievelstein RA, Luijten PR. Whole-body diffusion-weighted magnetic resonance imaging. Eur. J. Radiol. 2009;70:409–417. doi: 10.1016/j.ejrad.2009.03.054. [DOI] [PubMed] [Google Scholar]
- 194.Lenz C, Klarhofer M, Scheffler K, Winter L, Sommer G. Assessing extracranial tumors using diffusion-weighted whole-body MRI. Z. Med. Phys. 2010 Sep 29; doi: 10.1016/j.zemedi.2010.06.004. [Epub ahead of print]. [DOI] [PubMed] [Google Scholar]
- 195.Jaramillo D. Whole-body MR imaging, bone diffusion imaging: how and why? Pediatr. Radiol. 2010;40:978–984. doi: 10.1007/s00247-010-1608-8. [DOI] [PubMed] [Google Scholar]
- 196.Fischer MA, Nanz D, Hany T, Reiner CS, Stolzmann P, Donati OF, Breitenstein S, Schneider P, Weishaupt D, von Schulthess GK, Scheffel H. Diagnostic accuracy of whole-body MRI/DWI image fusion for detection of malignant tumours: a comparison with PET/CT. Eur. Radiol. 2011;21:246–255. doi: 10.1007/s00330-010-1929-x. [DOI] [PubMed] [Google Scholar]
- 197.Takenaka D, Ohno Y, Matsumoto K, Aoyama N, Onishi Y, Koyama H, Nogami M, Yoshikawa T, Matsumoto S, Sugimura K. Detection of bone metastases in non-small cell lung cancer patients: comparison of whole-body diffusion-weighted imaging (DWI), whole-body MR imaging without and with DWI, whole-body FDG-PET/CT, and bone scintigraphy. J. Magn. Reson. Imaging. 2009;30:298–308. doi: 10.1002/jmri.21858. [DOI] [PubMed] [Google Scholar]
- 198.Tsushima Y, Takano A, Taketomi-Takahashi A, Endo K. Body diffusion-weighted MR imaging using high b-value for malignant tumor screening: usefulness and necessity of referring to T2-weighted images and creating fusion images. Acad. Radiol. 2007;14:643–650. doi: 10.1016/j.acra.2007.02.006. [DOI] [PubMed] [Google Scholar]
- 199.Nagaraja TN, Knight RA, Ewing JR, Karki K, Nagesh V, Fenstermacher JD. Multiparametric magnetic resonance imaging and repeated measurements of blood–brain barrier permeability to contrast agents. Methods Mol. Biol. 2011;686:193–212. doi: 10.1007/978-1-60761-938-3_8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Scherfler C, Scholz SW, Donnemiller E, Decristoforo C, Oberladstatter M, Stefanova N, Diederen E, Virgolini I, Poewe W, Wenning GK. Evaluation of [123I]IBZM pinhole SPECT for the detection of striatal dopamine D2 receptor availability in rats. NeuroImage. 2005;24:822–831. doi: 10.1016/j.neuroimage.2004.10.005. [DOI] [PubMed] [Google Scholar]
- 201.Hassan M, Riley J, Chernomordik V, Smith P, Pursley R, Lee SB, Capala J, Gandjbakhche AH. Fluorescence lifetime imaging system for in vivo studies. Mol. Imaging. 2007;6:229–236. [PMC free article] [PubMed] [Google Scholar]
- 202.Abulrob A, Brunette E, Slinn J, Baumann E, Stanimirovic D. In vivo time domain optical imaging of renal ischemia-reperfusioninjury: discrimination based on fluorescence lifetime. Mol. Imaging. 2007;6:304–314. [PubMed] [Google Scholar]
- 203.Weissleder R, Tung CH, Mahmood U, Bogdanov A. In vivo imaging of tumors with protease-activated near-infrared fluorescent probes. Nat. Biotechnol. 1999;17:375–378. doi: 10.1038/7933. [DOI] [PubMed] [Google Scholar]
- 204.Asanuma D, Kobayashi H, Nagano T, Urano Y. Fluorescence imaging of tumors with “smart” pH-activatable targeted probes. Methods Mol. Biol. 2009;574:47–62. doi: 10.1007/978-1-60327-321-3_5. [DOI] [PubMed] [Google Scholar]
