Elsevier

Drug Discovery Today

Volume 27, Issue 1, January 2022, Pages 280-291
Drug Discovery Today

Post-screen (grey)
Positron emission tomographic imaging in drug discovery

https://doi.org/10.1016/j.drudis.2021.07.025Get rights and content

Highlights

  • PET imaging accelerates the selection of drug candidates during the early stages of development.

  • The estimation of receptor occupancy in the brain is one of the most important applications of PET.

  • PET is also widely used in pharmacokinetics, biodistribution and treatment monitoring.

  • There are several applications of PET imaging in oncological and cardiological drug discovery.

Abstract

Positron emission tomography (PET) is an extensively used nuclear functional imaging technique, especially for central nervous system (CNS) and oncological disorders. Currently, drug development is a lengthy and costly pursuit. Imaging with PET radiotracers could be an effective way to hasten drug discovery and advancement, because it facilitates the monitoring of key facets, such as receptor occupancy quantification, drug biodistribution, pharmacokinetic (PK) analyses, validation of target engagement, treatment monitoring, and measurement of neurotransmitter concentrations. These parameters demand careful analyses for the robust appraisal of newly formulated drugs during preclinical and clinical trials. In this review, we discuss the usage of PET imaging in radiopharmaceutical development; drug development approaches with PET imaging; and PET developments in oncological and cardiac drug discovery.

Introduction

The process of developing a new drug is expensive and lengthy. It has been estimated that the usual duration to advance a novel drug formulation from its conception to distribution in the market is 10 years and the projected cost is ~US$1.4 billion.1, 2 A longitudinal study on the length of drug development from 2000 to 2009 revealed that the average time taken for developing a new drug for CNS diseases is around 10 years. The same study showed the time of progression to be 5.1 years for antiviral drugs against AIDS and 7.6 years for anti-neoplastic drugs.3 This is a significant burden that not only weighs down the pharmaceutical industry, but also impacts an entire society. Although over the past two decades an increasing number of druggable targets have been identified as a result of rapid advancements in molecular biology, the number of approved drugs has not similarly increased.4 In addition, the development of new drugs for CNS disorders is imperative in the face of the growing incidence of such disorders, but hampered by our inability to fully map the progression of certain diseases. Reduced efficacy in drug development is also the result of our poor understanding of preclinical models.5 Moreover, the advancement of a CNS drug has several inherent impediments, such as inadequate access to the target organ, difficulties crossing the blood–brain barrier (BBB),6 and the challenges of being directly quantified in the brain. As a result of such issues, the typical cost of drug discovery is increasing and is likely to continue to do so. Consequently, approaches to expedite the drug discovery process and curb the rise in costs are being pursued.

The drug discovery process comprises several phases. Upon completion of the research phase of drug discovery, approval to examine new drug molecules in clinical subjects is acquired and a multistep clinical examination begins.7 The key intention of Phase I clinical trials is to assess drug safety in humans. It is also necessary to test a range of drug doses both above and below its intended dose, and to confirm that the drug candidate is well tolerated at its effective dosage. In this phase, the selected subjects are predominantly healthy volunteers (particularly in oncology). A focused patient population is required for Phase II clinical trials, during which the primary objective is to validate the safety and efficacy of the drug at its effective dose in the patient group. It is imperative to have positive data from Phase II trials before proceeding to the vital Phase III trials, the pivotal goal of which is to discern whether the new drug treatment can bring about considerable value and assess how the new formulation fares in relation to current drugs for the same disease.8 The length of the preclinical phase can change according to the diseases for which the drugs are purposed. Similarly, the duration of the clinical trial, inclusive of the approval steps, differs among the various drug categories.

It is anticipated that different imaging modalities, such as PET, could make an indispensable contribution to speed up the drug discovery process.9 Molecular imaging modalities could help in making decisions on effective dose administration and patient selection,10 and could also offer evidence regarding drug–target interactions in Phase I and phase II trials.11 By using molecular imaging techniques properly, the pharmaceutical industry can identify the leading compound in early stages. Compounds that do not successfully display qualified activity during the preliminary stages can be discarded, and the candidates that demonstrate substantial activity can be accelerated for further development. Using this approach, the drug discovery process could become more cost effective and less painstaking.

