ARTICLE IN PRESS
Imaging in Drug Development James Nairne*, Peter B. Iveson*, Andreas Meijer† *GE Healthcare, The Grove Centre, Amersham, Buckinghamshire, United Kingdom † GE Healthcare AS, Nydalen, Oslo, Norway
Contents 1. Introduction 2. Magnetic Resonance Imaging 2.1 Use of MRI in the Clinic 2.2 MRI in Drug Development 3. Nuclear Medicine Imaging 3.1 Single-Photon Emission Computed Tomography 3.2 Positron Emission Tomography 4. Summary and Future Prospects References
1 3 4 6 9 9 16 41 42
Keywords: Imaging, Radiochemistry, MRI, PET, SPECT, Fluorine-18, Clinical trials
1. INTRODUCTION In the past 50 years, imaging in medicine has become a staple in the diagnosis of disease. The technology for detection and processing of images has increased in complexity leading to an increase in sophistication in the information that can be derived from these images. Several different underlying technologies and techniques allow various aspects of the human body to be examined. Five basic modalities are used to provide non-invasive imaging for uncovering anatomical and functional detail in the clinic. These are X-ray, ultrasound, magnetic resonance imaging (MRI), nuclear medicine and optical imaging. X-ray was the earliest of the techniques and was able to show only hard tissue through attenuation of the signal from an X-ray source. Subsequent innovations included addition of contrast agents that absorb X-rays and allow complicated cardiac and pulmonary vasculature to be visualised. Abnormalities could then be identified prior to any surgical intervention. Progress in Medicinal Chemistry ISSN 0079-6468 http://dx.doi.org/10.1016/bs.pmch.2014.10.002
#
2014 Elsevier B.V. All rights reserved.
1
ARTICLE IN PRESS 2
James Nairne et al.
The early 1970s saw the development of computed tomography (CT) in which a 3D image of the anatomy is generated from exposure of the patient to X-rays. The information acquired from X-ray and CT is exclusively anatomical in detail. Its use within drug development has been limited to gross anatomical changes, for example, in tumour mass, and in conjugation with nuclear medicine techniques, in which it provides the anatomical context for the signal obtained from the radioactivity in dual modality techniques such as single-photon emission computed tomography (SPECT) and positron emission tomography (PET) (vide infra). Ultrasound imaging is another important method used in the diagnostic imaging field. In this technique, an ultrasound wave is produced by a transducer and guided into the body, where it is reflected from boundaries at which there are changes in tissue density. Its strength is in the investigation of soft tissue, and it benefits from the lack of use of ionising radiation, portability of equipment and the provision of real-time images. These features make ultrasound very useful in the field of antenatal care. Like X-ray, however, the major limitation of ultrasound is that it can provide information only on an anatomical level and as such has little role to play in drug development outside its diagnostic applications. MRI is an ionising radiation free technology that relies on the detection of a radiofrequency (RF) signal emitted by protons in a magnetic field. As with CT and ultrasound, MRI is capable of visualising anatomical detail; however, the MR technology is also capable of generating images based on the properties of endogenous molecules such as water and haemoglobin. For example, it is possible to assess the macroscopic movements of water to measure blood velocity. It is also possible to measure the microscopic movements of water molecules to assess cellular density. Another example is the possibility of generating images that are influenced by local concentration of haemoglobin and deoxyhaemoglobin, in order to assess the oxygenation of blood. The great capacity of MRI to visualise anatomical detail, without ionising radiation, and to simultaneously provide additional functional information on blood flow, cellular density and oxygenation renders it a very powerful imaging modality in drug development. MR is discussed in more detail in Section 2. Optical imaging is another modality that requires no ionising radiation. In general, the technique uses a fluorescence signal as the imaging output. Fluorescence is the excitation of a chromophore and dissipation of the energy absorbed in the form of light of longer wavelength. The human body is opaque to light in the visible portion of the electromagnetic spectrum and
ARTICLE IN PRESS 3
Imaging in Drug Development
CH3 CH3 N
H3C H3C N+
O S O O
OO S O
Figure 1 Structure of indocyanine green.
has absorption minima in the near infrared. Any reporter therefore needs to be able to absorb and emit light in the near infrared portion of the spectrum (750–900 nm) [1]. There are dyes that meet this criterion, the most widely used of which are the cyanine dyes. An example is the heptamethine cyanine, indocyanine green (Figure 1). The reporter is a large molecule and, as such, it is only really applicable for labelling of peptides and proteins. In addition, the absorption of light by tissue and photon scatter limits useful detection to a few millimetres [2]. Although some optical imaging agents have been used diagnostically in the clinic and in preclinical imaging, there are no examples of the modality being used in translational medicine. The last of the major modalities is nuclear medicine imaging. Early imaging agents tracked accumulation in target organs, for example, calcium-45 for bone and iodide-123 for thyroid. The development of radiochemistry has led to an increasingly sophisticated arsenal of chemical techniques that allow the production of radiotracers that are able to reveal the underlying biochemistry of disease with good spatial resolution. The description of these techniques and illustrations of their applications in the clinic forms the bulk of this chapter.
2. MAGNETIC RESONANCE IMAGING MRI is a non-invasive modality that uses strong magnetic fields and RF pulses to produce images of organs and internal structures in the body. Unlike CT, PET and SPECT imaging modalities, no ionising radiation is utilised. The main advantage with MRI over other imaging modalities is the contrast resolution that facilitates anatomical imaging with great soft tissue detail. CT is also capable of anatomical imaging; however, soft tissue detail is not as good as for MRI.
ARTICLE IN PRESS 4
James Nairne et al.
The MR modality is very flexible with a great number of possibilities for adjusting imaging parameters, in order to further enhance the contrast to background ratio for a certain pathology. It is also possible to inject a contrast agent to further enhance contrast between pathological and normal tissue; however, most MR procedures are performed without the use of a contrast agent. In a simplified view, an MRI scanner consists of three main components: a powerful magnet, an RF transmitter and an RF receiver. When a patient is inserted into an MRI scanner, the powerful magnet will magnetise hydrogen nuclei of molecules present in the patient (principally water and fat) so that they align, either parallel or anti-parallel, with the magnetic field giving rise to a net magnetisation (sum of magnetised hydrogen nuclei). This net magnetisation is excited by an RF pulse to give an unstable state that eventually will return to its initial state, emitting a detectable RF signal, in a process called relaxation. The relaxation proceeds via two independent processes (T1 and T2) which happen simultaneously. These two processes are called intrinsic contrast parameters as they are inherent to various types of tissue and cannot be changed. It is possible to weight the contrast of an image towards one of the two processes by adjusting imaging parameters. The relaxation process can be accelerated by introducing a contrast agent. Several MR contrast agents are available in the clinic and all incorporate a paramagnetic gadolinium ion [3]. The gadolinium ion will accelerate the relaxation process of protons that are in close proximity to the contrast agent, leading to a dramatically altered relaxation process that can be visualised by weighted imaging. The gadolinium ion is toxic and must be entrapped in a chelate to render it safe to use. There are several types of gadolinium chelates (Figure 2) in clinical use and they are all based on polyamino polycarboxylate structures that entrap the gadolinium ion, rendering it safe to use while maintaining the relaxation-enhancing properties.
2.1. Use of MRI in the Clinic MRI is used to diagnose or monitor treatment for a variety of medical conditions by visualisation of anatomical details, detection of pathological tissue or assessment of organ functionality. MR is frequently used to image the central nervous system (CNS) where different compartments and structures can be visualised in high detail in order to detect defects such as aneurysms, tumours, infections or stroke (Figure 3).
ARTICLE IN PRESS 5
Imaging in Drug Development
O
N O O
O
O
N
O
O
Gd N
H N
O
O
H3C
N
OO
Gadoterate meglumine Dotarem Guerbet
N H
N
O
O
N
O
N
O N
O
OO
O
Gadodiamide Omniscan GE
O
N
Gd
O N
O
O-
O
Gd
O
O
CH3
Gadopentetate dimeglumine Magnevist Bayer Schering
Figure 2 Examples of gadolinium chelates in use as MR contrast agents.
Figure 3 Anatomical head scan. Adapted with permission from GE Healthcare.
Another example is cardiovascular MR where the vascular anatomy, function, blood perfusion and tissue characteristics can be imaged and quantitated [4]. For some diagnostic situations, it is preferred to inject a contrast agent that will distribute in the blood and the extracellular space before excretion through the kidneys or the liver. Contrast agents are frequently used for angiography applications, where structural or flow related abnormalities are identified and characterised. Contrast agents are also used for oncology applications where the increased perfusion and permeability of tumours result in a high
ARTICLE IN PRESS 6
James Nairne et al.
concentration of contrast agent. The lesions are then characterised based on structural and morphological criteria (size and shape). MRI is especially powerful for the diagnosis of brain lesions, as the injected agent will leak into the brain only if the blood–brain barrier is damaged, such as when a tumour has disrupted the barrier.
2.2. MRI in Drug Development 2.2.1 Oncology The efficacy of an oncological treatment is often assessed by morphological criteria such as tumour size and shape [5]. Unfortunately, the progression or regression of a tumour following a specific treatment is not always reflected in a morphological change in a predictable and timely manner, potentially leading to unnecessarily long treatment regimes with expensive and ineffective therapeutic agents. There is, thus, a great need to assess therapy response on tumour function at an earlier stage, to allow for a change in treatment plan if ineffective. Currently there are two main MRI techniques, dynamic contrast enhancement (DCE) and diffusion weighted imaging (DWI) that are in clinical use to assess therapy response. A search of www.clinicaltrials.gov indicates that there are 52 active clinical trials using DCE or DWI as a diagnostic tool in evaluating therapeutic response. 2.2.1.1 Dynamic Contrast Enhancement
Imaging tumour vascularisation has received a lot of interest over the past few years as it gives a better understanding of tumour function, compared to morphological criteria alone. DCE is used to study the flow of contrast agent through the tumour to give data on blood volume, perfusion and permeability. There are two types of DCE in clinical use, qualitative and quantitative. Qualitative DCE studies the wash in and wash out pattern in a tumour to give crude information on its vascularisation that is used in conjunction with morphological data [6]. Quantitative DCE utilises pharmacokinetic modelling of blood flow through various compartments to give parameters that numerically describe perfusion, blood volume and permeability [7]. Although quantitative DCE holds great promise for the detailed study of tumour physiology, it is technically challenging and requires strict control of the imaging setup. The parameters obtained from quantitative DCE are very sensitive towards the choice of contrast agent, scanner type, pulse sequence and pharmacokinetic model [8].
ARTICLE IN PRESS 7
Imaging in Drug Development
2.2.1.2 Diffusion Weighted Imaging
DWI is a non-contrast technique that measures the microscopic mobility of water molecules, reported as ADC (Apparent Diffusion Coefficient) values. As the mobility of water molecules is hindered by various membranes in vivo, it is possible to assess the cellular density (Figure 4) [9]. In general, tumours have a higher cellular density than normal cells, whereas necrotic areas have a lower density [10]. DWI has been used in monitoring therapy response as a method of detecting early changes in the tumour microenvironment and much effort is in progress to establish it as a general response biomarker [11,12]. In some studies both DCE and DWI are used to improve the assessment of therapy response [13].
Distribution
2.2.2 Central Nervous System Drug discovery for CNS disorders is particularly challenging due to the often poorly defined pathophysiology and the subjective assessment of treatment efficacy [14]. Novel CNS drugs that enter clinical trials have a very high failure rate, and there is a clear need for biomarkers to objectively and quantifiably study the biological processes in the brain in order to better study the effect of treatments. Functional MRI (fMRI) has emerged as a very
Solid tumour
Water mobility or ⬙ADC⬙
Treatmentinduced cell death
Distribution
Effective therapy
Water mobility or ⬙ADC⬙
Figure 4 Cellular density and DWI. Adapted with permission from Wiley Publishers Ltd.: Ref. [9], © 2010.
ARTICLE IN PRESS 8
James Nairne et al.
promising tool in understanding how the various regions of the brain are involved in various diseases and it is providing a new way of screening patients and assessing treatment responses [15]. A search of www. clinicaltrials.gov indicates that there are 172 active clinical trials using fMRI as a diagnostic tool in evaluating therapeutic response. 2.2.2.1 Functional MRI
fMRI is a non-contrast technique that detects areas of the brain that are haemodynamically activated. Although it is mainly used to measure brain activity upon a sensory stimulus, it can also be used to assess the effect of pharmaceutical challenges and is sometimes called phMRI (pharmacologic MRI) [16]. There are two main techniques that are in use, bloodoxygenation-level dependent (BOLD) and arterial spin labelling (ASL). The BOLD effect utilises the different magnetic moments of oxygenated and de-oxygenated blood to indirectly measure the level of brain activity. A BOLD signal is a result of a complex interaction between changes in blood flow and blood oxygenation following an increase in neural activity (Figure 5). ASL is a non-contrast technique that can be used to measure cerebral blood flow (CBF) and, indirectly, neural activity. It is based on the magnetic labelling of blood water molecules that are flowing into a region of interest. Synapse
Astrocyte
Glutamate Glutamate
Glucose
Capillary O2
?