- 205.Kobayashi H, Ogawa M, Alford R, Choyke PL, Urano Y. New strategies for fluorescent probe design in medical diagnostic imaging. Chem. Rev. 2010;110:2620–2640. doi: 10.1021/cr900263j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206.Dafni H, Gilead A, Nevo N, Eilam R, Harmelin A, Neeman M. Modulation of the pharmacokinetics of macromolecular contrast material by avidin chase: MRI, optical, and inductively coupled plasma mass spectrometry tracking of triply labeled albumin. Magn. Reson. Med. 2003;50:904–914. doi: 10.1002/mrm.10638. [DOI] [PubMed] [Google Scholar]
- 207.Kobayashi H, Sakahara H, Hosono M, Yao ZS, Toyama S, Endo K, Konishi J. Improved clearance of radiolabeled biotinylated monoclonal antibody following the infusion of avidin as a “chase” without decreased accumulation in the target tumor. J. Nucl. Med. 1994;35:1677–1684. [PubMed] [Google Scholar]
- 208.Kobayashi H, Sakahara H, Endo K, Yao ZS, Toyama S, Konishi J. Repeating the avidin “chase” markedly improved the biodistribution of radiolabelled biotinylated antibodies and promoted the excretion of additional background radioactivity. Eur. J. Cancer. 1995;31A:1689–1696. doi: 10.1016/0959-8049(95)00244-d. [DOI] [PubMed] [Google Scholar]
- 209.Kobayashi H, Sakahara H, Endo K, Hosono M, Yao ZS, Toyama S, Konishi J. Comparison of the chase effects of avidin, streptavidin, neutravidin, and avidin–ferritin on a radiolabeled biotinylated anti-tumor monoclonal antibody. Jpn. J. Cancer Res. 1995;86:310–314. doi: 10.1111/j.1349-7006.1995.tb03056.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Ogawa M, Kosaka N, Choyke PL, Kobayashi H. Tumor-specific detection of an optically targeted antibody combined with a quencher-conjugated neutravidin “quencher-chaser”: a dual “quench and chase” strategy to improve target to nontarget ratios for molecular imaging of cancer. Bioconjugate Chem. 2009;20:147–154. doi: 10.1021/bc8003765. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Razkin J, Josserand V, Boturyn D, Jin ZH, Dumy P, Favrot M, Coll JL, Texier I. Activatable fluorescent probes for tumour-targeting imaging in live mice. Chem Med Chem. 2006;1:1069–1072. doi: 10.1002/cmdc.200600118. [DOI] [PubMed] [Google Scholar]
- 212.Lee S, Park K, Kim K, Choi K, Kwon IC. Activatable imaging probes with amplified fluorescent signals. Chem. Commun. 2008:4250–4260. doi: 10.1039/b806854m. [DOI] [PubMed] [Google Scholar]
- 213.Schellenberger E, Rudloff F, Warmuth C, Taupitz M, Hamm B, Schnorr J. Protease-specific nanosensors for magnetic resonance imaging. Bioconjugate Chem. 2008;19:2440–2445. doi: 10.1021/bc800330k. [DOI] [PubMed] [Google Scholar]
- 214.Hama Y, Urano Y, Koyama Y, Choyke PL, Kobayashi H. Activatable fluorescent molecular imaging of peritoneal metastases following pretargeting with a biotinylated monoclonal antibody. Cancer Res. 2007;67:3809–3817. doi: 10.1158/0008-5472.CAN-06-3794. [DOI] [PubMed] [Google Scholar]
- 215.Hama Y, Urano Y, Koyama Y, Kamiya M, Bernardo M, Paik RS, Shin IS, Paik CH, Choyke PL, Kobayashi H. A target cell-specific activatable fluorescence probe for in vivo molecular imaging of cancer based on a selfquenched avidin–rhodamine conjugate. Cancer Res. 2007;67:2791–2799. doi: 10.1158/0008-5472.CAN-06-3315. [DOI] [PubMed] [Google Scholar]
- 216.Urano Y. Sensitive and selective tumor imaging with novel and highly activatable fluorescence probes. Anal. Sci. 2008;24:51–53. doi: 10.2116/analsci.24.51. [DOI] [PubMed] [Google Scholar]