A relevant and non-invasive functional imaging technique that is applied widely in the clinic is PET imaging. In PET imaging, a biologically active molecule is labeled with a positron-emitting radiotracer. Ultimately, this analytical technique allows for the visualization and quantification of biochemical and physiological processes in vivo, and is predominantly used in neurology, oncology, and cardiology.12 The versatility of PET imaging gives it an edge over other functional imaging techniques. A diverse range of positron-emitting radionuclides, such as fluorine-18 (18F), carbon-11 (11C), nitrogen-13 (13N), and oxygen-11 (11O) are accessible. Moreover, owing to the distinct and variable characteristics of radionuclides, it is possible to select from a broad choice of radionuclides to label a wider range of biologically relevant compounds.2 Table 1 presents selected different 11C and 18F radioligands used for brain imaging.

As a marker of inflammation, translocator protein (TSPO), which is situated on the outer mitochondrial membrane, is upregulated on microglial cells.13 Neuroinflammation is characteristic of various neurodegenerative disorders, such as Alzheimer’s disease (AD) and Parkinson’s disease (PD), and can result in neuronal impairment. Although the PET tracer, [11C](R)-PK 11195, has been used as a selective antagonist for TSPO,14 it has several limitations, such as comparatively low brain permeability and enhanced lipophilicity, causing it to bind strongly to hydrophobic macromolecules. The dopamine and serotonin (5-HT) receptors and transporters are targets implicated in PD among other brain pathologies.5, 16 Among the tracers aimed at these targets, [11C]SCH 23390, [11C]Raclopride, [18F]Fallypride and [18F]FE-PE2I17, 18, 19 are well-established ones used to quantify parameters defining pharmacodynamic effects in response to drug treatment.20 Although antagonists for neuroreceptors have been primarily utilized as PET ligands, the usage of agonists forms a growing body of research be because they bind preferentially to the high-affinity state of the receptors.21 Similar to PD, numerous radiotracers have been developed for PET imaging of AD pathology. Amyloid β (Aβ)-directed radiotracers, such as the pioneering [11C]PIB and the further optimized [11C]AZD2184, and tau-targeted tracers, such as [11C]PBB3, show potential for marking their respective target biomolecules with high affinity and could provide a base for further improvement of their characteristics.22

PET imaging enables novel ‘precision pharmacology’, through which answers to whether new drugs are delivered to target tissues specifically in biologically effective dosages can be derived. This could significantly contribute to current understanding of drug discovery and development.20 Given the burgeoning costs associated with the clinical developmental stage of a drug, a well-planned experiment can address vital queries during the early stages when fewer human subjects are required, paving the way for more assertive and economic decision-making. Thus, PET imaging could yield additional significance in drug discovery, because the data obtained are translatable from bench to bedside. Such translatability is important for improving the efficiency of drug development and for applying it in a clinical setting. Moreover, it ensures that the costs of evaluating and advancing drugs in disease treatment are minimized.

The main application of PET in drug discovery is to evaluate receptor occupancy over a range of dosages, thus making it useful to ascertain the optimal dosage of the drug.23 Moreover, PET can be used to study drug PK, biodistribution, target validation, and disease monitoring.24, 25 It also aids our understanding of several important parameters, including the identification of possible drug metabolism its early stages of development, and the binding between the labeled radiometabolites and their respective receptors.26 PET imaging also provides important information on whether the radiometabolites are capable of crossing the BBB.27 The slow metabolism of the radiotracers suggests a long half-life and a comparatively high plasma concentration of the drug, which then offers valuable knowledge of the approximate dosage required for patients. Furthermore, PET is beneficial for selecting highly responsive patient populations for clinical trials and monitoring patient pathology in a longitudinal manner after drug treatment. This is because, through PET imaging, it is convenient to perform studies earlier and with a smaller patient sample size, which aids in identifying patient populations who might respond the best and benefit the most from treatment.28 Therefore, it would be prudent to implement PET imaging strategies more frequently during the early phases of drug development. The various applications of PET imaging in drug discovery are illustrated in Fig. 1. In this review, we mainly discuss the utilization of PET imaging in drug discovery based on the assessable PK and pharmacodynamics parameters. Although we focus on PET imaging in the discovery of drugs for CNS diseases, we also introduce its usage in oncological and cardiac diseases.

Section snippets

Approaches of drug development with PET

There are two primary approaches for drug development with PET29 (Fig. 2). The most common strategy is to use an unlabeled drug to inhibit the specific binding of a well-established PET radioligand. In the alternative approach, a drug molecule is labeled with a PET radioisotope to understand its anatomical distribution and binding to specific targets.

