Elsevier

European Journal of Cancer

Volume 38, Issue 16, November 2002, Pages 2094-2107
European Journal of Cancer

PET for in vivo pharmacokinetic and pharmacodynamic measurements

https://doi.org/10.1016/S0959-8049(02)00413-6Get rights and content

Abstract

Positron emission tomography (PET) scanning is evolving as a unique tool for drug development in oncology for improving both the efficacy of established treatment and in evaluating novel anticancer agents. As a non-invasive functional imaging modality, PET has an unrivalled sensitivity when monitoring the pharmacokinetics and pharmacodynamics of drugs and biochemicals when radiolabelled with short living positron-emitting radioisotopes. This is of particular relevance in assessing newer molecular-targeted therapy where conventional evaluation criteria (maximum tolerated dose and tumour shrinkage for example) may be inappropriate. PET has already been applied to a wide number of drugs to demonstrate activity in vivo from standard chemotherapy such as 5-fluorouracil (5-FU) [J Clin Oncol 17 (1999) 1580], to novel molecular agents such as those involved in tumour angiogenesis [Br J Cancer 83 (2000) P6] and antivascular therapy [Proc Annu Meet Am Soc Clin Oncol 19 (2000) 179a]. This review will evaluate the achievements of PET in the drug development process, an approach that promises to facilitate the rapid translation of scientific research into current clinical practice.

Introduction

The field of oncology has undergone a revolution in drug discovery and therapeutic approaches led primarily through an increased understanding of genetic and molecular mechanisms in cancer evolution. Correspondingly, the previously held empirical random screening method in oncological drug development is being gradually replaced with the concept of rational drug design. This process identifies specific targets responsible for malignant cellular transformation and more importantly, drugs that can overcome them, rather than simply using the antiproliferative activity of a drug as an endpoint. New agents discovered for possible use as anticancer therapy include genes, proteins, growth factors and receptors, and those involved in specific pathways, for example, angiogenesis, signal transduction, cell cycle, cell apoptosis, invasion, metastasis, drug resistance and blood flow. Further expansion of drug development has resulted from combinatorial chemistry, another new technology that has grown at a tremendous rate to produce a plethora of novel therapeutic targets [4]. However, drug development is still a protracted and expensive business. It is estimated that of 5000 possible drugs screened, only one drug is successfully approved and introduced into the market [5]. In addition, the candidate drug finally selected takes an average of 10 years of development and requires an investment of several million pounds before eventually proceeding to clinical trial [6]. Consequently, there is a need for an additional method of evaluating potential new drugs and to optimise existing treatment strategies. There is an increasing realisation that radiotracer drug imaging as used in positron emission tomography (PET) has a major role to play in expediting drug development both in the clinical and preclinical setting, and which may in addition reduce the substantial costs currently incurred.

The objectives of drug development in oncology are clearly defined. The aims involve the determination of optimum delivery of drug to the site of the tumour with minimum exposure to normal tissue, thereby achieving maximum therapeutic benefit. This requires accurate monitoring of drug pharmacology including pharmacokinetics (absorption, distribution, metabolism and elimination) and pharmacodynamics (tumour response, enzyme induction/inhibition receptor binding, tolerance, etc.). At present, these parameters are measured by analysis of blood and urine samples and occasionally using biopsy specimens of relevant tissues. Whilst this strategy is suitable for certain anticancer drugs, plasma monitoring may be irrelevant with the next generation of therapies such as anti-angiogenesis, where treatment is designed to target specific tissues. In this situation, the availability of a positron emitting radiotracer to quantitate drug delivery and response in the relevant tissue can significantly advance our knowledge of drug pharmacology. The ability of PET to meet the challenges and demands of drug development relies on a multidisciplinary team consisting of radiochemists, biologists, mathematical modellers, pharmacologists and clinicians.

Section snippets

Radiotracers

There are many positron emitting isotopes available for commonly occurring elements including carbon, oxygen, nitrogen and fluorine with half-lives ranging from seconds to several days (Table 1). They are usually made in a cyclotron, although some (copper 62) can be manufactured in a nuclear generator. Radiotracers are produced after replacement of a molecule in a compound of interest with a radionuclide without modifying their pharmaceutical, biological or biochemical properties. For example,

Acquisition and modelling of PET images

A 3D image of the distribution of the radiolabelled drug within the body comes from the simultaneous detection of two gamma rays from the decay of the positron emitting radionuclide. The two 511 keV gamma rays are emitted at approximately 180° and are each recorded coincidentally by rings of external nuclear detectors, with the inference that the decay event occurred between the opposing two detectors. Data are obtained as sinograms that are reconstructed into tomographic images after

PET and drug evaluation

There are a number of generic issues in drug development that can be addressed by PET imaging. These can be described in the form of questions: (1) does the drug sufficiently distribute to target (tumour) and how much goes to normal tissue, where toxicity may be produced, (2) how is the drug eliminated from tumour and normal tissue, (3) does the drug modulate its target in a predictable way, (4) is the drug efficacious. Points (1) and (2) can be classified as pharmacokinetic studies, and (3)

Summary

Advances in technology have made in vivo assessment of physiological and biochemical processes in humans a reality. This has been achieved by the integrated work of a multidisciplinary team within a PET department. PET is an invaluable tool in the preclinical assessment of drugs prior to further development. Clinical assessment of drugs can help elucidate mechanisms of action, and resistance, at tracer doses. Newer therapies can be investigated where conventional methods of drug analysis will

United references

Refs. 29, 33 are not cited.