- 217.Wu AM, Senter PD. Arming antibodies: prospects and challenges for immunoconjugates. Nat. Biotechnol. 2005;23:1137–1146. doi: 10.1038/nbt1141. [DOI] [PubMed] [Google Scholar]
- 218.Saga T, Weinstein JN, Jeong JM, Heya T, Lee JT, Le N, Paik CH, Sung C, Neumann RD. Two-step targeting of experimental lung metastases with biotinylated antibody and radiolabeled streptavidin. Cancer Res. 1994;54:2160–2165. [PubMed] [Google Scholar]
- 219.Kosaka N, Ogawa M, Paik DS, Paik CH, Choyke PL, Kobayashi H. Semiquantitative assessment of the microdistribution of fluorescence-labeled monoclonal antibody in small peritoneal disseminations of ovarian cancer. Cancer Sci. 2010;101:820–825. doi: 10.1111/j.1349-7006.2009.01423.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Kobayashi H, Kawamoto S, Jo SK, Bryant HL, Brechbiel MW, Star RA. Macromolecular MRI contrast agents with small dendrimers: pharmacokinetic differences between sizes and cores. Bioconjugate Chem. 2003;14:388–394. doi: 10.1021/bc025633c. [DOI] [PubMed] [Google Scholar]
- 221.Ogawa M, Regino CA, Marcelino B, Williams M, Kosaka N, Bryant LH, Choyke PL, Kobayashi H. New nanosized biocompatible MR contrast agents based on lysine-dendri-graft macromolecules. Bioconjugate Chem. 2010;21:955–960. doi: 10.1021/bc9005442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Ahmed N, Langlois R, Rodrigue S, Benard F, van Lier JE. Automated synthesis of 11beta-methoxy-4,16alpha-[16alpha-(18)F]difluoroestradiol (4F-M[(18)F]FES) for estrogen receptor imaging by positron emission tomography. Nucl. Med. Biol. 2007;34:459–464. doi: 10.1016/j.nucmedbio.2007.02.001. [DOI] [PubMed] [Google Scholar]
- 223.Yoshida Y, Kurokawa T, Sawamura Y, Shinagawa A, Okazawa H, Fujibayashi Y, Kotsuji F. The positron emission tomography with F18 17beta-estradiol has the potential to benefit diagnosis and treatment of endometrial cancer. Gynecol. Oncol. 2007;104:764–766. doi: 10.1016/j.ygyno.2006.10.024. [DOI] [PubMed] [Google Scholar]
- 224.Linden HM, Stekhova SA, Link JM, Gralow JR, Livingston RB, Ellis GK, Petra PH, Peterson LM, Schubert EK, Dunnwald LK, Krohn KA, Mankoff DA. Quantitative fluoroestradiol positron emission tomography imaging predicts response to endocrine treatment in breast cancer. J. Clin. Oncol. 2006;24:2793–2799. doi: 10.1200/JCO.2005.04.3810. [DOI] [PubMed] [Google Scholar]
- 225.Levashova Z, Backer MV, Horng G, Felsher D, Backer JM, Blankenberg FG. SPECT and PET imaging of EGF receptors with site-specifically labeled EGF and dimeric EGF. Bioconjugate Chem. 2009;20:742–749. doi: 10.1021/bc800443w. [DOI] [PubMed] [Google Scholar]
- 226.Yang W, Barth RF, Wu G, Huo T, Tjarks W, Ciesielski M, Fenstermaker RA, Ross BD, Wikstrand CJ, Riley KJ, Binns PJ. Convection enhanced delivery of boronated EGF as a molecular targeting agent for neutron capture therapy of brain tumors. J. Neuro-Oncol. 2009;95:355–365. doi: 10.1007/s11060-009-9945-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Yeh HH, Ogawa K, Balatoni J, Mukhapadhyay U, Pal A, Gonzalez-Lepera C, Shavrin A, Soghomonyan S, Flores L, 2nd, Young D, Volgin AY, Najjar AM, et al. Molecular imaging of active mutant L858R EGF receptor (EGFR) kinaseexpressing nonsmall cell lung carcinomas using PET/CT. Proc. Natl. Acad. Sci. U. S. A. 2011;108:1603–1608. doi: 10.1073/pnas.1010744108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Nakada H, Inoue M, Tanaka N, Yamashina I. Coexpression of cancer-associated carbohydrate antigens Tn and sialyl Tn. Glycoconjugate J. 1994;11:262–265. doi: 10.1007/BF00731227. [DOI] [PubMed] [Google Scholar]