In the first approach, an unlabeled drug competes with a radioligand to occupy a receptor system,30 revealing, for example, whether the

Receptor occupancy by PET

Receptor occupancy has been calculated to be the percentage of the receptor population that is engaged by an unlabeled drug.33 The best-known usage of PET is for the examination of antipsychotic drug binding.34 Receptor occupancy can be determined either by the well-established indirect approach (discussed earlier) or by the direct approach. The receptor occupancy study of a radiolabeled drug provides several key pieces of information, such as the reachability of the drug to its intended

PET imaging in drug biodistribution studies

Biodistribution studies are essential during initial-phase drug discovery, because they confirm whether the drug can reach the target tissue, and whether it has a propensity to accumulate in nontarget sites or have a negative effect on target sites, which could suggest possible toxicity.20 For example, if a drug molecule is unable to cross the BBB and penetrate the brain in sufficient quantity, the preferred effect in the CNS will not be achieved.42 PET scans can monitor the concentration of

PET imaging and pharmacokinetic studies

The essential PK information of a drug can be easily acquired by PET imaging when the drug molecule can itself be radiolabeled.48 The relevant PK data can be quantified from whole-body biodistribution data using mathematical/statistical modeling and information about the molar activity of the radiopharmaceutical. By taking into account the molar activity in conjunction with the mean injected mass, the mean injected activity can be derived to be used in biodistribution studies. Given its

Validation of target engagement by PET imaging

When a PET radiotracer and a drug molecule share the same target, the PET technique can be applied to check any interaction of the candidate drug with its target. [11C]AZ10419369 was recently used as a suitable radioligand for the quantification of in vivo 5-HT1B receptor binding.51, 52 Currently, this radiotracer is being used as a candidate to quantify the receptor occupancy effected by compounds that target the 5-HT1B receptor. Serotonin binding to the 5-HT1B receptor reduces the synaptic

Drug treatment monitoring by PET imaging

One of the most important translations of PET imaging in drug discovery is to track the progression of diseases, which alters the treatment course of therapeutic candidates, such as the drug development process for Alzheimer’s disease (AD). According to the β-amyloid (Aβ) cascade hypothesis, the pathological hallmark of AD is the development of insoluble amyloid plaques, incorporated mainly because of the aggregation of Aβ in the brains of patients58; a key aim for AD drug development is to

Quantification of neurotransmitter concentration by PET imaging

PET has also been used to quantify the changes in endogenous neurotransmitter concentrations.62 In accordance with the competition model, radiotracer binding to a neuroreceptor decreases after an increase in the neurotransmitter concentration. The serotonin system has a significant role in pathophysiology, as well as in the treatment of psychiatric disorders.63 Hence, it is important to develop a PET imaging tracer that would allow the quantification of serotonin release in the brain. As

Use of PET imaging in the discovery of drug for oncological disease

Apart from having a vital role in CNS diseases, the importance of PET imaging in the drug discovery for oncological diseases should be noted. PET with 2-[18F]fluoro-2-deoxyglucose (FDG-PET) is often used as an imaging accessory in the clinical setting.65 Its use is effective when monitoring therapy response in lymphomas, providing this method with an edge over other imaging modalities.66 Interim PET imaging in patients after a fixed time after drug treatment administration allows for the

Usage of PET imaging in the drug discovery of cardiac diseases

In contrast to the widespread usage of PET imaging in the drug development course of CNS and oncological diseases, such as breast and lung cancer, its applications in cardiac disease remain relatively sparse. Nevertheless, studies suing PET imaging as an aid in the drug discovery of cardiac diseases are beginning to emerge. Heart failure has been long correlated with sympathetic activation, and a reduced neuronal noradrenaline (NA) transporter (NET) function and NA concentration in the heart.76

Concluding remarks

The use of PET is mostly centered in the fields of CNS and oncology-based therapeutics development and, to a smaller extent, in the cardiac disorders. It has the potential to be used in drug discovery and development, including investigating drug biodistribution, plasma binding, absorption, distribution, metabolism, metabolic clearance, and effective dosing. From a clinical point of view, PET imaging has been used for a long time to diagnose and track disease in a quantitative manner. Hence,

Acknowledgments

Authors acknowledge support from Lee Kong Chian School of Medicine, NTU Austrian Institute of Technology and Medical University of Vienna internal grant (NAM/15006), the LKC Medicine Imaging Probe Development Platform, Singapore, and the Cognitive Neuroimaging Centre (CONIC) at Nanyang Technological University, Singapore.

References (83)

  • C.P. Adams et al.