Glucose Glucose
· Less Hb · Slower dephasing · Stronger MRI signal
O2
· More Hb · Faster dephasing · Weaker MRI signal
Figure 5 BOLD effect. Adapted with permission from Macmillan Publishers Ltd.: Ref. [17], © 2002.
ARTICLE IN PRESS Imaging in Drug Development
9
The magnetic labelling will alter the signal compared to a non-labelled background and the amount of arterial blood delivered to the region can be measured. Recently, fMRI has been shown to be a promising tool for translational drug discovery as certain spatial and functional characteristics of brain network architectures across species have been mapped. These intrinsic networks could potentially be used as translational biomarkers in early screening for CNS drug discovery [18].
3. NUCLEAR MEDICINE IMAGING The use of radiotracers is a particularly powerful tool for the development of therapeutics. This is mainly due to the high sensitivity of the signal. Typically, radiolabelled compounds are administered intravenously in very low quantities (around 10 μg), levels below that which induces a pharmaceutical response. The radionuclides of use in nuclear medicine imaging have to fulfil certain specific criteria to minimise the exposure of the patient to the deleterious effects of ionising radiation. This is achieved by using nuclides of low energy, those with short half-lives and γ or positronemitting isotopes. Isotopes that emit α-particles are inappropriate for imaging as the α-particles are absorbed by tissue in the immediate vicinity, thus causing damage. The mode of emitting detectable radiation dictates the mode of detection of the signal. The radiation from γ emitting radionuclides is detected directly in a technique called SPECT, whereas the radiation detected from positron-emitting nuclides is detected indirectly; this technique is known as positron emission tomography (PET).
3.1. Single-Photon Emission Computed Tomography Images generated using SPECT are built up from a series of planar images. The γ-rays are captured by a camera that is rotated about the patient. The camera consists of up to three heads, each consisting of a collimator in front of a sodium iodide crystal backed with an array of photomultiplier tubes, which are used to pick up an image of the scanned area. This is reconstructed to give a 3D image of the accumulated radiotracer or radiopharmaceutical. There are three main radionuclides used in SPECT imaging (Table 1); two of these, technetium-99m and indium-111, are radiometals and the third is iodine-123. Good metal-ion chelate stability under physiological conditions is required for imaging applications. If this is not achieved,
ARTICLE IN PRESS 10
James Nairne et al.
Table 1 Common Radioisotopes Used in SPECT Isotope Production Emissions 99m
Tc
Generator
90% IsT, 10% InT
Half-life
Daughter
6h
99
Tc
111
In
Cyclotron
100% EC
2.8 days
111
123
I
Cyclotron
100% EC
13.2 h
123
Cd Te
EC, electron capture; IsT, isomeric transition (γ emission); InT, internal transition. In internal transition, the nucleus decays by transferring energy to an orbital electron which is ejected instead of the γ-ray.
imaging artefacts can be observed that can complicate image interpretation; for example, free technetium will accumulate in the thyroid gland. 3.1.1 Technetium-99m Tracers Technetium-99m is produced relatively inexpensively using a generator. Molybdenum-99 suspended on an alumina column decays (t½ ¼ 66 h) to form technetium-99m. The singly charged 99mTcO 4 is eluted in preference to the doubly charged 99 MoO4 2 using saline. Commercially available technetium-99m radiotracers are generally prepared by the simple addition of technetium-99m eluted from the generator to a kit vial containing a freeze-dried formulation of the active ingredient. The technetium-99m half-life of 6 h allows time for preparation of the radiotracer, distribution and patient imaging. The energy of the γ-ray emission (140 keV) is ideal for imaging using gamma cameras. Technetium has a rich coordination chemistry with several potential oxidation states [19]. Most nuclear imaging agents contain technetium99m in the +5, +3 or +1 oxidation states, although it is also present in the +7 oxidation state in the thyroid imaging agent 99m TcO4 , as formed in the generator. Technetium has good affinity for nitrogen, oxygen, phosphorus and sulphur in the most common oxidation states. The preparation of technetium-99m imaging agents is relatively straightforward (Scheme 1); a kit comprising a reducing agent, usually stannous chloride, a weak chelating agent and the cheland is treated with the generator eluate and the mixture incubated for a short time, often at room temperature, giving a preparation that is ready for injection without purification. Approximately 85% of nuclear medicine diagnostic imaging procedures are still done with technetium-99m. The three most widely used technetium-99m tracers in the USA are 99mTc-Sestamibi (Cardiolite) and 99m Tc-Tetrofosmin (Myoview) (Figure 6) primarily for imaging myocardial perfusion and 99mTc-MDP (methylenediphosphonate) for imaging bone
ARTICLE IN PRESS 11
Imaging in Drug Development
Sn4+
Sn2+
HO
O
O
99mTc
HO
[99mTcVOx] −
[99mTcVIIO4] −
O
O
O
O
O
OH CO2H
HO
OH OH CO2H
CH3 H3C NO N H3C CH3 Tc NO N H3C CH3 OH OH
Scheme 1 Formation of 99mTc-HMPAO (hexamethylpropyleneamine oxime).
H3C
CH3
H3CO CH3
H3CO
CH3 H3 CO
N N
N
OEt O
EtO P
CH3
99m
Tc
H3C
OCH3 H3C
P
EtO
N N
OEt
P Tc P
99m
H3C N
OEt
CH3
OCH3 CH3
OEt
O OEt
OEt
OCH3 H3C CH3
99m
99m
Tc-Sestamibi
Figure 6 Structures of
99m
Tc-Sestamibi and
99m
Tc-Tetrofosmin
Tc-Tetrofosmin.
metastases. The delivery of these tracers to the organ of interest is based on a non-target specific localisation mechanism. 99m Tc-Sestamibi and 99mTc-Tetrofosmin are also both transport substrates recognised by the multidrug resistance (MDR) P-glycoprotein (Pgp). Pgp can confer resistance to many cytotoxic cancer therapeutics. 99m Tc-Sestamibi has been used in cancer clinical trials to predict the response to chemotherapy. In a recent meta-analysis, lung cancer patients who had less 99mTc-Sestamibi initial uptake in tumours were found to be less likely to respond to chemotherapy. The sensitivity, specificity and
ARTICLE IN PRESS 12
James Nairne et al.
accuracy of 99mTc-Sestamibi in identifying chemotherapy responders were 94%, 90% and 92%, respectively [20]. 3.1.2 Indium-111 Tracers Indium-111 is produced in a cyclotron using a cadmium-112 target which is bombarded with protons to produce indium-111 by the (p,2n) reaction. The energies of the γ-ray emissions (171 and 245 keV) are higher than that of technetium-99m. Indium-111 forms complexes in which indium is present in the +3 oxidation state. Either open-chain DTPA or macrocyclic DOTA chelating agents are generally used in indium-111 nuclear medicine applications (Figure 7). In contrast to indium-DTPA which can be formed at room temperature, heat is required to form indium-DOTA complexes [21]. As a result, indium-DOTA complexes may not be suitable for applications in which the peptide or antibody is sensitive to heat. The half-life of indium-111 (2.8 days) is especially suited to imaging antibodies which tend to have longer biological half-lives. 3.1.3 Iodine-123 Tracers Iodine-123 is produced in a cyclotron by bombardment of enriched xenon124 by the (p,2n) reaction via the shorter lived caesium-123 and xenon-123. The energy of the γ-ray emission (159 keV) is close to the ideal for imaging using gamma cameras. Several different iodination methods are employed to synthesise radiolabelled iodine-123 compounds. The most commonly used involve either O O O N O O
N
111In
O
N
N O O
O O
N
N
N
O O
O
111In
O O O
111In-DTPA
111In-DOTA
Figure 7 Structures of indium-DTPA and -DOTA complexes.
ARTICLE IN PRESS 13
Imaging in Drug Development
direct electrophilic substitution on activated aromatic systems, electrophilic radio-iododestannylation reactions or nucleophilic exchange [22]. Direct electrophilic substitution of protein or peptide is carried out using iodogen, an oxidant that converts [123I]iodide to [123I]iodonium, which then reacts with an electron-rich amino acid residue such as tyrosine [23]. 123 I-FP-CIT (DaTSCAN, 123I-2-β-carbomethoxy-3β-(4-iodophenyl)N-(3-fluoropropyl)nortropane) is used in patients with clinically uncertain Parkinsonian syndromes and in patients to help differentiate probable dementia with Lewy bodies from Alzheimer’s disease. It is one of the few commercially available iodine-123 radiopharmaceuticals and is prepared by radio-iododestannylation (Scheme 2). The combination of [123I]iodide, part labelled elemental iodine and the hydrogen peroxide oxidising agent, produces the electrophilic species HO*I and H2O*I which then react rapidly with the trimethylstannyl precursor at room temperature. The 13.2 h half-life of iodine-123 allows central manufacturing and a relatively wide distribution area. 123 I-BMIPP ([123I]-beta-methyl-p-iodophenylpentadecanoic acid (Cardiodine/Japan)) is another commercially available radioiodinated tracer which is currently being used as a metabolic imaging agent in drug therapy clinical trials. Long-chain fatty acids such as BMIPP are the main energy source for normal oxidised myocardium and are rapidly metabolised by β-oxidation. With ischaemia, energy metabolism moves to anaerobic metabolism and the main energy substrate changes from fatty acids to glucose. Reduced uptake of 123I-BMIPP can indicate a history of previous ischaemia. This is due to the fact that the recovery of oxidative metabolism of fatty acids within cardiomyocytes lags behind the recovery of perfusion. 123 I-BMIPP is synthesised by nucleophilic exchange of iodine-127 in the presence of a catalytic amount of copper and ascorbic acid as reductant (Scheme 3).
F
F
O
O N
OCH3
N SnMe3
Scheme 2 Preparation of
OCH3
[123I]-iodide
123
I-FP-CIT.
123I
H2O2 / H2SO4
ARTICLE IN PRESS 14
James Nairne et al.
I OH CH3 O
[123I]-iodide Cu(NO3)2 / ascorbic acid
123I
OH CH3 O Scheme 3 Preparation of
123
I-BMIPP.
3.1.4 Applications of SPECT to Translational Medicine The number of applications of technetium-99m radiotracers in clinical drug development is likely to increase in the future as more target-specific technetium radiotracers are developed. One recent example involved the use of a 99m Tc-labelled folate imaging agent 99mTc-Etarfolatide in the clinical trials of the related drug Vintafolide [24] (Figure 8). In 99mTc-Etarfolatide, 99mTc is stabilised by chelation to a peptide-based N3S chelate which forms a negatively charged complex in which 99mTc is present in the +5 oxidation state. The formation of this hydrophilic complex facilitates the desired rapid clearance through the kidneys. Vintafolide is under development for the treatment of cancers of the ovary, lung and breast among others. 99mTc-Etarfolatide imaging has primarily been used in clinical trials to identify patients expressing high levels of the folate receptor who may be suitable for Vintafolide therapy. The longer half-life of indium-111 makes it suitable for use with antibody-based therapies. 111In-ibritumomab tiuxetan has been used to test the mechanistic hypothesis behind the combination of radiotherapy using 90 Y-iritumomab tiuxetan (Zevalin) and oligonucleotide therapy using CpG 7909 in patients with relapsed indolent non-Hodgkin lymphoma [25]. It had been proposed that CpG 7909 therapy could enhance the delivery of the Zevalin as in vitro experiments had shown increased CD20 expression by malignant B cells. However, no increase in uptake of the indium-111
ARTICLE IN PRESS 15
Imaging in Drug Development
O HN
O N NH
N H
HO2C O
N
NH2
N
HN O NH N HO2C 99mTc=O
N
O
S HO2C 99m
Tc-Etarfolatide
O HN
O N
HO2C
N H
NH
O NH2
N
N
O HN NH O HO2C HN
CO2H H2N
NH NH
O NH
CO2H
O
O HN
S S
NH
O
CO2H
HN
O OH
HO
CH3 N
H3C OCH3 HN O H3CO H3C
Vintafolide
Figure 8 Structures of
99m
Tc-Etarfolatide and Vintafolide.
H N HO
ARTICLE IN PRESS 16
James Nairne et al.
analogue of Zevalin was observed, indicating that there was no enhanced delivery of the radiotherapeutic. 123 I-FP-CIT imaging has been used in a number of clinical trials of disease modifying drugs. A recent study used 123I-FP-CIT imaging to show that the dopamine agonist pramipexole had no modifying properties in patients with early Parkinson’s disease [26]. Baseline 123I-FP-CIT scans were carried out before therapy and at 15 months following treatment with pramipexole. A decrease in striatal dopamine transporter binding observed by imaging at 15 months indicated that pramipexole was not effective in slowing disease progression. A trial in Japan has been set up to determine the efficacy of the Na–H exchange (NHE) inhibitor TY-51924 as an adjunctive therapy to primary percutaneous coronary intervention (pPCI) for patients with ST-elevation myocardial infarction (STEMI) [27]. The trial illustrates the important role radiotracers can play in the clinic. In the acute phase (3–5 days after pPCI), thallium-201 SPECT imaging will be used to determine infarct size, and left ventricular (LV) volume and function, while 123IBMIPP will be used to determine the area at risk. In the follow-up, 3 months after pPCI, 99mTc-Tetrofosmin will be used to determine infarct size and LV volume and function. 201Tl chloride and 99mTc-Tetrofosmin are both myocardial perfusion imaging agents. GlaxoSmithKline (GSK) have used [123I]iodobenzamide and [123I]R91150 123 ( I-4-amino-N-[1-[3-(4-fluorophenoxy)propyl]-4-methyl-4-piperidinyl]5iodo-2-methoxybenzamide) in the study of SB-773812, a putative antipsychotic treatment. The two SPECT tracers bind D2 receptors and [123I] R91150 also binds to 5HT2A[28]. The clinical study was able to show that not only did SB-773812 cross the blood–brain barrier, but it also bound the two receptors in humans. The study contributed towards the decision to proceed with clinical development into Phase III.