- 229.Ikeda Y, Kuwano H, Baba K, Ikebe M, Matushima T, Adachi Y, Mori M, Sugimachi K. Expression of Sialyl-Tn antigens in normal squamous epithelium, dysplasia, and squamous cell carcinoma in the esophagus. Cancer Res. 1993;53:1706–1708. [PubMed] [Google Scholar]
- 230.Doerr RJ, Herrera L, Abdel-Nabi H. In-111 CYT-103 monoclonal antibody imagingin patients with suspected recurrent colorectal cancer. Cancer. 1993;71:4241–4247. doi: 10.1002/1097-0142(19930615)71:12+<4241::aid-cncr2820711813>3.0.co;2-g. [DOI] [PubMed] [Google Scholar]
- 231.Lamki LM, Buzdar AU, Singletary SE, Rosenblum MG, Bhadkamkar V, Esparza L, Podoloff DA, Zukiwski A, Hortobagyi GN, Murray JL. Indium-111-labeled B72.3 monoclonal antibody in the detection and staging of breast cancer: a phase I study. J. Nucl. Med. 1991;32:1326–1332. [PubMed] [Google Scholar]
- 232.Krug LM, Milton DT, Jungbluth AA, Chen LC, Quaia E, Pandit-Taskar N, Nagel A, Jones J, Kris MG, Finn R, Smith-Jones P, Scott AM, et al. Targeting Lewis Y (Le(y)) in small cell lung cancer with a humanized monoclonal antibody, hu3S193: a pilot trial testing two dose levels. J. Thorac. Oncol. 2007;2:947–952. doi: 10.1097/JTO.0b013e3181560dcc. [DOI] [PubMed] [Google Scholar]
- 233.Quaia E, Krug LM, Pandit-Taskar N, Nagel A, Reuter VE, Humm J, Divgi C. The value of gamma camera and computed tomography data set coregistration to assess Lewis Y antigen targeting in small cell lung cancer by (111)Indium-labeled humanized monoclonal antibody 3S193. Eur. J. Radiol. 2008;67:292–299. doi: 10.1016/j.ejrad.2007.07.004. [DOI] [PubMed] [Google Scholar]
- 234.Davis SC, Samkoe KS, O’Hara JA, Gibbs-Strauss SL, Payne HL, Hoopes PJ, Paulsen KD, Pogue BW. MRI-coupled fluorescence tomography quantifies EGFR activity in brain tumors. Acad. Radiol. 2010;17:271–276. doi: 10.1016/j.acra.2009.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Nitin N, Rosbach KJ, El-Naggar A, Williams M, Gillenwater A, Richards-Kortum RR. Optical molecular imaging of epidermal growth factor receptor expression to improve detection of oral neoplasia. Neoplasia. 2009;11:542–551. doi: 10.1593/neo.09188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Arndt-Jovin DJ, Kantelhardt SR, Caarls W, de Vries AH, Giese A, Jovin Ast TM. Tumor-targeted quantum dots can help surgeons find tumor boundaries. IEEE Trans. NanoBiosci. 2009;8:65–71. doi: 10.1109/TNB.2009.2016548. [DOI] [PubMed] [Google Scholar]
- 237.Manning HC, Merchant NB, Foutch AC, Virostko JM, Wyatt SK, Shah C, McKinley ET, Xie J, Mutic NJ, Washington MK, LaFleur B, Tantawy MN, et al. Molecular imaging of therapeutic response to epidermal growth factor receptor blockade in colorectal cancer. Clin. Cancer Res. 2008;14:7413–7422. doi: 10.1158/1078-0432.CCR-08-0239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Remy S, Reilly RM, Sheldon K, Gariepy J. A new radioligand for the epidermal growth factor receptor: 111In labeled human epidermal growth factor derivatized with a bifunctional metal-chelating peptide. Bioconjugate Chem. 1995;6:683–690. doi: 10.1021/bc00036a004. [DOI] [PubMed] [Google Scholar]