    Estimating the cost of new drug development: is it really $802 million?

    Health Aff

    (2006)
  • C. Halldin et al.

    Brain radioligands-–state of the art and new trends

    Quart J Nucl Med Mol Imaging

    (2001)
  • K.I. Kaitin et al.

    Pharmaceutical innovation in the 21st century: new drug approvals in the first decade, 2000–2009

    Clin Pharmacol Ther

    (2011)
  • FDA. Innovation or stagnation? Challenge and opportunity on the critical path to new medical products....
  • National Academies of Sciences and Medicine. Therapeutic Development in the Absence of Predictive Animal Models of...
  • D.J. Begley

    Delivery of therapeutic agents to the central nervous system: the problems and the possibilities

    Pharmacol Ther

    (2004)
  • T. Takebe et al.

    The current status of drug discovery and development as originated in United States academia: the influence of industrial and academic collaboration on drug discovery and development

    Clin Transl Sci

    (2018)
  • C.A. Umscheid et al.

    Key concepts of clinical trials: a narrative review

    Postgrad Med

    (2011)
  • E.G. de Vries et al.

    Toward molecular imaging-driven drug development in oncology

    Cancer Discovery

    (2011)
  • A.R. Pantel et al.

    Molecular imaging to guide systemic cancer therapy: Illustrative examples of PET imaging cancer biomarkers

    Cancer Lett

    (2017)
  • L. Cunha et al.

    The role of molecular imaging in modern drug development

    Drug Discovery Today

    (2014)
  • H. Schoder et al.

    PET/CT: a new imaging technology in nuclear medicine

    Eur J Nucl Med Mol Imaging

    (2003)
  • S. Lavisse et al.

    Reactive astrocytes overexpress TSPO and are detected by TSPO positron emission tomography imaging

    J Neurosci

    (2012)
  • D.R. Owen et al.

    Imaging brain microglial activation using positron emission tomography and translocator protein-specific radioligands

    Int Rev Neurobiol

    (2011)
  • C. Güzey et al.

    Radioligand binding to brain dopamine and serotonin receptors and transporters in Parkinson’s disease: relation to gene polymorphisms

    Int J Neurosci

    (2012)
  • J.O. Rinne et al.

    A post-mortem study on striatal dopamine receptors in Parkinson’s disease

    Brain Res

    (1991)
  • P.H. Elsinga et al.

    PET tracers for imaging of the dopaminergic system

    Curr Med Chem

    (2006)
  • C. Halldin et al.

    Preparation of 11C-labelled SCH 23390 for the in vivo study of dopamine D-1 receptors using positron emission tomography

    Int J Radiation Appl Instrum Part A Appl Radiat Isotopes

    (1986)
  • V.S. Kerstens et al.

    Reliability of dopamine transporter PET measurements with [18F] FE–PE2I in patients with Parkinson’s disease

    EJNMMI Res

    (2020)
  • P.M. Matthews et al.

    Positron emission tomography molecular imaging for drug development

    Br J Clin Pharmacol

    (2012)
  • L. Lemoine et al.

    Radiosynthesis and preclinical evaluation of 18F–F13714 as a fluorinated 5-HT1A receptor agonist radioligand for PET neuroimaging

    J Nucl Med

    (2012)
  • M.D. Ikonomovic et al.

    Post-mortem correlates of in vivo PiB-PET amyloid imaging in a typical case of Alzheimer’s disease

    Brain

    (2008)
  • Y. Zhang et al.

    PET imaging for receptor occupancy: meditations on calculation and simplification

    J Biomed Res

    (2012)
  • M.M. Moein et al.

    Sample preparation techniques for radiometabolite analysis of positron emission tomography radioligands; trends, progress, limitations and future prospects

    Trends Anal Chem

    (2019)
  • K.K. Ghosh et al.

    Dealing with PET radiometabolites

    EJNMMI Res

    (2020)
  • C. Halldin et al.

    Radioligand disposition and metabolism—key information in early drug development

    Develop Nucl Med

    (1995)
  • V.W. Pike

    PET radiotracers: crossing the blood–brain barrier and surviving metabolism

    Trends Pharmacol Sci

    (2009)
  • N.M. Scheinin et al.

    Amyloid imaging as a surrogate marker in clinical trials in Alzheimer’s disease

    Quart J Nucl Med Mol Imaging

    (2011)
  • C. Halldin et al.