3.2. Positron Emission Tomography Positrons are particles that have the same mass as an electron, but are positively charged and as such are very unstable. They have a very short half-life in tissue as they interact with electrons in a process called annihilation, in which the mass of the two particles is converted into energy in the form of two photons, with an energy of 511 keV, emitted in the opposite directions. It is these two photons that are detected in a PET camera. The major advantage of the use of PET over SPECT is that the technique can be used to quantify the uptake of a particular tracer. This is
ARTICLE IN PRESS 17
Imaging in Drug Development
possible due to the simultaneous emission of the two photons during annihilation. The detectors in the camera are arranged in a ring around the patient and only signals that arrive at the same time are counted in image accumulation. It is, therefore, certain that the signal is accrued from single emission events and that each event has occurred on a straight line between the two detectors that have detected the signal Most of the common PET radioisotopes have shorter half-lives that those used in SPECT (Table 2). Nitrogen-13 and oxygen-15 are used clinically in the form of [13N]ammonia and [15O]water as blood perfusion agents. However, the most widely used nuclides are carbon-11 and fluorine-18. These are attractive isotopes in drug development as they can form covalent bonds to carbon, allowing them to be incorporated into analogues of drug-like molecules which then to behave in the same way as the drug when introduced into the body. 3.2.1 Carbon-11 Tracers The half-life of carbon-11 is only 20 min. This, therefore, necessitates two prerequisites for the radiochemist. First is in the design of the radiotracer; the radiolabel should be designed to be introduced at the latest possible stage in a synthesis. The second is that the chemistry used needs to be rapid and efficient. Carbon-11 is generated in a cyclotron by proton bombardment of nitrogen gas with ejection of an α-particle from the nucleus (14N2 (p,a)11C). Incorporation of a small amount of oxygen in the target gives rise to carbon-11 as carbon dioxide, while if around 4% hydrogen is mixed with the nitrogen, carbon-11 methane is obtained. These two compounds Table 2 Common PET Radionuclides and Their Properties Isotope Production Emissions Half-life 11 13
C N
Cyclotron
100% β +
Cyclotron
100% β +
Daughter
20.3 min
11
B
10 min
13
C N
15
O
Cyclotron
100% β +
2.1 min
15
18
F
Cyclotron
100% β +
109.8 min
18
O
Generator
89% β +, 11% EC
68 min
68
Zn
68
Ga
89
Zr
Cyclotron
76.6% EC, 23% β +
78.4 h
89m
64
Cu
Cyclotron
44% EC, 38% β , 18% β +
12.7 h
64
Y,
Ni,
89 64
Y
Zn
ARTICLE IN PRESS 18
James Nairne et al.
provide the basic feedstocks for introduction of carbon-11 atoms into compounds. [11C]Carbon dioxide can be used directly in reactions to incorporate the radiolabel. The radiosynthesis of [11C]WAY100635 (Scheme 4), a 5HT1A receptor ligand [29], and the dopamine D2/D3 binder [11C] PHNO [30] (Scheme 5) was prepared by trapping Grignard reagents with [11C]carbon dioxide, conversion to the corresponding acid chlorides and reaction with amines. Over the years, radiochemists have been able to develop a number of chemical approaches that fulfil the criterion of rapid incorporation of the label. [11C]Carbon dioxide has been converted to [11C]methanol by treatment with lithium aluminium hydride [31] and subsequent treatment with hydriodic acid [32] to form [11C]methyl iodide. This synthon is one of the most widely used methylating agents for introducing labelled methyl groups in nucleophilic methylation reactions (Scheme 6). It has been used in the preparation of methyl ethers, as in the adenosine A2A ligand [11C]KW6002 [33], N-methylated compounds such as the benzodiazepine receptor binding compound, [11C]flumazenil [34], and S-methylated compounds
MgCl [11CO2]
O11 OMgCl C SOCl2
O11 Cl C
11C
O
N
N
OCH3
N
N
[11C]WAY100635 11
Scheme 4 Radiosynthesis of [ C]WAY100635.
CH3CH2MgBr
[11CO2]
CH3CH211CO2MgBr
(i) PDC (ii) DTBP
CH3CH211COCl
O N
11CH CH CH 2 2 3
O [11C]PHNO PDC–phthaloyl dichloride; DTBP–2,6-di-tert-butylpyridine
Scheme 5 Radiosynthesis of [11C]PHNO.
N O
11C
CH2CH3
ARTICLE IN PRESS 19
Imaging in Drug Development
O Et O
N N Et
CH3 N
OCH3
N
N
O 11CH 3
[11C]KW-6002
N
O OEt
N
F O
11CH 3
[11C]flumazenil
[11C]CO2
[11C]CH3I
[11C]CH3OH
CO2H H311C
NH2
[11C]methionine
O H311C
S
NH N
O
HO O F OH [11C]FMAU
Scheme 6 Some applications of [11C]methyl iodide in the radiosyntheses of important carbon-11 labelled radiotracers.
in the preparation of [11C]methionine [35], used for imaging tumours. [11C] Methyl iodide has also been used as a substrate in Stille [36], for the preparation of 1-(20 -deoxy-20 -β-D-arabinofuranosyl)-[methyl-11C]thymine ([11C]FMAU), and Suzuki [37] coupling reactions and as a substrate for the preparation of [11C]methylmagnesium iodide [38]. [11C]Methyl triflate has also been used as a methylating agent. This is prepared by treatment [11C]methyl iodide with silver triflate. [11C]Methyl triflate is then used as a very reactive electrophile [39] in similar reactions to [11C]methyl iodide. For example, Langer et al. have prepared [11C] raclopride (Scheme 7) using this approach [40]. [11C]Carbon dioxide can be reduced to [11C]carbon monoxide by reduction over zinc [41] or molybdenum [42] and the resultant gas used in carbonylation reactions using transition metals such as palladium and rhodium. These synthetic routes have been used to prepare malonates [43], ureas [44], carbamates [45], ketones [46,47] and amides [48] among other compound classes.
ARTICLE IN PRESS 20
James Nairne et al.
H311C OH Cl
O
Et N
N H OH
O
O
Cl
11CH OSO CF 2 3 3
aq NaOH acetone, RT
Et N
N H OH Cl
Cl
[11C]raclopride 11
11
Scheme 7 Radiosynthesis of [ C]raclopride using [ C]methyl triflate.
F
Br N
F
11CN
K11CN
N
N N O
F
Pd(PPh3)4 DMSO
N N O
F
11
Scheme 8 Radiosynthesis of [ C]AZD9272.
Cu11CN 11
I
CN
DMF N
N
Scheme 9 Radiosynthesis of [11C]LY223645.
[11C]Methane is also a valuable cyclotron product used for introducing a carbon-11 label into compounds. It has been utilised to prepare [11C]methyl iodide by treatment with iodine [49]; [11C]hydrogen cyanide via a platinum-catalysed reaction with ammonia [50]; and [11C]phosgene via perchlorination then hydrolysis of the resultant [11C]carbon tetrachloride [51,52]. The latter two carbon-11 reagents have been used to make a variety of tracers. [11C]Hydrogen cyanide is used to introduce a [11C]cyano group using a number of different chemistries, including transition metal-catalysed cyanations, such as used by Andersson et al. in the radiosynthesis of the mGluR5 radioligand [11C]AZD9272 [53] (Scheme 8), and copper cyanide reactions such as that used by Mathews et al. to prepare [11C]LY2232645 (Scheme 9), also an mGluR5 antagonist [54]. 3.2.2 Fluorine-18 Tracers Fluorine-18 has a half-life of 109.8 min, some five-and-a-half times that of carbon-11. This provides the isotope with several advantages over the use of carbon-11 tracers; the greater shelf-life of the tracer once prepared can
ARTICLE IN PRESS 21
Imaging in Drug Development
translate as the ability either to image more subjects from a single radiosynthesis or to allow imaging of patients to occur at sites remote from the production of the tracer. Fluorine-18 is also produced by a reaction in a cyclotron; in this case, proton bombardment of oxygen-18. In the case of [18F]fluorine gas, the target is [18O]oxygen gas and for [18F]fluoride the target is [18O]water. The methods for introducing fluorine-18 into a molecule are more limited than is the case for carbon-11, but the same principle for introducing the isotope as late as possible still applies. Much of the early work with fluorine18 used electrophilic reactions. These involved the use of [18F]fluorine gas directly. As fluorine gas is very reactive, the substrates that could be used were limited and the chemoselectivity of the introduction of the label is low. An example of this is the synthesis of [18F]6-F-dopa, a tracer for the quantification of dopamine receptors. The reaction is only 59% regioselective [55], the remaining 41% of radiofluorinated product being a mixture of the 2- and 5-fluoro analogues (Scheme 10). Other electrophilic reagents have been developed are [18F]acetyl hypofluorite, which has been used to prepare [18F]fluorodeoxyglucose (FDG) in its early radiosyntheses [56] (Scheme 11), and N-fluorobenzenesulfonamide [57]. The nuclear chemistry to produce [18F]fluoride is much more straightforward—bombardment of [18O] water with protons gives [18F] fluoride that can be used as a nucleophile in radiofluoridation reactions. HO
CO2H 18F
HO
NH2
[18F]6-F-dopa HO HO
CO2H NH2
[18F]F2/HF
HO
CO2H NH2
HO 18F
[18F]-5F-dopa 18F
HO HO
CO2H NH2 [18F]-2F-dopa
Scheme 10 Electrophilic radiosynthesis of [18F]6-F-dopa.
ARTICLE IN PRESS 22
James Nairne et al.
(i) [18F]CH3COOF (ii) HCl
O
HO
O
HO
F
HO
HO
OH
OH
OH
Scheme 11 Electrophilic radiosynthesis of [18F]FDG.
AcO
O
AcO
18
OAc
(i) [ F]KF, K222
OTf
(ii) HCl
OAc
HO
O
OH 18
HO
F
OH
Scheme 12 Nucleophilic radiosynthesis of [18F]FDG.
As fluoride is a relatively poor nucleophile, it needs to be processed to improve its ability to participate in SN2 reactions. [18F]Fluoride is trapped on an ion exchange cartridge, the [18O]water recovered for reuse and the [18F]fluoride eluted with a mixture of potassium carbonate and kryptofix222 (K222). This gives a solution that, once dried, gives anhydrous fluoride that is rendered more reactive by the sequestration of the potassium ion by the kryptofix-222 [58]. In this form, the fluoride can react in both SN2 and SNAr reactions used in the preparation of [18F]fluoroalkanes and [18F] fluoroarenes, respectively. 3.2.2.1 Preparation of [18F]Fluoroalkanes
[18F]Fluoroalkanes can be prepared from precursors such as alkyl tosylates, mesylates and triflates in solvents such as acetonitrile, DMF and DMSO. The basicity of fluoride means that use of protecting groups is often required to prevent side reactions or suppress [18F]hydrogen fluoride formation by proton abstraction. This chemistry has been exploited widely in the preparation of radiotracers for applications in neurology and oncology. The most widely used PET radiopharmaceutical is [18F]FDG, now prepared via the nucleophilic substitution of a triflate group in mannose triflate tetraacetate [59] (Scheme 12). [18F]FDG is a marker for glucose hypometabolism—it is taken into cells via the glucose transporter mechanism and then undergoes phosphorylation on the 6-hydroxyl function, but cannot undergo the second step of glucose metabolism—phosphorylation at the 2-position—as this is blocked by the fluorine-18 atom (Scheme 13). The phosphorylated derivative is then trapped in the cell, leading to an accumulation of the monophosphorylated
ARTICLE IN PRESS 23
Imaging in Drug Development
HO
-
18
HO
O P O O
O
OH
O
hexokinase
F
OH
O
18
HO
OH
F
OH
Scheme 13 Metabolism of [18F]FDG. 18F
O
O 18F
N
N H OCH3
N
N
N
OCH3 N
OCH3 [18F]Fallypride
H3C
[18F]MeFWAY
H N 18F
N
O
O
O
18F
[18F]Florbetapir 18
CO2H NH2
[18F]Fluciclovine 18
18
Figure 9 Structures of [ F]fallypride, [ F]MeFWAY, [ F]florbetapir and [18F] fluciclovine.