- 239.Fuller GN, Bigner SH. Amplified cellular oncogenes in neoplasms of the human central nervous system. Mutat. Res. 1992;276:299–306. doi: 10.1016/0165-1110(92)90016-3. [DOI] [PubMed] [Google Scholar]
- 240.Naruki Y, Carrasquillo JA, Reynolds JC, Maloney PJ, Frincke JM, Neumann RD, Larson SM. Differential cellular catabolism of 111In: 90Y and 125I radiolabeled T101 anti-CD5 monoclonal antibody. Int. J. Rad. Appl. Instrum. B. 1990;17:201–207. doi: 10.1016/0883-2897(90)90148-t. [DOI] [PubMed] [Google Scholar]
- 241.Kobayashi H, Kao CH, Kreitman RJ, Le N, Kim MK, Brechbiel MW, Paik CH, Pastan I, Carrasquillo JA. Pharmacokinetics of 111In- and 125I-labeled antiTac single-chain Fv recombinant immunotoxin. J. Nucl. Med. 2000;41:755–762. [PubMed] [Google Scholar]
- 242.Foged C, Nielsen HM. Cell-penetrating peptides for drug delivery across membrane barriers. Expert Opin. Drug Delivery. 2008;5:105–117. doi: 10.1517/17425247.5.1.105. [DOI] [PubMed] [Google Scholar]
- 243.Jiang T, Olson ES, Nguyen QT, Roy M, Jennings PA, Tsien RY. Tumor imaging by means of proteolytic activation of cell-penetrating peptides. Proc. Natl. Acad. Sci. U. S. A. 2004;101:17867–17872. doi: 10.1073/pnas.0408191101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Watkins GA, Jones EF, Scott Shell M, VanBrocklin HF, Pan MH, Hanrahan SM, Feng JJ, He J, Sounni NE, Dill KA, Contag CH, Coussens LM, et al. Development of an optimized activatable MMP-14 targeted SPECT imaging probe. Bioorg. Med. Chem. 2009;17:653–659. doi: 10.1016/j.bmc.2008.11.078. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Chen B, Liu Q, Zhang Y, Xu L, Fang X. Transmembrane delivery of the cell-penetrating peptide conjugated semiconductor quantum dots. Langmuir. 2008;24:11866–11871. doi: 10.1021/la802048s. [DOI] [PubMed] [Google Scholar]
- 246.Kersemans V, Kersemans K, Cornelissen B. Cell penetrating peptides for in vivo molecular imaging applications. Curr. Pharm. Des. 2008;14:2415–2447. doi: 10.2174/138161208785777432. [DOI] [PubMed] [Google Scholar]
- 247.Kobayashi H, Choyke PL. Target-Cancer-Cell-Specific Activatable Fluorescence Imaging Probes: Rational Design and in vivo Applications. Acc. Chem. Res. 2011;44:83–90. doi: 10.1021/ar1000633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Bremer C, Bredow S, Mahmood U, Weissleder R, Tung CH. Optical imaging of matrix metalloproteinase-2 activity in tumors: feasibility study in a mouse model. Radiology. 2001;221:523–529. doi: 10.1148/radiol.2212010368. [DOI] [PubMed] [Google Scholar]
- 249.Sheth RA, Maricevich M, Mahmood U. In vivo optical molecular imaging of matrix metalloproteinase activity in abdominal aortic aneurysms correlates with treatment effects on growth rate. Atherosclerosis. 2010;212:181–187. doi: 10.1016/j.atherosclerosis.2010.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Bremer C, Tung CH, Weissleder R. In vivo molecular target assessment of matrix metalloproteinase inhibition. Nat. Med. 2001;7:743–748. doi: 10.1038/89126. [DOI] [PubMed] [Google Scholar]
- 251.Mahmood U. Science to practice: can an enzyme-sensitive MR contrast agent be used to image inflammation in aneurysms? Radiology. 2009;252:627–628. doi: 10.1148/radiol.2523090736. [DOI] [PubMed] [Google Scholar]