    PET studies with carbon-11 radioligands in neuropsychopharmacological drug development

    Curr Pharm Des

    (2001)
  • G. Vauquelin

    Determination of drug–receptor residence times by radioligand binding and functional assays: experimental strategies and physiological relevance

    MedChemComm

    (2012)
  • A. Lindberg et al.

    Potential for imaging the high-affinity state of the 5–HT 1B receptor: a comparison of three PET radioligands with differing intrinsic activity

    EJNMMI Res

    (2019)
  • P. Hein et al.

    Receptor and binding studies

  • L. Farde

    The advantage of using positron emission tomography in drug research

    Trends Neurosci

    (1996)
  • R. Arakawa et al.

    PET technology for drug development in psychiatry

    Neuropsychopharmacol Rep

    (2020)
  • E. Kim et al.

    Predicting brain occupancy from plasma levels using PET: superiority of combining pharmacokinetics with pharmacodynamics while modeling the relationship

    J Cereb Blood Flow Metab

    (2012)
  • L. Pani et al.

    Antipsychotic efficacy: relationship to optimal D2-receptor occupancy

    Eur Psych

    (2007)
  • S. Kapur et al.

    The newer antipsychotics: underlying mechanisms and the new clinical realities

    Curr Opin Psych

    (2004)
  • S. Nyberg et al.

    Suggested minimal effective dose of risperidone based on PET-measured D2 and 5-HT2A receptor occupancy in schizophrenic patients

    Am J Psychiatry

    (1999)
  • F. Yokoi et al.

    Dopamine D 2 and D 3 receptor occupancy in normal humans treated with the antipsychotic drug aripiprazole (OPC 14597): a study using positron emission tomography and [11 C] raclopride

    Neuropsychopharmacology

    (2002)
  • A. Lingford-Hughes et al.

    A proof-of-concept study using [11 C] flumazenil PET to demonstrate that pagoclone is a partial agonist

    Psychopharmacology

    (2005)
  • B. Andree et al.

    Positron emission tomographic analysis of dose-dependent MDL 100,907 binding to 5-hydroxytryptamine-2A receptors in the human brain

    J Clin Psychopharmacol

    (1998)
  • Cited by (18)

    • Blood-brain barrier permeability following conventional photon radiotherapy – A systematic review and meta-analysis of clinical and preclinical studies

      2022, Clinical and Translational Radiation Oncology
      Citation Excerpt :

      The effect of RT on the BBB in patients can be studied in more detail and longitudinally during and after radiotherapy, using advanced MRI and PET studies. Drug imaging with PET after RT, will provide more insight on possible RT-induced enhancement of drug delivery to the brain, avoiding toxicity and optimizing concomitant and adjuvant treatment strategies for an optimal therapeutic index [119]. The review has been supported by the KWF Young Investigator Award (KWF 10911, Dr. D.G. van Vuurden.

    • Why 90% of clinical drug development fails and how to improve it?

      2022, Acta Pharmaceutica Sinica B
      Citation Excerpt :

      This hypothesis believes that only free unbound drug from plasma (not plasma protein bound drug) can distribute to disease-targeted tissues to interact with its molecular target; while free drug exposure in the plasma would be similar to the disease-targeted tissues at steady state41,42; and thus drug exposure in the plasma can be used to predict the pharmacodynamic effect of the drug candidate. However, this “free drug” hypothesis may only apply to a limited class of drug candidates but not applicable to many other compounds since many factors can cause an asymmetric free drug distribution between plasma and tissue42–64. Therefore, drug exposure in the plasma, without knowing the exposure in disease-targeted tissue/normal tissues, may mislead the selection of drug candidate to clinical trials51–53,65.

    • Structure‒tissue exposure/selectivity relationship (STR) correlates with clinical efficacy/safety

      2022, Acta Pharmaceutica Sinica B
      Citation Excerpt :

      This hypothesis believes that only free unbound drug from plasma (not plasma protein bound drug) can distribute to disease-targeted tissues to interact with its molecular target; while free drug concentration in the plasma and in the disease-targeted tissues would be similar at steady state12,13, and thus drug exposure in the plasma can be used as surrogate to predict the pharmacodynamic effect of the drug candidate. However, this “free drug hypothesis” may only apply to a limited class of drug candidates but not applicable to many other compounds since many factors can cause an asymmetric free drug distribution between plasma and tissue4,10,13–34. This hypothesis also completely ignores the active transport of plasma proteins themselves from systemic circulations to all tissues that also contributes to drug exposure in the tissues4,16,17.

    View all citing articles on Scopus
    View full text