[18F]FDG that is detected by the PET camera [60]. The greater the glucose demand of the cells in question, the greater the radioactive signal. This tracer has, therefore, found particular use in oncology, as well as cardiology and neurology [61]. Many tracers have been developed using this type of chemistry via direct labelling methods. These include analogues of carbon-11 tracers such as the dopamine D2/D3 binder fallypride [62], an analogue of raclopride, and the 5HT1A tracer MeFWAY [63], an analogue of WAY100635; and tracers that have been approved for clinical use. This latter group includes florbetapir [64], a radiopharmaceutical for the detection of amyloid plaques, and others undergoing development, for example, fluciclovine (FACBC) [65], an L-amino acid transporter marker that has potential applications in prostate cancer diagnosis (Figure 9). In all of the above cases, the fluorine is introduced by a direct method, that is, one in which a pharmacophore containing a leaving group is treated
ARTICLE IN PRESS 24
James Nairne et al.
with [18F]fluoride. Some compounds contain functionality that is sensitive to the conditions for reaction with fluoride or contain functionality that prevent fluoride acting as a nucleophile. In these cases, a so-called indirect method of fluorination can be used. The fluorine is introduced to a small molecule that undergoes a reaction with the pharmacophore under mild conditions. Examples of this approach are demonstrated by the synthesis of [18F]fluorocholine [66] and [18F]fluoroethyltyrosine [67], both used in oncology, and [18F]GE-179 [68], a marker for activated NMDA receptors. Radiosynthesis of [18F]fluorocholine is carried out via the synthesis of [18F] fluorobromomethane from dibromomethane (Scheme 14) and the radiosyntheses of [18F]fluoroethyltyrosine and [18F]GE-179 via the synthesis of [18F]fluoroethyl tosylate from ethylene ditosylate (Scheme 15). 3.2.2.2 Preparation of [18F]Fluoroarenes
Fluorination of aromatic rings with [18F]fluoride follows the same rules as those for preparative SNAr reactions in that, generally, there is a requirement CH3 H3 C Br
18F
Br
N
OH 18F
Br
CH3 N
H 3C
OH
Scheme 14 Radiosynthesis of [18F]fluorocholine using [18F]bromofluoromethane.
TsO
OTs
CH3 N
H N
18
NH Cl
F
CO2H
OTs HO
SH H N
NH2
CH3 N CO2H
NH Cl
18
S 18
F
O
NH2
F [18F] fluoroethyltyrosine
[18F] GE-179 18
18
Scheme 15 Radiosyntheses of [ F]GE-179 and [ F]fluoroethyltyrosine.
ARTICLE IN PRESS 25
Imaging in Drug Development
for an electron-withdrawing group capable of delocalising the incoming negative charge. For simple direct labelling reactions, therefore, there needs to be suitable functionality in the ortho- or para-position to the [18F]fluoride label. Leaving groups are typical for the types of reaction—nitro, trimethylammonium and halides have all been used, though nitro and trimethylammonium are preferred due to ease of separation of the product from the excess of precursor. The two dopamine tracers [18F]haloperidol [69] (Scheme 16) and [18F]spiperone [70] (Scheme 17) have been prepared in one step from their nitro precursors. There are some examples where the standard SNAr-activating groups are not present, so particularly stringent reaction conditions are required to effect the radiosyntheses; [18F]flutemetamol [71] (the fluorinated equivalent of the carbon-11 amyloid tracer [11C]PiB [72]) and [18F]flumazenil (a GABA tracer) [73] have been prepared from [18F]fluoride (Figure 10).
OH
OH
O
O Cl
Cl
O2N
18
F
[18F]Haloperidol 18
Scheme 16 Radiosynthesis of [ F]haloperidol.
O O
H N
O O
N
H N N
N
N 18F
O2N
18
[ F]Spiperone
Scheme 17 Radiosynthesis of [18F]spiperone. N
18F
HO
HO
S N
NH CH3
S N
18F
N O
[18F]Flutemetamol
[11C]PiB
CO2Et
N
NH 11CH 3
CH3
[18F]Flumazenil
Figure 10 Structures of [18F]flutemetamol, [11C]PiB and [18F]flumazenil.
ARTICLE IN PRESS 26
James Nairne et al.
The constraints placed on the electronic requirements of SNAr reactions have led to a number of different approaches to introduce fluorine-18 into aromatic systems. Initial attempts were to redesign syntheses so that the radiochemical precursor was activated to nucleophilic attack, and then carry out subsequent chemistry to provide the tracer required. Much of this effort was aimed at radiosyntheses of the dopamine transporter tracer [18F] 6-F-dopa. Lemaire et al. reported a four-step radiosynthesis of this tracer in which the fluoride was introduced via nucleophilic displacement of a trimethylammonium salt derived from veratraldehyde. Subsequent reduction, iodination, asymmetric alkylation and deprotection gave [18F]6-F-dopa [74] (Scheme 18). The Machulla group developed a similar nucleophilic method using a veratraldehyde [75]. Although it was not used for the synthesis of [18F] 6-F-dopa, the same group developed a method for the introduction of [18F] fluoride by incorporation of a formyl group acting as a transient activating group that was removed using Wilkinson’s catalyst [76] (Scheme 19). A further strategy towards [18F]6-F-dopa was reported by Coenen who demonstrated incorporation of fluoride in the 6-position via activation through a ketone in the 3-position. The ketone was subsequently converted to the required phenol via a Baeyer–Villiger oxidation reaction [77]. Each of these syntheses has the disadvantage of a large number of steps with the fluorine-18 in the molecule leading to low radiochemical yields of tracer. A more direct approach has been shown in the use of iodonium salts by Carroll and DiMagno. Carroll showed that it was possible to access 3-fluoropyridine using this technology in 55–63% radiochemical yields (Scheme 20) [78]. Hitherto, the yields of this compound using fluoride were significantly worse than for the activated 2- and 4-fluoro analogues [79].
H3CO
CHO
H3CO
H3CO
R
H3CO
CHO
HO
18F
HO
CO2H 18
F
NH2
Scheme 18 Outline of radiosynthesis of [18F]F-dopa [73]. H3CO
CH3
H3CO
H3CO
F
H3CO
CHO
CH3 18
F
H3CO H3CO
CH3 18
F
CHO
Scheme 19 Use of a formyl group to activate an aromatic ring towards SNAr using [18F] fluoride [75].
ARTICLE IN PRESS 27
Imaging in Drug Development
Although the use of iodonium salts shows promise for simple molecule and synthons [80], the chemistry used to prepare them is incompatible with functionality that is sensitive to oxidation or strong acid. Recent years have seen the extension of transition metal chemistry for the introduction of fluoride. Initial work by the Buchwald group showed that the introduction of 19F fluoride was possible using palladium chemistry [81]. The use of palladium-catalysed chemistry to introduce [18F]fluoride into unactivated positions was reported by Ritter [82]. A fluorine-18 palladium(IV) complex is prepared from the 4-methylpyridine precursor (Scheme 21), and it is this that is used to react with palladium(II) complexes of the arene that is to be fluorinated to give the desired [18F]fluoroarene. This chemistry was illustrated by the syntheses of [18F]fluorodeoxyestrone and [18F]AS-252424 (Figure 11). Ritter recognised the need for further simplification of his method and followed up by publishing on the fluorination of nickel(II) complexes in aqueous solutions [83] in the presence of an oxidant. These single-step radiosyntheses also remove the need for the time-consuming drying step. 18
I+ N
OCH3
F
N
18
Scheme 20 Radiosynthesis of [ F]3-fluoropyridine using an iodonium salt. CH3
18
F
N N
N N Pd
N
N
B N N N N N
Pd N
N N N B N N N N
Scheme 21 Synthesis of [18F]-containing palladium(IV) complex for preparation of [18F] fluoroarenes [81]. CH3O
O NH
H H 18F
S
OH H
18F
O O
Figure 11 Structures of [18F]fluorodeoxyestrone and [18F]AS-252424.
ARTICLE IN PRESS 28
James Nairne et al.
The utility of the method was exemplified by the formal syntheses of [18F]6F-dopa as well as [18F]fluorodeoxyestrone. In a further development in the flexibility of preparation of [18F]fluoroarenes, Tredwell et al. have recently published a paper on the use of copper(II)-mediated chemistry to introduce [18F]fluorine into aromatic systems via boronic esters [84] (Scheme 22). This methodology has the benefits of well-established chemistry for the preparation of the precursors and relatively mild reaction conditions for the fluorination reaction itself. The method was illustrated with the radiosyntheses of [18F]DAA1106, a tracer for translocator protein (TSPO), [18F]6fluorotyrosine and [18F]6-F-dopa. 3.2.2.3 Labelling of Peptides and Proteins with Fluorine-18
All the methods discussed earlier are applicable to the preparation of small molecules, but outside neurology, there is an increasing emphasis on the use of biopharmaceuticals, consisting of peptide or proteins. The requirements of most of the methods are for anhydrous conditions or often the absence of acidic protons to allow fluoride to be introduced. Moreover, site-selective chemistry for peptides and proteins has long been a challenge for molecular biologists. The radiolabelling of radiotracers for in vivo use presents an additional challenge due to the stoichiometry of the radiochemistry reactions. A considerable amount of work has been put into realising methodologies for the introduction of [18F]fluorine to this type of molecule. One of the advantages of dealing with high molecular weight pharmacophores is that the addition of extra functional groups can be done with little effect on the binding properties and pharmacokinetics, provided that the group is added away from the molecule’s binding site. This strategy has been broadly exploited. The groups added are often called “synthons” or “prosthetic groups” and are designed to react with functional groups not found in native amino acid residues. By definition, these strategies are two-step H3CO
H3CO
O H3C
OCH3 N
18
OPh O
B O
[Cu(OTf)2(py)4] [ F]KF/K222 DMF, 110 °C, 20 min
O H3C
OCH3 N OPh
18F
Scheme 22 Radiosynthesis of [18F]DAA1106 using copper(II)-mediated chemistry.
ARTICLE IN PRESS 29
Imaging in Drug Development
processes—the synthons must be readily radiolabelled with [18F]fluoride and then undergo rapid chemoselective reactions to give the labelled macromolecule (Scheme 23). The most commonly used synthon is [18F]4-fluorobenzaldehyde. The conjugation chemistry to add this to proteins and peptides is through the reaction of the aldehyde with either an aminoxy or hydrazine group. The aminoxy group can be introduced by the reaction of an amine in the peptide or protein with aminooxyacetic acid, as exemplified by the αvβ3 integrin binding peptide, [18F]fluciclatide [85] (Figure 12), and the hydrazine group can be introduced using the HYNIC group, for example, the LTB4 antagonist, MB67 [86] (Figure 13). This strategy works well for peptides that can be synthesised, as the introduction of the reactive group can be done in a site-specific manner before deprotection of the amino acid side chains. More complex molecules such as the HER-2-binding Affibody ZHER2:2891 can be labelled via cysteine–maleimide conjugates [87] (Figure 14). The 4-fluorobenzaldehyde group is a relatively lipophilic synthon; the O’Hagan group has developed a simple, more hydrophilic equivalent in a synthon derived from ribose, 5-deoxy-5-fluororibofuranose (FDR) (Scheme 24). FDR has an advantage over FDG in that the conjugate is Peptide-ONH 2 CHO
18 -
CHO
F
Me3N
18
TfO-
N 18
F
O
Peptide
F
Scheme 23 Outline scheme for labelling peptides and proteins using [18F]4fluorobenzaldehyde as a synthon. 18
F
N O HN O
NH2
NH NH
H N
HN O
O
O N H S
H N O
O N H
CO2H H N O
Ph
O N H
S
H N O S
Figure 12 Structure of [18F]fluciclatide.
O
O N H
O
O
O
N H
O O
NH2
ARTICLE IN PRESS 30
James Nairne et al.
N
O
N N N N H3C CH3
O
O
O
O
N H
O
NH
NaO3S HN O O
SO3Na
NH
NaO3S HN O
SO3Na N NH
HN O
18
F
H N
N O
O
NH O
SO3Na
NaO3S HN O
NH O
N
O
SO3Na
NaO3S HN
N N N N
O
H3C CH 3
O N H
O
O
NH
O
18
Figure 13 Structure of [ F]MB67. 18F
H N
O
N O
N S
O O CO2H
ZHER2:2891
NH
Figure 14 Structure of [18F]-labelled HER-2 affibody.
18
F HO
O
OH
O OH OH
O
N
18
F
N OH
glutathione S
O
Scheme 24 Reaction of [18F]FDR with aminoxy-tagged glutathione.
OH
ARTICLE IN PRESS 31
Imaging in Drug Development
formed more rapidly than the corresponding FDG equivalent and that it exists predominantly in the open-chain form [88]. Another method for introducing fluorine-18 via a synthon approach has been to use the Huisgen 1,3-dipolar cycloaddition reaction, also known as Click chemistry. Usually, the peptide or protein is derivatised with a terminal acetylene then treated with an [18F]ω-fluoroalkyl azide which reacts to give the target molecule tagged with a [18F]fluoroalkyl-1,2,3-triazole. This has been demonstrated by the Sutcliffe group in the preparation of a labelled version of A20FDMV, a binder to αvβ6-expressing tumours [89]. Ramenda et al. also used click chemistry to label human serum albumin (HSA), but in this case, they prepared a fluorine-18 substituted alkyne and reacted it with azide-functionalised HSA [90] (Figure 15). There have been successful attempts to label peptides and proteins directly with fluoride. Wa¨ngler et al. developed a silicon-based prosthetic group that reacts with [18F]fluoride to give a compound with a hydrolytically stable silicon–fluorine bond [91] (Figure 16). The second recent development has been reported by D’Souza et al. who used a chelated aluminium ion linked to a targeting agent and then carried out a ligand exchange reaction with [18F]fluoride [92] (Scheme 25). This chemistry has recently been reported as being used to prepare the integrin imaging agent [18F]-alfatide (Figure 17) for use in lung cancer patients [93]. 3.2.3 Iodine-124 Tracers Iodine-124 is produced in a cyclotron by bombardment of enriched tellurium-124 either by protons in a (p,n) reaction or by deuterons in a O N N
O
O
S
N N H
N
A20FMDV2 18
F
H3C
18
F
Figure 15 Structures of [18F]-labelled A20FDMV and the [18F]-labelled alkyne synthon developed by Ramenda.