- 252.Hama Y, Urano Y, Koyama Y, Gunn AJ, Choyke PL, Kobayashi H. A self-quenched galactosamine–serum albumin–rhodamineX conjugate: a “smart” fluorescent molecular imaging probe synthesized with clinically applicable material for detecting peritoneal ovarian cancer metastases. Clin. Cancer Res. 2007;13:6335–6343. doi: 10.1158/1078-0432.CCR-07-1004. [DOI] [PubMed] [Google Scholar]
- 253.Jares-Erijman EA, Jovin TM. Imaging molecular interactions in living cells by FRET microscopy. Curr. Opin. Chem. Biol. 2006;10:409–416. doi: 10.1016/j.cbpa.2006.08.021. [DOI] [PubMed] [Google Scholar]
- 254.Jares-Erijman EA, Jovin TM. FRET imaging. Nat. Biotechnol. 2003;21:1387–1395. doi: 10.1038/nbt896. [DOI] [PubMed] [Google Scholar]
- 255.Ogawa M, Kosaka N, Regino CA, Mitsunaga M, Choyke PL, Kobayashi H. High sensitivity detection of cancer in vivo using a dual-controlled activation fluorescent imaging probe based on H-dimer formation and pH activation. Mol. BioSyst. 2010;6:888–893. doi: 10.1039/b917876g. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256.Ogawa M, Kosaka N, Choyke PL, Kobayashi H. H-type dimer formation of fluorophores: a mechanism for activatable in vivo optical molecular imaging. ACS Chem. Biol. 2009;4:535–546. doi: 10.1021/cb900089j. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Bader AN, Hoetzl S, Hofman EG, Voortman J, van Bergen En Henegouwen PM, van Meer G, Gerritsen HC. Homo-FRET Imaging as a Tool to Quantify Protein and Lipid Clustering. ChemPhysChem. 2010 doi: 10.1002/cphc.201000801. [DOI] [PubMed] [Google Scholar]
- 258.Hudson GA, Cheng L, Yu J, Yan Y, Dyer DJ, McCarroll ME, Wang L. Computational studies on response and binding selectivity of fluorescence sensors. J. Phys. Chem. B. 2010;114:870–876. doi: 10.1021/jp908368k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Zhao Y, Zheng Q, Dakin K, Xu K, Martinez ML, Li WH. New caged coumarin fluorophores with extraordinary uncaging cross sections suitable for biological imaging applications. J. Am. Chem. Soc. 2004;126:4653–4663. doi: 10.1021/ja036958m. [DOI] [PubMed] [Google Scholar]
- 260.Matsumoto T, Urano Y, Shoda T, Kojima H, Nagano T. A thiol-reactive fluorescence probe based on donor-excited photoinduced electron transfer: key role of ortho substitution. Org. Lett. 2007;9:3375–3377. doi: 10.1021/ol071352e. [DOI] [PubMed] [Google Scholar]
- 261.Sunahara H, Urano Y, Kojima H, Nagano T. Design and synthesis of a library of BODIPY-based environmental polarity sensors utilizing photoinduced electron-transfer-controlled fluorescence ON/OFF switching. J. Am. Chem. Soc. 2007;129:5597–5604. doi: 10.1021/ja068551y. [DOI] [PubMed] [Google Scholar]
- 262.Mineno T, Ueno T, Urano Y, Kojima H, Nagano T. Creation of superior carboxyfluorescein dyes by blocking donor-excited photoinduced electron transfer. Org. Lett. 2006;8:5963–5966. doi: 10.1021/ol0623926. [DOI] [PubMed] [Google Scholar]
- 263.Gabe Y, Ueno T, Urano Y, Kojima H, Nagano T. Tunable design strategy for fluorescence probes based on 4-substituted BODIPY chromophore: improvement of highly sensitive fluorescence probe for nitric oxide. Anal. Bioanal. Chem. 2006;386:621–626. doi: 10.1007/s00216-006-0587-y. [DOI] [PubMed] [Google Scholar]
- 264.Ueno T, Urano Y, Setsukinai K, Takakusa H, Kojima H, Kikuchi K, Ohkubo K, Fukuzumi S, Nagano T. Rational principles for modulating fluorescence properties of fluorescein. J. Am. Chem. Soc. 2004;126:14079–14085. doi: 10.1021/ja048241k. [DOI] [PubMed] [Google Scholar]