References (120)

  • F.L. Flanagan et al.

    PET in breast cancer

    Semin. Nucl. Med.

    (1998)
  • U.A. Germann et al.

    P-glycoproteinamediators of multidrug resistance

    Semin. Cell Biol.

    (1993)
  • R.A.H. Harte et al.

    Tumour, normal tissue, and plasma pharmacokinetic studies of fluorouracil biomodulation with N-phosphacetyl-L-aspartate, folinic acid, and interferon alfa

    J. Clin. Oncol.

    (1999)
  • V.A. Carroll et al.

    Imaging vascular endothelial growth factor in vivo with positron emission tomography

    Br. J. Cancer

    (2000)
  • H. Anderson et al.

    Measurement of tumour and normal tissue (NT) perfusion by positron emission tomography (PET) in the evaluation of antivascular therapyresults in the phase1 study of Combretastatin A4 Phosphate (CA4P)

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (2000)
  • K.A. Leonard et al.

    Combinatorial chemistry in cancer drug development

    Cancer J.

    (2001)
  • J.L. Tatum et al.

    Imaging drug development

    Acad. Radiol.

    (2000)
  • B. Cambell

    Drug development and positron emission tomography

  • M.A. Mintun et al.

    Breast CancerPET imaging of oestrogen receptors

    Radiology

    (1988)
  • S.C. Huang et al.

    Principles of tracer kinetic modelling in positron tomography

  • R.N. Gunn et al.

    A general method to correct PET data for tissue metabolites using a dual-scan approach

    J. Nucl. Med.

    (2000)
  • R.G. Blasberg et al.

    Imaging brain tumour proliferative activity with [124I]iododeoxyuridine

    Cancer Res.

    (2000)
  • V.J. Cunningham et al.

    Spectral analysis of dynamic PET studies

    J. Cereb. Blood Flow Metab.

    (1993)
  • C.S. Patlak et al.

    Graphical evaluation of blood-to-brain transfer constant from multiple-time uptake data

    J. Cereb. Blood Flow Metab.

    (1983)
  • W.K. Yung et al.

    A phase 11 study of temozolomide vs. procarbazine in patients with glioblastoma multiforme at first relapse

    Br. J. Cancer

    (2000)
  • A.A. Brandes et al.

    Temozolomide in patients with high grade glioma

    Oncology

    (2000)
  • M.D. Prados

    Future directions in the treatment of malignant gliomas with temozolomide

    Semin. Oncol.

    (2000)
  • M. Bower et al.

    Multicentre CRC phase 11 trial of temozolomide in recurrent or progressive high grade gliomas

    Cancer Chemother. Pharmacol.

    (1997)
  • M.R. Middleton et al.

    Randomised phase 111 study of temozolomide vs. dacarbazine in the treatment of patients with advanced metastatic melanoma

    J. Clin. Oncol.

    (2000)
  • S.S. Agarwala et al.

    Temozolomide, a novel alkylating agent with activity in the central nervous system, may improve the treatment of advanced metastatic melanoma

    Oncologist

    (2000)
  • N.M. Bleehan et al.

    CRC phase 11 trial of temozolomide metastatic melanoma

    J. Clin. Oncol.

    (1995)
  • E.J. Estlin et al.

    Phase 1 study of temozolomide in paediatric patients with advanced cancer. UK Children's Cancer Study Group

    Br. J. Cancer

    (1998)
  • H.S. Nicholson et al.

    Phase 1 study of temozolomide in children and adolescents with recurrent solid tumoursa report from the Children's Cancer Group

    J. Clin. Oncol.

    (1998)
  • P.J. Woll et al.

    Phase 11 trial of temozolomide in low-grade non-Hodgin's lymphoma

    Br. J. Cancer

    (1995)
  • J.P. Van Brussel et al.

    A phase 11 study of temozolomide in hormone-refractory prostate cancer

    Cancer Chemother. Pharmacol.

    (2000)
  • M.J. Moore et al.

    A phase 11 study of temozolomide in advanced untreated pancreatic cancer

    Invest. New Drugs

    (1998)
  • A.T. Chan et al.

    Phase 11 study of Temodal in the treatment of patients with advanced nasopharyngeal carcinoma

    Cancer Chemother. Pharmacol.

    (1998)
  • M.F. Stevens et al.

    From triazines and triazenes to temozolomide

    Eur. J. Cancer

    (1996)
  • L.L. Tsang et al.