O N t-Bu 18
F
O
N H
Aux--Peptide
Si t-Bu
Figure 16 General structure of peptide labelled with a stable [18F]fluoride–silicon bond.
ARTICLE IN PRESS 32
James Nairne et al.
O HN
O HO
N
HO
N
D -Lys(HSG)- D -Tyr- D -Lys(HSG)-NH2
N
O
AlCl3
O HN
O O O
D -Lys(HSG)- D -Tyr- D -Lys(HSG)-NH2
N
Al N N
Na18F
OH O
O HN
O O O
D -Lys(HSG)-D -Tyr- D -Lys(HSG)-NH2
N
Al N N 18F
O
Scheme 25 Preparation of [18F]-labelled aluminium conjugate.
(d,2n) reaction. The imaging characteristics of iodine-124 are not ideal as it has a complex decay scheme with many high energy γ emissions and positron emissions: only 23% of its decay leads to positron emissions [94]. However, data correction with modern PET scanners can solve most of the associated imaging problems. The half-life of 4.2 days allows imaging studies at longer timepoints necessary for tracers with slow pharmacokinetics such as antibodies. The strategies for synthesis of iodine-124-labelled compounds are the same as for iodine-123 tracers (vide supra). The utility of iodine-124 has recently been demonstrated in a study of colorectal cancer patients in which an 124I-labelled humanised anti-A33 antibody (hUA33) was used to determine the treatment strategy [95].
ARTICLE IN PRESS 33
Imaging in Drug Development
O
18F
N N Al N
O O O
S NH NH O O NH2 HN O OH
NH O
O
HN
O HN
O
HN
NH
NH
O
O
O
NH O HO2C HN
HN O
O HN
HO
HN O
NH
O NH HO2C
NH
HN O
HN NH2
Figure 17 Structure of [18F]-alfatide.
[124I]huA33 is produced by direct electrophilic substitution on the tyrosine residues in the antibody. Na[124I] is added to a tube coated with the oxidant iodogen (1,3,4,6-tetrachloro-3α,6α-diphenylglycouril) followed by addition of a solution containing hUA33. The reaction is terminated by removal of the reaction mixture from the tube. Damage to the antibody is limited due to the poor water solubility of iodogen. PET/CT scans were carried out 1 week after injection and just before surgical removal of tumour. A linear relationship was observed between tissue A33 antigen and uptake of [124I]huA33 in both tumour and normal colon. The amount of [124I]huA33 in A33 antigen-positive colon tumour tissue was three times the amount in the average colon tissue section. As the A33 antigen-positive tumour regions were typically 1–2 mm in dimension, it was concluded that a lower energy/shorter range β emitter would be more suitable for radiotherapy. Due to the persistent tumour retention of [124I]huA33 and uptake in normal gut, a multistep approach to therapy
ARTICLE IN PRESS 34
James Nairne et al.
was suggested. This could involve initial administration of a modified antibody construct with specificity for a small molecule. Injection of the smallmolecule radiotherapeutic would then take place after clearance of the antibody construct from non-tumour sites. 3.2.4 Zirconium-89 Tracers The most widely used cyclotron production method of zirconium-89 is from yttrium-89 using a (p,n) reaction. The cost of zirconium-89 is relatively low for a cyclotron produced isotope as the 100% naturally abundant yttrium-89 target requires no enrichment and so is commercially available. Although zirconium-89 has a relatively low probability of positron emission (23%), good-quality images can be obtained because of its relatively low energy (397 keV). Zirconium-89 forms complexes in which zirconium is present in the +4 oxidation state. Desferrioxamine (DFO) is the most widely used chelating agent (Figure 18) and the number of studies has increased significantly since bifunctional DFO-chelating agents became commercially available [21]. No zirconium-89 molecular imaging agent is commercially available although there are an increasing number of published preclinical and clinical studies with 89Zr-labelled radiotracers. There is some evidence of undesirable demetallation of the complexes in vivo. Zirconium is a bone seeker and demetallation can lead to increased radiation dose to bone marrow. As a result, several groups have recently H N
O
N
O
O
N
O 89Zr
HN
O
CH3
O O
O
N N H
Figure 18 Structure of 89Zr-desferrioxamine.
ARTICLE IN PRESS 35
Imaging in Drug Development
N N
N
O
O
O
O
O O N
O
O
N
HN
89Zr
O
N
O
O
O
O
O
N
89Zr
O
O
O
NH
N
O
O
N
O
N
Figure 19 Structures of improved 89Zr complexes.
developed new chelating agents for zirconium (Figure 19) that appear to form more stable complexes than those formed by DFO [96,97]. DFO provides only six of eight required donor atoms for zirconium. The improved stability of the new compounds is due to optimisation of the chelate ring size and complete satisfaction of the zirconium coordination sphere. Most clinical studies are with zirconium-89-labelled antibodies as the long half-life (78.4 h) allows imaging after several days. One recent study utilised 89Zr-trastuzumab, 89Zr-bevacizumab and 18F-FDG imaging to assess the ability of an HSP90 inhibitor to degrade client proteins in breast cancer patients [98]. HER-2 and ER oestrogen receptor are two important HSP90 client proteins involved in metastatic breast cancer. Imaging scans were performed at 2 and 4 days post-injections. One injection was carried out prior to therapy with the HSP90 inhibitor NVP-AUY922. A second injection was carried out 15 days later. 89Zr-trastuzumab tumour uptake was found to correlate with change in tumour size as measured by CT. Quantification of 89Zr-trastuzumab uptake in the tumour lesions showed heterogeneity within tumour lesions in the same and in different patients. No correlation was observed between 89Zr-bevacizumab uptake and tumour size. One reason for this could be that most 89Zr-bevacizumab tumour uptake was in bone lesions not measurable by CT. A clinical benefit was observed in 50% of heavily pretreated patients with progressive breast cancer although larger studies are required to confirm this. 3.2.5 Copper-64 Tracers The most widely used cyclotron production method of copper-64 is from nickel-64 using a (p,n) reaction. Copper-64 radiopharmaceuticals almost
ARTICLE IN PRESS 36
James Nairne et al.
always contain copper-64 in the +2 oxidation state as complexes containing copper in the +1 oxidation state tend to be unstable in biological media. Macrocyclic chelating agents are used to prepare copper-64 radiopharmaceuticals as the resulting copper complexes are kinetically more stable than those formed with acyclic chelating agents. A variety of chelating agents have been developed in recent years to find ones which can form copper-64 complexes at relatively low temperatures, but which will still have sufficient kinetic inertness to be stable in vivo [99]. The focus of most of these efforts is now on cross-bridged macrocycles. A recent example of a cyclam-based chelating agent with pendant carboxylic and phosphonic acid groups (Figure 20) shows that high labelling yields of the peptide-derivatised chelating agents (>90%) can be obtained at close to room temperature after 1 h [100]. It has also been suggested, however, that copper-64 complexes can be formed at room temperature with good in vivo stability with more conventional macrocyclic chelating agents such as TETA (Figure 20), if high purity copper-64 is used [101]. There are still relatively few literature examples of the use of copper-64 radiopharmaceuticals in clinical trials. The relatively short half-life of 12.7 h means that iodine-124 and zirconium-89 tend to be favoured for antibody applications. However, a recent study in HER-2-positive metastatic breast cancer patients using 64Cu-trastuzumab shows that good-quality images can be obtained up to 48 h post-injection if 18F-FDG is used beforehand to determine tumour locations [102]. A trastuzumab dose of 50 mg before the 64Cu-trastuzumab injection was found to improve image quality by increasing blood retention and decreasing liver uptake.
O O
O
N
O
P N
N O
64Cu
O
N
O
O
N N
64Cu
N
N
−
O O O
64Cu
CB-TE1A1P
Figure 20 Structures of
64
64Cu-TETA
Cu CB-TE1A1P and TETA.
ARTICLE IN PRESS 37
Imaging in Drug Development
3.2.6 Gallium-68 Tracers Generator produced gallium-68 is an attractive isotope for PET imaging as the 271-day half-life of the parent germanium-68 means that generators can be used for approximately 1 year [103]. The gallium-68 generator contains germanium-68 absorbed on alumina, SnO2 or TiO2 columns. Dilute HCl solutions are used to elute the decay product gallium-68 from the column. The recent approval of the Eckert and Ziegler pharmaceutical grade gallium-68 generator in Europe is likely to increase the number of future clinical studies. Gallium-68 forms complexes in which gallium is present in the +3 oxidation state. DOTA is the mostly widely used chelating agent as the 68GaDOTA complex (Figure 21) has good stability in vivo [21,103]. Formation of the 68Ga-DOTA complex, however, requires heating to 90 C and as such is not suitable for the labelling of compounds which are sensitive to heat. In addition to the rediscovery of NOTA as a chelating agent for gallium-68 [104], a number of new chelating agents have recently been developed which can coordinate gallium to form more stable complexes than those formed with DOTA. Of particular interest are the TRAP and CP256 chelating agents illustrated in Figure 22[105,106]. 68Ga-TRAP complexes can be formed on heating in acid solution as low as pH 0.5 and TRAP can potentially be used to directly label acidic [68Ga]gallium trichloride solutions eluting from the generator. CP256 was found to complex gallium-68 at room temperature using lower chelating agent amounts than required to form gallium-68 complexes with established chelating agents such as DOTA and NOTA. Radiochemical yields of 68Ga-CP256 of greater than 70% could be obtained at chelating agent concentrations of just 1 μM. O O
N
N
O N -
O
68Ga
N
O O O
Figure 21 Structure of 68Ga-DOTA complex.
ARTICLE IN PRESS 38
James Nairne et al.
O P
NH
OH O
O N
O
O 68Ga
O
N
P O
N
H3C
O OH
68Ga
N
P O
N
O
O
NH
N
O
N
H3C O
NH
O O
68Ga
O O
O
N
N H
NH
N
CH3
O
O
O O
68
68Ga-TRAP
Ga-NOTA
Figure 22 Structures of
68
68
Ga-CP256
Ga NOTA, TRAP and CP256.
3.2.7 Application of PET to Translational Medicine The chemical methodologies illustrated above demonstrate that there is a broad range of methods for introducing PET isotopes into molecules. The choice of chemistry and isotope is dependent on the application. There are four main areas for the application of PET in the drug development arena. These are to provide information on the biodistribution of a candidate; receptor occupancy studies that can be used to predict efficacy of drug candidates; monitoring of the effectiveness of therapy in early drug development phases; and as a method for pre-screening patient populations for clinical trials. Information on the biodistribution of a drug will require a radioactive analogue of the drug itself. This is most commonly done by the preparation of carbon-11 versions of the drug candidate or, if it has an accessible fluorine atom, fluorine-18 can be considered. Bergstr€ om et al. developed a radiosynthesis of [11C]-zolmitriptan for studying the biodistribution of the drug itself [107]. The study was aimed at demonstrating blood–brain barrier penetration of the drug. As most radiotracers are administered by bolus injection, delivery is usually very different to the drug studies and as such the tracer by itself cannot be used to measure bioavailability. In this study, two imaging measurements were carried out—one before and one 30 min after administration of the unlabelled drug. The difference in the uptake of the tracer was used to infer the biodistribution of the drug itself. Kiesewetter developed a radiosynthesis of racemic [18F]BMS-204352 for use in biodistribution studies to support the development of MaxiPost™ [108]. Radiometals may also be used for studying biodistribution of drugs. The therapeutic is likely to be a biologic as the biodistribution will be affected less by addition of a chelated metal. Roche is currently using a radiolabelled antibody against HER-3 89Zr-RO5479599 to provide information on their trial
ARTICLE IN PRESS 39
Imaging in Drug Development
of RO5479599 alone or in combination with erlotinib or cetuximab in patients with HER-3-positive solid tumours [109]. One of the biggest applications of PET in translational medicine is in investigating the receptor occupancy of drugs and drug candidates. This has been used as an end point in a number of studies in neuropsychiatry in particular. The key aspect of use of PET in this arena is that there needs to be a valid biomarker of the receptor to be studied that is not structurally similar to the drug under study. This will ensure that any displacement of the tracer can be attributed to the drug. The antipsychotic drug quetiapine has been shown to have efficacy in a range of psychiatric disorders including schizophrenia, depression and anxiety. The drug has been shown to have high affinity for the 5-HT1A and dopamine D2 receptors, while its human metabolite, norquetiapine, also has high affinity to the norepinephrine transporter (NET). It was this latter property that was shown to be prevalent in a clinical study using the specific NET radiotracer (s,s)-[18F]FMeNER-D2 [110]. PET has also been used to study the pharmacokinetics of drugs. For example, the occupancy of dopamine D2 and D3 receptors by ziprasidone across a 23 h period has been reported. The occupancy was measured by assessing the uptake of [11C]raclopride, which binds to both D2 and D3 receptors. In this case, the occupancy half-life was found to be 8.3 h, data that support the clinical twice-daily treatment [111]. [11C]Raclopride has also been used to assess drug occupancy of the investigational antipsychotic drugs JNJ-37822681 [112], YKP1358 (structure not disclosed) [113] and BL-1020 [114] (Figure 23). More selective tracers for dopamine receptors
F
N
N
N
CF3
N
S N
F
N
Cl
BL-1020 NH2 N
H311C
O
N N
NH2 O
N H JNJ-37822681
O
N N O N
[11C]SCH442416
Figure 23 Structures of JNJ-37822681, BL-1020 and [11C]SCH442416.