- 265.Hanaoka K, Muramatsu Y, Urano Y, Terai T, Nagano T. Design and synthesis of a highly sensitive off-on fluorescent chemosensor for zinc ions utilizing internal charge transfer. Chem.–Eur. J. 2010;16:568–572. doi: 10.1002/chem.200901591. [DOI] [PubMed] [Google Scholar]
- 266.Kobayashi T, Urano Y, Kamiya M, Ueno T, Kojima H, Nagano T. Highly activatable and rapidly releasable caged fluorescein derivatives. J. Am. Chem. Soc. 2007;129:6696–6697. doi: 10.1021/ja070376d. [DOI] [PubMed] [Google Scholar]
- 267.Wang Q, Scheigetz J, Roy B, Ramachandran C, Gresser MJ. Novel caged fluorescein diphosphates as photoactivatable substrates for protein tyrosine phosphatases. Biochim. Biophys. Acta. 2002;1601:19–28. doi: 10.1016/s1570-9639(02)00412-0. [DOI] [PubMed] [Google Scholar]
- 268.Kiskin NI, Chillingworth R, McCray JA, Piston D, Ogden D. The efficiency of two-photon photolysis of a “caged” fluorophore, o-1-(2-nitrophenyl)ethylpyranine, in relation to photodamage of synaptic terminals. Eur. Biophys. J. 2002;30:588–604. doi: 10.1007/s00249-001-0187-x. [DOI] [PubMed] [Google Scholar]
- 269.Kozlowski DJ, Weinberg ES. Photoactivatable (caged) fluorescein as a cell tracer for fate mapping in the zebrafish embryo. Methods Mol. Biol. 2000;135:349–355. doi: 10.1385/1-59259-685-1:349. [DOI] [PubMed] [Google Scholar]
- 270.Desai A, Mitchison TJ. Preparation and characterization of caged fluorescein tubulin. Methods Enzymol. 1998;298:125–132. doi: 10.1016/s0076-6879(98)98014-4. [DOI] [PubMed] [Google Scholar]
- 271.Kenmoku S, Urano Y, Kojima H, Nagano T. Development of a highly specific rhodamine-based fluorescence probe for hypochlorous acid and its application to real-time imaging of phagocytosis. J. Am. Chem. Soc. 2007;129:7313–7318. doi: 10.1021/ja068740g. [DOI] [PubMed] [Google Scholar]
- 272.Xiang Y, Tong A. A new rhodamine-based chemosensor exhibiting selective Fe(iii)-amplified fluorescence. Org. Lett. 2006;8:1549–1552. doi: 10.1021/ol060001h. [DOI] [PubMed] [Google Scholar]
- 273.Hargreaves RJ. The role of molecular imaging in drug discovery and development. Clin. Pharmacol. Ther. 2008;83:349–353. doi: 10.1038/sj.clpt.6100467. [DOI] [PubMed] [Google Scholar]
- 274.Gross S, Piwnica-Worms D. Molecular imaging strategies for drug discovery and development. Curr. Opin. Chem. Biol. 2006;10:334–342. doi: 10.1016/j.cbpa.2006.06.028. [DOI] [PubMed] [Google Scholar]
- 275.Ogawa M, Regino CA, Seidel J, Green MV, Xi W, Williams M, Kosaka N, Choyke PL, Kobayashi H. Dual-modality molecular imaging using antibodies labeled with activatable fluorescence and a radionuclide for specific and quantitative targeted cancer detection. Bioconjugate Chem. 2009;20:2177–2184. doi: 10.1021/bc900362k. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Ogawa M, Regino CA, Choyke PL, Kobayashi H. In vivo target-specific activatable near-infrared optical labeling of humanized monoclonal antibodies. Mol. Cancer Ther. 2009;8:232–239. doi: 10.1158/1535-7163.MCT-08-0862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Alford R, Ogawa M, Hassan M, Gandjbakhche AH, Choyke PL, Kobayashi H. Fluorescence lifetime imaging of activatable target specific molecular probes. Contrast Media Mol. Imaging. 2010;5:1–8. doi: 10.1002/cmmi.360. [DOI] [PMC free article] [PubMed] [Google Scholar]