    Comparisom of the cytotoxicity of temozolomide and dacarbazine, prodrugs of 3-methyl-(triazen-1-yl)imidazole-4-carboxamide

    Cancer Chemother. Pharmacol.

    (1991)
  • G.D. Brown et al.

    Synthesis of [11C-methyl]methylisocyanate and application of microwave heating to label the novel anticancer agent temozolomide

    J. Label Radiopharm.

    (1994)
  • S.R. Meikl et al.

    Pharmacokinetic assessment of novel anti-cancer drugs using spectral analysis and positron emission tomographya feasibility study

    Cancer Chemother. Pharmacol.

    (1998)
  • C.S. Brock et al.

    The kinetic behaviour of temozolomide in man

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (1996)
  • C.S. Brock et al.

    Temozolomide uptake in human astrocytomas demonstrated in vivo

    Proc. Annu. Meet. Am. Soc. Clin. Oncol.

    (1997)
  • C.S. Brock et al.

    Response to temozolomide (TEM) in recurrent high grade gliomas (HGG) is related to drug concentration

    Annals. Oncol.

    (1998)
  • G.J. Atwell et al.

    Potential antitumour agents50. In vivo solid-tumour activity of derivatives of N-[2-(dimethylamino)ethyl]acridine-4-carboxamide

    J. Med. Chem.

    (1987)
  • G.J. Finlay et al.

    In vitro assessment of N-[2-(dimethylamino)ethyl]acridine-4-carboxamide, a DNA-intercalating antitumour drug with reduced sensitivity to multidrug resistance

    Cancer Chemother. Pharmacol.

    (1993)
  • B.C. Baguley et al.

    Design of DNA intercalators to overcone topoisomerase 2-mediated multidrug resistance

    J. Natl. Cancer Inst.

    (1990)
  • B.C. Baguley et al.

    Resisitance mechanisms to topoisomerase poisonsthe application of cell culture methods

    Oncol. Res.

    (1992)
  • B.C. Baguley et al.

    Selectivity of N-[2-(dimethylamino)ethyl]acridine-4-carboxamide towards Lewis lung carcinoma and human tumoue cell lines in vitro

    Eur. J. Cancer Clin. Onc.

    (1989)
  • F. Brady et al.

    Automated radiosynthesis of [6-O-Methyl-11C]buprnorphine from 3-O-trityl protected precursors

    Appl. Radiat. Isot.

    (1994)
  • Cited by (103)

    • <sup>64</sup>Cu-labeling of small extracellular vesicle surfaces via a cross-bridged macrocyclic chelator for pharmacokinetic study by positron emission tomography imaging

      2022, International Journal of Pharmaceutics
      Citation Excerpt :

      Therefore, imaging techniques without these limitations are required for a more detailed analysis of sEV pharmacokinetics. With a temporal resolution of seconds, positron emission tomography (PET) is one of the most promising alternative imaging technologies (Gupta et al., 2002; Massoud and Gambhir, 2003). Non-clinical and clinical pharmacokinetic studies using PET have been conducted for many small molecules and antibody drugs, and recent advances in positron-emitter labeling have led to applications in the pharmacokinetic evaluation of nucleic acid drugs, peptide drugs, nano-drug delivery systems (DDSs), cells, and bacteria (Bansal et al., 2015; Mukai et al., 2014; Mukai et al., 2019; Nomura et al., 2020; Ren et al., 2019; Sakai et al., 2019; Tamura et al., 2013).

    • Recent advances in lipid nanoparticles for delivery of nucleic acid, mRNA, and gene editing-based therapeutics

      2022, Drug Metabolism and Pharmacokinetics
      Citation Excerpt :

      Positron emission tomography (PET) is an imaging technique that can be used seamlessly from rodents to humans. Its application to pharmacokinetic research and drug development has been explored, owing to its high quantitative and temporal resolution [33,53–56]. The ability to directly evaluate drug concentrations in human tissues, which was previously impossible, is a breakthrough [53–56], and is expected to lead to efficient drug development that strongly links pharmacokinetics to drug efficacy.

    • Positron emission tomography as a noninvasive tool in pharmacokinetic studies

      2021, Biopharmaceutics and Pharmacokinetics Considerations: Volume 1 in Advances in Pharmaceutical Product Development and Research
    • Listening for the therapeutic window: Advances in drug delivery utilizing photoacoustic imaging

      2019, Advanced Drug Delivery Reviews
      Citation Excerpt :

      The clinical utility of these photothermal/photodynamic agents could be local administration or systemic administration to target a disease site followed by activation and treatment monitoring using near infrared excitation via photoacoustic imaging [61]. The clinical state of the art for imaging pharmacokinetics is PET/CT with drug conjugated or co-loaded radiotracers [62]. In the preclinical space, photoacoustic imaging is being used for monitoring drug release and localized measurements of delivery.

    View all citing articles on Scopus
    View full text