ARTICLE IN PRESS 40
James Nairne et al.
can be used to improve the understanding of mechanisms of action; [11C] PHNO has been used to understand the effects of antipsychotics on the D3 receptor [115]. This study showed that the receptor was possibly upregulated after short-term treatment with either risperidone or olanzapine. Dose occupancy studies have been used to justify the continuation of development of compounds. [11C]SCH442416 has been used to assess the binding of vipadenant to the A2A receptor [116]. Applications of PET in translation medicine in oncology have focussed on the effectiveness of treatment. Several tracers that are proliferation markers have been developed, and the wide availability of [18F]FDG makes it a commonly used marker for therapeutic trials [117]. The application of other markers is much less widespread. One of the features of FDG is that it is cleared through the urinary system and so accumulates in the bladder. This makes it suboptimal for imaging bladder and prostate conditions. Imaging agents that are under evaluation in the clinic for monitoring the effectiveness of therapies for such conditions are those derived from choline—[11C]choline [118] and [18F]fluorocholine [119]—and the amino acid analogue [18F] fluciclovine (FACBC) [120]. These three tracers have been used in other oncology applications too. [18F]Fluorothymidine ([18F]FLT) uptake has been shown to change in response to treatment of patients with capecitabine for breast cancer [121], erlotinib for non-small cell lung cancer [122] and bevacizumab for recurrent malignant glioma [123] among others. There are opportunities to show effectiveness earlier in studies by using tracers that probe the mechanism of action of the drug. For example, the integrin binder [18F]fluciclatide is being used in the early determination of the effectiveness of anti-angiogenic therapies in clinical studies [124], following preclinical work showing the ability of the tracer to demonstrate the effectiveness of sunitinib in reducing microvessel density ahead of reduction of tumour volume [125]. A further application of imaging in translational medicine is as a method for selection of patients for therapy groups. This can be particularly useful if the aim of the therapy is to treat diseases in their early phases. An example of this is in the attempts to treat neurodegenerative disorders such as Alzheimer’s disease. It is known that some of the underlying pathology of the disease exists before any symptoms show [126]. This leads to the possibility that healthy volunteers may be on the path to the disease, thus having an adverse effect on the outcome of the drug under study. Merck are using this strategy in assessing the diagnosis of prodromal Alzheimer’s disease by using [18F]flutemetamol in the initial trial of MK-8931 [127]. Lilly used
ARTICLE IN PRESS Imaging in Drug Development
41
[18F]AV-45 ([18F]-florbetapir) as a secondary endpoint in the Phase III assessment of LY450139 [128]. Similar strategies could be employed in the development of treatment of Parkinson’s disease where it has already been shown that a reduction in uptake of [18F]F-dopa precedes observation of symptoms [129]. Clinical trials involving targeted gallium-68 tracers are dominated by the three somatostatin receptor imaging agents 68Ga-DOTA TOC, 68 Ga-DOTA TATE and 68Ga-DOTA NOC. Although not commercially available, all three tracers are now widely used in Europe for the diagnosis of neuroendocrine tumours (NET). The gallium-68 NET imaging agents have been used extensively during the development of the corresponding yttrium-90 and lutetium-177 radiotherapeutics. A recent study concluded that high baseline tracer uptake values obtained with 68Ga DOTA TOC imaging were suggestive of a better therapeutic response to 90Y/177Lu DOTA TOC radiotherapy in patients with hepatic metastases from gastro-entero-pancreatic NETs [130].
4. SUMMARY AND FUTURE PROSPECTS Imaging has the ability to show functional and biochemical changes that can help not only the understanding of disease mechanisms but also the response of the body to treatment. fMRI has the capability to look at these changes in real time, thereby providing a great opportunity to give an early indication of the effect of a putative treatment. The number of clinical trials that already have an fMRI study as part of the protocol indicates that the technique is making big strides in its impact on drug development. This is likely to continue as instrument sensitivity improves and the development of new sequences enables the extraction of more information from endogenous molecules. There is also the prospect of the introduction of molecular imaging techniques; use of hyperpolarised carbon-13 pyruvate has been tried [131] and there is potential of using fluorine-19 [132], but these techniques are some way in the future. Nuclear medicine is a little further back in the scale of its adoption in translational medicine. There are a few reasons for this. Despite the large installed base of SPECT cameras and the ready availability of radionuclides for the modality, there has been less emphasis on the development of SPECT radiotracers for molecular imaging techniques than for PET. This has been due to the reduced accuracy of absolute quantification available for SPECT until the recent development of improved cameras and software. In addition, the
ARTICLE IN PRESS 42
James Nairne et al.
isotopes available for SPECT are less amenable for application to small molecules, though we have shown some examples of compounds that meet this criterion. As the pharmaceutical industry increases the use of peptides, proteins, antibodies and even cells, these drawbacks become less relevant. PET has long been considered by the nuclear medicine community as the tool that can deliver information at a biochemical level, and as such a great deal of research has gone into the development of tracers that can indicate the mechanism of action of compounds by use of labelled analogues of drugs and observing the effect of such drugs on the biochemistry of the body. Much of this early work has been done using carbon-11 as the reporter. The major challenge in using this radionuclide is the short half-life that has prevented widespread use in large clinical trials. Fluorine-18 has been the main radionuclide of choice for the development of radiotracers due to its longer half-life. This makes the application of PET to large-scale multi-centre clinical trials feasible. As well as the ability to detect changes in the biochemistry of disease early, PET can be used to select clinical trial groups based on the underlying pathology they show. Many neurodegenerative diseases remain asymptomatic until the disease is well advanced; inclusion of patients with early stage in control groups can mask the benefits of a treatment in the treated group. Although a disadvantage in this case, PET could be used in the future to identify asymptomatic patient groups that would benefit from treatment. Recent advances in nuclear medicine imaging highlighted in this review provide the opportunity for the use of radiotracers in the development of small molecule drugs with the use of iodine-123, carbon-11 and fluorine18-labelled molecules and peptide therapies and biopharmaceuticals through labelling with technetium-99m, gallium-68 and zirconium-89. Following the biochemical changes in response to drug treatment gives the possibility for early indications of clinical efficacy; it is anticipated that the application of imaging in this area will continue to grow.
REFERENCES [1] Weissleder R, Ntziachristos V. Shedding light onto live molecular targets. Nat Med 2003;9:123–8. [2] Ntziachristos V. Going deeper than microscopy: the optical imaging frontier in biology. Nat Methods 2010;7:603–14. [3] Bellin M-F, Van Der Molen AJ. Extracellular gadolinium-based contrast media: an overview. Eur J Radiol 2008;66:160–7. [4] Hundley WG, Bluemke DA, Finn JP, Flamm SD, Fogel MA, Friedrich MG, et al. ACCF/ACR/AHA/NASCI/SCMR 2010 expert consensus document on
ARTICLE IN PRESS Imaging in Drug Development
[5] [6] [7] [8] [9] [10] [11] [12] [13]
[14] [15] [16] [17] [18] [19] [20] [21] [22] [23]
43
cardiovascular magnetic resonance: a report of the American College of Cardiology Foundation Task Force on Expert Consensus Documents. Circulation 2010;121:2462–508. McLaughlin R, Hylton N. MRI in breast cancer therapy monitoring. NMR Biomed 2011;24:712–20. Agrawal G, Su MY, Nalcioglu O, Feig SA, Chen JH. Significance of breast lesion descriptors in the ACR BI-RADS MRI lexicon. Cancer 2009;115:1363–80. O’Connor JPB, Jackson A, Parker GJM, Roberts C, Jayson GC. Dynamic contrastenhanced MRI in clinical trials of antivascular therapies. Nat Rev Clin Oncol 2012;9:167–77. Leach MO, Morgan B, Tofts PS, Buckley DL, Huang W, Horsfield MA, et al. Imaging vascular function for early stage clinical trials using dynamic contrast-enhanced magnetic resonance imaging. Eur Radiol 2012;22:1451–64. Thoeny HC, Ross BD. Predicting and monitoring cancer treatment response with diffusion-weighted MRI. J Magn Reson Imaging 2010;32:2–16. Moffat BA, Chenevert TL, Lawrence TS, Meyer CR, Johnson TD, Dong Q, et al. Functional diffusion map: a noninvasive MRI biomarker for early stratification of clinical brain tumour response. Proc Natl Acad Sci USA 2005;102:5524–9. Bains LJ, Zweifel M, Thoeny HC. Therapy response with diffusion MRI: an update. Cancer Imaging 2012;12:395–402. Padhani AR, Liu G, Mu-Koh D, Chenevert TL, Thoeny HC, Takahara T, et al. Diffusion-weighted magnetic resonance imaging as a cancer biomarker: consensus and recommendations. Neoplasia 2009;11:102–25. Wu L-M, Hu J-N, Gu H-Y, Hua J, Chen J, Xu J-R. Can diffusion-weighted MR imaging and contrast-enhanced MR imaging precisely evaluate and predict pathological response to neoadjuvant chemotherapy in patients with breast cancer? Breast Cancer Res Treat 2012;135:17–28. Borsook D, Hargreaves R, Becerra L. Can functional magnetic resonance imaging improve success rates in CNS drug discovery? Expert Opin Drug Discov 2011;6:597–617. Nutt DJ, Attridge J. CNS drug development in Europe—past progress and future challenges. Neurobiol Dis 2014;61:6–20. Jenkins BG. Pharmacologic magnetic resonance imaging (phMRI): imaging drug action in the brain. Neuroimage 2012;62:1072–85. Heeger DJ, Ress D. What does fMRI tell us about neuronal activity? Nat Rev Neurosci 2002;3:142–51. Smucny J, Wylie KP, Tregellas JR. Functional magnetic resonance imaging of intrinsic brain networks for translational drug discovery. Trends Pharmacol Sci 2014;35:397–405. Bartholoma MD, Louie AS, Valliant JF, Zubieta J. Technetium and gallium derived radiopharmaceuticals: comparing and contrasting the chemistry of two important radiometals for the molecular imaging era. Chem Rev 2010;110:2903–20. Mohan HK, Miles KA. Cost-effectiveness of 99mTc sestamibi on predicting response to chemotherapy in patients with lung cancer: systematic review and meta-analysis. J Nucl Med 2009;50:376–81. Wadas TJ, Wong EH, Weisman GR, Anderson CJ. Coordinating radiometals of copper, gallium, indium, yttrium and zirconium. Chem Rev 2010;110:2858–902. Eersels JLH, Travis MJ, Herscheid JDM. Manufacturing I-123-labelled radiopharmaceuticals. Pitfalls and solutions. J Label Compd Radiopharm 2005;48:241–57. Richardson AP, Mountford PJ, Baird AC, Heyderman E, Coakley AJ. An improved method of labelling antibodies with 123-I. Nucl Med Commun 1986;7:355–61.
ARTICLE IN PRESS 44
James Nairne et al.
[24] Maurer AH, Elsinga P, Fanti S, Nguyen B, Oyen WJG, Weber WA. Imaging the folate receptor on cancer cells with 99mTc-etarfolatide: properties, clinical use and future potential of folate receptor imaging. J Nucl Med 2014;55:701–4. [25] Witzig TE, Wiseman GA, Maurer MJ, Habermann TM, Micallef INM, Nowakowski GS, et al. A phase I trial of immunostimulatory CpG 7909 oligodeoxynucleotide and 90-yttrium ibritumomab tiuxetan radioimmunotherapy for relapsed B-cell non-Hodgkin lymphoma. Am J Hematol 2013;88:589–93. [26] Schapira AHV, McDermott MP, Barone P, Comella CL, Albrecht S, Hsu HH, et al. Pramipexole in patients with early Parkinson’s disease (PROUD): a randomised delayed-start trial. Lancet Neurol 2013;12:747–55. [27] Ishihara M, Asakura M, Kimura K, Nakao K, Hamada C, Hirayama A. Trial design and rationale of TY-51924 as a novel Na +/H + exchanger inhibitor in patients with ST-elevation acute myocardial infarction undergoing percutaneous coronary intervention. J Cardiol 2014;63:82–7. [28] Catafau AM, Bullich S, Nucci G, Burgess C, Gray F, Merlo-Pich E, et al. Contribution of SPECT measurements of D2 and 5-HT2A occupancy to the clinical development of the antipsychotic SB-773812. J Nucl Med 2011;52:526–34. [29] McCarron JA, Turton DR, Pike VW, Poole KG. Remotely-controlled production of the 5-HT1A receptor radioligand, [carbonyl-11C]WAY100635, via11 C-carboxylation of an immobilized Grignard reagent. J Label Compd Radiopharm 1996;38:941–53. [30] Wilson AA, McCormick P, Kapur S, Willeit M, Garcia A, Hussey D, et al. Radiosynthesis and evaluation of [11C]-(+)-propyl-3,4,4a,5,6,10b-hexahydro-2Hnaphtho[1,2-b][1,4]oxazin-9-ol as potential radiotracer for in vivo imaging of the dopamine D2 high affinity state with positron emission tomography. J Med Chem 2005;48:4153–60. [31] Bolton R. Isotopic methylations. J Label Compd Radiopharm 2001;44:701–36, and refs therein. [32] Halldin C, Farde L, H€ ogberg T, Hall H, Sedvall G. Carbon-11 labelling of eticlopride in two different positions—a selective high-affinity ligand for the study of dopamine D-2 receptors using PET. Appl Radiat Isot 1990;41:669–74. [33] Hirani E, Gillies J, Karaswa A, Shimada J, Kase H, Opacka-Juffry J, et al. Evaluation of [4-O-methyl-11C]KW-6002 as a potential PET ligand for mapping central adenosine A2A receptors in rats. Synapse 2001;42:164–72. [34] Halldin C, Stone-Elander S, Thorell J-O, Persson A, Sedvall G. 11C-labelling of Ro 15-1788 in two different positions, and also 11C-labelling of its main metabolite Ro 15-3890, for PET studies of benzodiazepine receptors. Int J Radiat Appl Instrum A 1988;39:993–7. [35] Pascali C, Bogni A, Iwata R, Decise D, Crippa F, Bombardieri E. Simple and efficient production of L-(S-methyl-11C)methionine by on-column [11C]methylation on C18 Sep-Pak. J Label Compd Radiopharm 1999;42:715–24. [36] Samuelsson L, La˚ngstr€ om B. Synthesis of 1(20 -deoxy-20 -fluoro-β-Darabinofuranosyl)-[Methyl-11C]thymine ([11C]FMAU) via a Stille cross-coupling reaction with [11C]methyl iodide. J Label Compd Radiopharm 2003;46:263–72. [37] Hostetler ED, Terry GE, Burns HD. An improved synthesis of substituted [11C]toluenes via Suzuki coupling with [11C]methyl iodide. J Label Compd Radiopharm 2005;48:629–34. [38] Elsinga PH, Keller E, De Groot TJ, Visser GM, Vaalburg W. Synthesis of [11C]methyl magnesium iodide and its application to the introduction of [11C]-N-tert-butyl groups and [11C]-sec-alcohols. Appl Radiat Isot 1995;46:227–31. [39] Jewett DM. A simple synthesis of [11C]methyl triflate. Appl Radiat Isot 1992;43:1383–5.
ARTICLE IN PRESS Imaging in Drug Development
45
[40] Langer O, Nagren K, Dolle´ F, Lundkvist C, Sandell J, Swahn C-G, et al. Precursor synthesis and radiolabelling of the dopamine D2 receptor ligand [11C]raclopride from [11C]methyl triflate. J Label Compd Radiopharm 1999;42:1183–93. [41] Clark JC, Buckingham PD. Short-lived radioactive gases for clinical use. London: Butterworths; 1975. p. 231. [42] Zeisler SK, Nader M, Theobald A, Oberdorfer F. Conversion of no-carrier-added [11C]carbon dioxide to [11C]carbon monoxide on molybdenum for the synthesis of 11 C-labeled aromatic ketones. Appl Radiat Isot 1997;48:1091–5. [43] Barletta J, Karimi F, Doi H, La˚ngstr€ om B. Synthesis of diethyl [carbonyl-11C]malonate from [11C]carbon monoxide by rhodium-promoted carbonylation and its application as a reaction intermediate. J Label Compd Radiopharm 2006;49:801–9. [44] Kealey S, Husbands SM, Bennacef I, Gee AD, Passchier J. Palladium-mediated oxidative carbonylation reactions for the synthesis of 11C-radiolabelled ureas. J Label Compd Radiopharm 2014;57:202–8. [45] Doi H, Barletta J, Suzuki M, Noyori R, Watanabe Y, La˚ngstr€ om B. Synthesis of 11 C-labelled N,N0 -diphenylurea and ethyl phenylcarbamate by a rhodium-promoted carbonylation via [11C]isocyanatobenzene using phenyl azide and [11C]carbon monoxide. Org Biomol Chem 2006;2:3063–6. [46] Karimi F, Barletta J, La˚ngstr€ om B. Palladium-mediated 11C-carbonylative crosscoupling of alkyl/aryl iodides with organostannanes: an efficient synthesis of unsymmetrical alkyl/aryl [11C-carbonyl]ketones. Eur J Org Chem 2005;2374–8. [47] Lidstr€ om P, Kihlberg T, La˚ngstr€ om B. [11C]Carbon monoxide in the palladiummediated synthesis of 11C-labelled ketones. J Chem Soc Perkin Trans 1 1997;2701–6. [48] Eriksson J, Aberg O, La˚ngstr€ om B. Synthesis of [11C]/[13C]acrylamides by palladiummediated carbonylation. Eur J Org Chem 2007;455–61. [49] Larsen P, Ulin J, Dahlstr€ om K, Jensen M. Synthesis of [11C]iodomethane by iodination 11 of [ C]methane. Appl Radiat Isot 1997;48:153–7. [50] Iwata R, Ido T, Takahashi T, Nakanishi H, Iida S. Optimization of [11C]HCN production and no-carrier-added [1-11C]amino acid synthesis. Int J Radiat Appl Instrum A Appl Radiat Isot 1987;38:97–102. [51] Landais P, Crouzel C. A new synthesis of [11C]phosgene. Int J Radiat Appl Instrum A Appl Radiat Isot 1987;38:297–300. [52] Nishijima K-I, Kuge Y, Seki K-I, Ohkura K, Motoki N, Nagatsu K, et al. A simplified and improved synthesis of [11C]phosgene with iron and iron (III) oxide. Nucl Med Biol 2002;29:345–50. [53] Andersson JD, Seneca N, Trong P, Wensbo D, Raboisson P, Farde L, et al. Palladium mediated 11C-cyanation and characterization in the non-human primate brain of the novel mGluR5 radioligand [11C]AZD9272. Nucl Med Biol 2013;40:547–53. [54] Mathews WB, Monn JA, Ravert HT, Holt DP, Schoep DD, Dannals RF. Synthesis of a mGluR5 antagonist using [11C]copper(I) cyanide. J LabelCompd Radiopharm 2006;49:829–34. [55] Firnau G, Chirakal R, Garnett ES. Aromatic radiofluorination with [18F] fluorine gas: 6-[18F]fluoro-L-dopa. J Nucl Med 1984;25:1228–33. [56] Ehrenkaufer RE, Potocki JF, Jewett DM. Simple synthesis of F-18-labelled 2-fluoro-2-deoxy-D-glucose: concise communication. J Nucl Med 1984;25:333–7. [57] Teare H, Robins EG, Arstad E, Luthra SK, Gouverneur V. Synthesis and reactivity of [18F]-N-fluorobenzenesulfonamide. Chem Commun 2007;2330–2. [58] Block D, Klatte B, Kn€ ochel A, Beckmann R, Holm U. N.C.A. [18F]-labeling of aliphatic compounds in high yields via aminopolyether-supported nucleophilic substitution. J Label Compd Radiopharm 1986;23:467–77. [59] Hamacher K, Coenen HH, St€ ocklin G. Efficient stereospecific synthesis of no-carrieradded 2-[18F]-fluoro-2-deoxy-D-glucose using aminopolyether supported nucleophilic substitution. J Nucl Med 1986;27:235–8.
ARTICLE IN PRESS 46
James Nairne et al.
[60] Pauwels EKJ, Ribiero MJ, Stoot JHMB, McCready VR, Bourguignon M, Mazie`re B. FDG accumulation and tumour biology. Nucl Med Biol 1998;25:317–22. [61] Gambhir SS, Czernin J, Schwimmer J, Silverman DHS, Coleman RE, Phelps ME. A tabulated summary of the FDG PET literature. J Nucl Med 2001;42:1S–93S. [62] Mukherjee J, Christian BT, Dunigan KA, Shi B, Narayanan TK, Satter M, et al. Brain imaging of 18F-fallypride in normal volunteers: blood analysis, biodistribution, testretest studies, and preliminary assessments of sensitivity to aging effects on dopamine D-2/D-3 receptors. Synapse 2002;46:170–88. [63] Saigal N, Pichika R, Easwaramoorthy B, Collins D, Shi B, Narayanan TK, et al. Synthesis and biologic evaluation of a novel serotonin 5-HT1A receptor ligand, 18Flabeled mefway, in rodents and imaging by PET in a nonhuman primate. J Nucl Med 2006;47:1697–706. [64] Yao C-H, Lin K-J, Weng C-C, Hisao I-T, Ting T-C, Jan T-R, et al. GMP-compliant automated synthesis of [18F]AV-45 (Florbetapir F18) for imaging β-amyloid plaques in human brain. Appl Radiat Isot 2010;68:2293–7. [65] Shoup TM, Goodman MM. Synthesis of [F-18]-1-amino-3-fluorocyclobutane1-carboxylic acid (FACBC): a PET tracer for tumour delineation. J Label Compd Radiopharm 1999;42:215–25. [66] Iwata R, Pascali C, Bogni A, Furumoto S, Terasaki K, Yanai K. [18F]Fluoromethyl triflate, a novel and reactive [18F]fluoromethylating agent: preparation and application to the on-column preparation of [18F]fluorocholine. Appl Radiat Isot 2002;57:347–52. [67] Wester HJ, Herz M, Weber W, Heiss P, Senekowitsch-Schmidtke R, Schweiger M, et al. Synthesis and radiopharmacology of O-(2-[18F]fluoroethyl)-L-tyrosine for tumour imaging. J Nucl Med 1999;40:205–12. [68] McGinnity CJ, Hammers A, Barros DAR, Luthra SK, Jones PA, Trigg W, et al. Initial evaluation of 18F-GE-179, a putative PET tracer for activated N-methyl D-aspartate receptors. J Nucl Med 2014;55:423–30. [69] Kilbourn MR, Welch MJ, Dence CS, Tewson TJ, Saji H, Maeda M. Carrier-added and no-carrier-added syntheses of [18F]spiperidol and [18F]haloperidol. Int J Appl Radiat Isot 1984;35:591–8. [70] Hamacher K, Hamkens W. Remote controlled one-step production of 18F labelled butyrophenone neuroleptics exemplified by the synthesis of N.C.A. [18F]Nmethylspiperone. Appl Radiat Isot 1995;46:911–6. [71] Vandenberghe R, Van Laere K, Ivanoiu A, Salmon E, Bastin C, Triau E, et al. 18Fflutemetamol amyloid imaging in Alzheimer disease and mild cognitive impairment: a phase 2 trial. Ann Neurol 2010;68:319–29. [72] Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, Holt DP, et al. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh compound B. Ann Neurol 2004;55:306–19. [73] Mandap KS, Ido T, Kiyono Y, Kobayashi M, Lohith TG, Mori T, et al. Development of microwave-based automated nucleophilic [18F]fluorination system and its application to the production of [18F]flumazenil. Nucl Med Biol 2009;36:403–9. [74] Lemaire C, Gillet S, Guillouet S, Plenevaux A, Aerts J, Luxen A. Highly enantioselective synthesis of no-carrier-added 6-[18F]fluoro-L-dopa by chiral phasetransfer alkylation. Eur J Org Chem 2004;2899–904. [75] Shen B, Ehrlichmann W, Uebele M, Machulla H-J, Reischl G. Automated synthesis of N.C.A. [18F]FDOPA via nucleophilic aromatic substitution with [18F]fluoride. Appl Radiat Isot 2009;67:1650–3. [76] Shen B, L€ offler D, Zeller K-P, Uebele M, Reischl G, Machulla H-J. Decarbonylation of multi-substituted [18F]benzaldehydes for modelling syntheses of 18F-labelled aromatic amino acids. Appl Radiat Isot 2007;65:1227–31.
ARTICLE IN PRESS Imaging in Drug Development
47
[77] Wagner FM, Ermert J, Coenen HH. Three-step, “one-pot” radiosynthesis of 6-fluoro-3,4-dihydroxy-l-phenylalanine by isotopic exchange. J Nucl Med 2009;50:1724–9. [78] Carroll MA, Nairne J, Woodcraft JL. Diaryl iodonium salts: a solution to 3-[18F] fluoropyridine. J Label Compd Radiopharm 2007;50:452–4. [79] Karramkam M, Hinnen F, Vaufrey F, Dolle´ F. 2-, 3-, and 4-[18F]fluoropyridine by no-carrier-added nucleophilic aromatic substitution with K[18F]F-K222—a comparative study. J Label Compd Radiopharm 2003;46:979–92. [80] Chun J-H, Pike VW. Single-step syntheses of no-carrier-added functionalized [18F] fluoroarenes as labelling synthons from diaryliodonium salts. Org Biomol Chem 2013;11:6300–6. [81] Watson DA, Su MJ, Teverovskiy G, Zhang Y, Garcia-Fortanet J, Kinzael T, et al. Formation of ArF from LPdAr(F): catalytic conversion of aryl triflates to aryl fluorides. Science 2009;325:1661–4. [82] Lee E, Kamlet AS, Powers DC, Neumann CN, Boursalian GB, Furuya T, et al. A fluoride-derived electrophilic late-stage fluorination reagent for PET imaging. Science 2011;334:639–42. [83] Lee E, Hooker JM, Ritter T. Nickel-mediated oxidative fluorination for PET with aqueous [18F]fluoride. J Am Chem Soc 2012;134:17456–8. [84] Tredwell M, Preshlock SM, Taylor NJ, Gruber S, Huiban M, Passchier J, et al. A general copper-mediated nucleophilic 18F fluorination of arenes. Angew Chem Int Ed Engl 2014;50:7751–5. [85] Glaser M, Morrison M, Solbakken M, Arukwe J, Karlsen H, Wiggen U, et al. Radiosynthesis and biodistribution of cyclic RGD peptides conjugated with novel [18F] fluorinated aldehyde-containing prosthetic groups. Bioconjug Chem 2008;19:951–7. [86] Rennen HJJM, Laverman P, van Eerd JEM, Oyen WJG, Cortstens FHM, Boerman OC. PET imaging of infection with a HYNIC-conjugation LTB4 antagonist labelled with F-18 via hydrazine formation. Nucl Med Biol 2007;34:691–5. [87] Glaser M, Iveson P, Hoppmann S, Indrevoll B, Wilson A, Arukwe J, et al. Three methods for the 18F labelling of the HER-2 affibody molecule ZHER2:2891 including preclinical assessment. J Nucl Med 2013;54:1981–8. [88] Li X-G, Dall’Angelo S, Schweiger LF, Zanda M, O’Hagan D. [18F]-5Fluoro-5-deoxyribose, an efficient peptide bioconjugation ligand for positron emission tomography (PET) imaging. Chem Commun 2012;48:5247–9. [89] Hausner SH, Marik J, Gagnon KMJ, Sutcliffe JL. In vivo positron emission tomography (PET) imaging with an αvβ6 specific peptide radiolabelled using 18F-“Click” chemistry: evaluation and comparison with 4-[18F]fluorobenzoyl- and 2-[18F] fluoropropionyl-peptides. J Med Chem 2008;51:5901–4. [90] Ramenda T, Kneiss T, Bergmann R, Steinbach J, Wuest F. Radiolabelling of proteins with fluorine-18 via click chemistry. Chem Commun 2009;7521–3. [91] Wa¨ngler C, Niedermoser S, Chin J, Orchowski K, Schirrmacher E, Jurkschat K, et al. One-step 18F-labeling of peptides for positron emission tomography imaging using the SiFA methodology. Nat Protoc 2012;7:1946–55. [92] D’Souza CA, McBride WJ, Sharkey RM, Todaro LJ, Goldenberg DM. High-yielding aqueous 18F-labeling of peptides via Al18F chelation. Bioconjug Chem 2011;22:1793–803. [93] Wan W, Guo N, Pan D, Yu C, Weng Y, Luo S, et al. First experience of 18F-alfatide in lung cancer patients using a new lyophilized kit for rapid radiofluorination. J Nucl Med 2013;54:691–8. [94] Cascini GL, Asabella AN, Notaristefano A, Restuccia A, Ferrari C, Rubini D, et al. 124 Iodine: a longer-life positron emitter isotope—new opportunities in molecular imaging. Biomed Res Int 2014, Article ID 672094.
ARTICLE IN PRESS 48
James Nairne et al.
[95] O’Donoghue JA, Smith-Jones PM, Humm JL, Ruan S, Pryma DA, Jungbuth AA, et al. 124 I-huA33 antibody uptake is driven by A33 antigen concentration in tissues from colorectal cancer patients imaged by immuno-PET. J Nucl Med 2011;52:1878–85. [96] Deri MA, Ponnala S, Zeglis BM, Pohl G, Dannenberg JJ, Lewis JS, et al. Alternative chelator for 89Zr radiopharmaceuticals: radiolabelling and evaluation of 3,4,3-(LI-1,2HOPO). J Med Chem 2014;57:4849–60. [97] Guerard F, Lee Y-S, Brechbiel MW. Rational design, synthesis and evaluation of tetrahydroxamic acid chelators for stable complexation of zirconium (IV). Chem Eur J 2014;20:5584–91. [98] Gaykema SBM, Schroder CP, Vitfell-Rasmussen J, Chua S, Oude Munnink TH, Brouwers AH, et al. 89Zr-trastuzumab and 89Zr-bevacizumab PET to evaluate the effect of the HSP90 inhibitor NVP-AUY922 in metastatic breast cancer patients. Clin Cancer Res 2014;20:3945–54. [99] Cai Z, Anderson CJ. Chelators for copper radionuclides in positron emission tomography radiopharmaceuticals. J Label Compd Radiopharm 2014;57:224–30. [100] Jiang M, Ferdani R, Shokeen M, Anderson CJ. Comparison of two crossbridged macrocyclic chelators for the evaluation of 64Cu-labeled-LLP2A, a peptidomimetic ligand targeting VLA-4-positive tumours. Nucl Med Biol 2013;40:245–51. [101] Cooper MS, Ma MT, Sunassee K, Shaw KP, Williams JD, Paul RL, et al. Comparison of 64Cu-complexing bifunctional chelators for radioimmunoconjugation: labelling efficiency, specific activity, and in vitro/in vivo stability. Bioconjug Chem 2012;23:1029–39. [102] Mortimer JE, Bading JR, Colcher DM, Conti PS, Frankel PH, Carroll MI, et al. Functional imaging of human epidermal growth factor 2-positive metastatic breast cancer using 64Cu-DOTA-trastuzumab PET. J Nucl Med 2014;55:23–9. [103] Velikyan I. Prospective of 68Ga-radiopharmaceutical development. Theranostics 2014;4:47–79. [104] Velikyan I, Maecke H, La˚ngstr€ om B. Convenient preparation of 68Ga-based PET-radiopharmaceuticals at room temperature. Bioconjug Chem 2008;19:569–73. [105] Notni J, Simecek J, Wester H-J. Phosphonic acid functionalised polyazacyclononane chelators for radiodiagnostics and radiotherapeutics: unique characteristics and applications. Chem Med Chem 2014;9:1107–15. [106] Berry DJ, Ma Y, Ballinger JR, Tavare R, Koers A, Sunassee K, et al. Efficient bifunctional gallium-68 chelators for positron emission tomography: tris(hydroxypyridinone)ligands. Chem Commun 2011;47:7068–70. [107] Bergstr€ om M, Yates R, Wall A, Ka˚gedal M, Syva¨nen S, La˚ngstr€ om B. Blood-brain barrier penetration of zolmitriptan—modelling of positron emission tomography data. J Pharmacokinet Pharmacodyn 2006;33:75–91. [108] Kiesewetter DO, Jagoda EM, Starrett JE, Gribkoff VK, Hewawasam P, Srinivas N, et al. Radiochemical synthesis and biodistribution of a novel maxi-K potassium channel opener. Nucl Med Biol 2002;22:55–9. [109] A study of RO5479599 alone or in combination with cetuximab or erlotinib in patients with metastatic and/or locally advanced malignant HER3-positive solid tumours. Clinicaltrials.gov Identifier NCT01482377. [110] Nyberg S, Jacuite A, Takano A, Ka˚gedal M, Csele´nyi Z, Halldin C, et al. Norepinephrine transporter occupancy in the human brain after oral administration of quetiapine XR. Int J Neuropsychopharmacol 2013;16:2235–44. [111] Suzuki T, Graff-Guerrero A, Uchida H, Remington G, Caravaggio F, Borlido C, et al. Dopamine D2/D3 occupancy of ziprasidone across a day: a within-subject PET study. Psychopharmacology 2013;228:43–51.
ARTICLE IN PRESS Imaging in Drug Development
49
[112] Te Beek ET, De Boer P, Moerland M, Schmidt ME, Hoetjes NJ, Windhorst AD, et al. In vivo quantification of striatal dopamine D2 receptor occupancy by JNJ-3782281 using [11C]raclopride and positron emission tomography. J Psychopharmacol 2012;26:1128–35. [113] Kim E, Howes OD, Kim B-H, Yu K-S, Jeong JM, Lee JS, et al. The use of healthy volunteers instead of patients to inform drug dosing studies: a [11C]raclopride PET study. Psychopharmacology 2011;217:515–23. [114] Appel L, Geffen Y, Heurling K, Eriksson C, Antoni G, Kapur S. BL-1020, a novel antipsychotic candidate with GABA enhancing effects: D2 receptor occupancy study in humans. Eur Neuropsychopharmacol 2009;19:841–50. [115] Mizrahi R, Agid O, Borlido C, Suridjan I, Rusjan P, Houle S, et al. Effects of antipsychotics on D3 receptors: a clinical PET study in first episode antipsychotic naı¨ve patients with schizophrenia using [11C]-+-PHNO. Schizophr Res 2011;131:63–8. [116] Brooks DJ, Papapetropoulos S, Vandenhende F, Tomic D, He P, Coppell A, et al. An open label positron emission tomography study to assess adenosine A2A brain receptor occupancy of vipadenant (BIIB014) at steady-state levels in healthy male volunteers. Clin Neuropharmacol 2010;33:55–60. [117] Ben-Haim S, Ell P. 18F-FDG PET and PET/CT in the evaluation of cancer treatment response. J Nucl Med 2009;50:88–99. [118] Challapalli A, Barwick T, Tomasi G, O’Doherty M, Contractor K, Stewart S, et al. Exploring the potential of [11C]choline-PET/CT as a novel imaging biomarker for predicting early treatment response in prostate cancer. Nucl Med Commun 2014;35:20–9. [119] Chemotherapy response monitoring with 18F-choline PET/CT in hormone refractory prostate cancer. Clinicaltrial.gov Identifier NCT00928252. [120] Nanni C, Schiavini R, Boschi S, Ambrosini V, Pettinato C, Brunocilla E, et al. Comparison of 18F-FACBC and 11C-Choline PET/CT in patients with radically treated prostate cancer and biochemical relapse: preliminary results. Eur J Nucl Med Mol Imaging 2013;40:11–7. [121] Kenny LM, Contractor KB, Stebbing J, Al-Nahhas A, Palmieri C, Shousha S, et al. Altered tissue 30 -deoxy-30 -[18F]fluorothymidine pharmacokinetics in human breast cancer following capecitabine treatment detected by positron emission tomography. Clin Cancer Res 2009;15:6649–57. [122] Scheffler M, Kobe C, Zander T, Nogova L, Kahraman D, Thomas R, et al. Monitoring reversible and irreversible EGFR inhibition with erlotinib and afatinib in a patient with non-small cell lung cancer (NSCLC) using sequential [18F]fluorothymidine (FLT-)PET. Lung Cancer 2012;77:617–20. [123] Schwarzenberg J, Czernin J, Cloughesy TF, Ellingson BM, Pope WB, Geist C, et al. 30 -Deoxy-30 -18F-fluorothymidine PET and MRI for early survival predictions in patients with recurrent malignant glioma treated with bevacizumab. J Nucl Med 2012;53:29–36. [124] [18F]Fluciclatide PET imaging of pazopanib response. Clinicaltrials.gov Identifier NCT01961583. [125] Battle MR, Goggi JL, Allen L, Barnett J, Morrison MS. Monitoring tumour response to antiangiogenic therapy with 18F-fluciclatide, an 18F labelled αvβ3-integrin imaging agent. J Nucl Med 2011;52:424–30. [126] Braak H, Braak E. Neuropathological stageing of Alzheimer-related changes. Acta Neuropathol 1991;82:239–59. [127] Efficacy and safety trial of MK-8931 in participants with prodromal Alzheimer’s disease (MK-8931-19) (APECS). Clinicaltrials.gov Identifier NCT01953601. [128] Effect of LY450139 on the long term progression of Alzheimer’s disease. Clinicaltrials. gov Identifier NCT00594568.
ARTICLE IN PRESS 50
James Nairne et al.
[129] Burn DJ, Mark MH, Playford ED, Maraganore DM, Zimmerman Jr TR, Duvoisin RC, et al. Parkinson’s disease in twins studied with 18F-dopa and positron emission tomography. Neurology 1992;42:1894–900. [130] Wulfert S, Kratochwil C, Choyke PL, Afshar-Oromieh A, Mier W, Kauczor H-U, et al. Multimodal imaging for early functional response assessment of 90 Y/177Lu-DOTATOC peptide receptor targeted radiotherapy with DW-MRI and 68Ga-DOTATOC-PET/CT. Mol Imaging Biol 2014;16:586–94. [131] Viale A, Reineri F, Dastru` W, Aime S. Hyperpolarized 13C-pyruvate magnetic resonance imaging in cancer diagnostics. Expert Opin Med Diagn 2012;6:335–45. [132] Ruiz-Cabello J, Barnett BP, Bottomley PA, Bulte JW. Fluorine (19F) MRS and MRI in biomedicine. NMR Biomed 2011;24:112–29.