1
|
The Unique Pharmacometrics of Small Molecule Therapeutic Drug Tracer Imaging for Clinical Oncology. Cancers (Basel) 2020; 12:cancers12092712. [PMID: 32971780 PMCID: PMC7563483 DOI: 10.3390/cancers12092712] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/11/2020] [Accepted: 09/17/2020] [Indexed: 12/30/2022] Open
Abstract
Simple Summary New clinical radiology scans using trace amounts of therapeutic cancer drugs labeled with radioisotope injected into patients can provide oncologists with fundamentally unique insights about drug delivery to tumors. This new application of radiology aims to improve how cancer drugs are used, towards improving patient outcomes. The article reviews published clinical research in this important new field. Abstract Translational development of radiolabeled analogues or isotopologues of small molecule therapeutic drugs as clinical imaging biomarkers for optimizing patient outcomes in targeted cancer therapy aims to address an urgent and recurring clinical need in therapeutic cancer drug development: drug- and target-specific biomarker assays that can optimize patient selection, dosing strategy, and response assessment. Imaging the in vivo tumor pharmacokinetics and biomolecular pharmacodynamics of small molecule cancer drugs offers patient- and tumor-specific data which are not available from other pharmacometric modalities. This review article examines clinical research with a growing pharmacopoeia of investigational small molecule cancer drug tracers.
Collapse
|
2
|
Li J, Kim S, Shields AF, Douglas KA, McHugh CI, Lawhorn-Crews JM, Wu J, Mangner TJ, LoRusso PM. Integrating Dynamic Positron Emission Tomography and Conventional Pharmacokinetic Studies to Delineate Plasma and Tumor Pharmacokinetics of FAU, a Prodrug Bioactivated by Thymidylate Synthase. J Clin Pharmacol 2017; 56:1433-1447. [PMID: 27095537 DOI: 10.1002/jcph.751] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 04/12/2016] [Accepted: 04/13/2016] [Indexed: 11/05/2022]
Abstract
FAU, a pyrimidine nucleotide analogue, is a prodrug bioactivated by intracellular thymidylate synthase to form FMAU, which is incorporated into DNA, causing cell death. This study presents a model-based approach to integrating dynamic positron emission tomography (PET) and conventional plasma pharmacokinetic studies to characterize the plasma and tissue pharmacokinetics of FAU and FMAU. Twelve cancer patients were enrolled into a phase 1 study, where conventional plasma pharmacokinetic evaluation of therapeutic FAU (50-1600 mg/m2 ) and dynamic PET assessment of 18 F-FAU were performed. A parent-metabolite population pharmacokinetic model was developed to simultaneously fit PET-derived tissue data and conventional plasma pharmacokinetic data. The developed model enabled separation of PET-derived total tissue concentrations into the parent drug and metabolite components. The model provides quantitative, mechanistic insights into the bioactivation of FAU and retention of FMAU in normal and tumor tissues and has potential utility to predict tumor responsiveness to FAU treatment.
Collapse
Affiliation(s)
- Jing Li
- Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Seongho Kim
- Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Anthony F Shields
- Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kirk A Douglas
- Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Christopher I McHugh
- Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jawana M Lawhorn-Crews
- Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jianmei Wu
- Karmanos Cancer Institute, Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Thomas J Mangner
- Department of Radiology, Wayne State University, Detroit, MI, USA
| | | |
Collapse
|
3
|
Saleem A, Murphy P, Plisson C, Lahn M. Why are we failing to implement imaging studies with radiolabelled new molecular entities in early oncology drug development? ScientificWorldJournal 2014; 2014:269605. [PMID: 25202719 PMCID: PMC4151371 DOI: 10.1155/2014/269605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 07/18/2014] [Indexed: 11/18/2022] Open
Abstract
In early drug development advanced imaging techniques can help with progressing new molecular entities (NME) to subsequent phases of drug development and thus reduce attrition. However, several organizational, operational, and regulatory hurdles pose a significant barrier, potentially limiting the impact these techniques can have on modern drug development. Positron emission tomography (PET) of radiolabelled NME is arguably the best example of a complex technique with a potential to deliver unique decision-making data in small cohorts of subjects. However, to realise this potential the impediments to timely inclusion of PET into the drug development process must be overcome. In the present paper, we discuss the value of PET imaging with radiolabelled NME during early anticancer drug development, as exemplified with one such NME. We outline the multiple hurdles and propose options on how to streamline the organizational steps for future studies.
Collapse
Affiliation(s)
- Azeem Saleem
- Imanova Ltd., Centre for Imaging Sciences, Imperial College Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Philip Murphy
- GlaxoSmithKline Global Imaging Unit, Stockley Park West, 1-3 Ironbridge Road, Uxbridge, Middlesex UB11 1BT, UK
| | - Christophe Plisson
- Imanova Ltd., Centre for Imaging Sciences, Imperial College Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - Michael Lahn
- Early Phase Oncology Clinical Investigation, Eli Lilly Corporate Center, Building 31/4, 893 S. Delaware Street, Indianapolis, IN 46285, USA
| |
Collapse
|
4
|
|
5
|
van der Veldt AAM, Smit EF, Lammertsma AA. Cancer therapy: could a novel test predict the amount of drug that reaches its target? Expert Rev Anticancer Ther 2013; 13:377-9. [PMID: 23560831 DOI: 10.1586/era.13.13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
6
|
van der Veldt AAM, Smit EF, Lammertsma AA. Positron Emission Tomography as a Method for Measuring Drug Delivery to Tumors in vivo: The Example of [(11)C]docetaxel. Front Oncol 2013; 3:208. [PMID: 23986880 PMCID: PMC3742054 DOI: 10.3389/fonc.2013.00208] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Accepted: 07/30/2013] [Indexed: 01/26/2023] Open
Abstract
Systemic anticancer treatments fail in a substantial number of patients. This may be caused by inadequate uptake and penetration of drugs in malignant tumors. Consequently, improvement of drug delivery to solid tumors may enhance its efficacy. Before evaluating strategies to enhance drug uptake in tumors, better understanding of drug delivery to human tumors is needed. Positron emission tomography (PET) is an imaging technique that can be used to monitor drug pharmacokinetics non-invasively in patients, based on radiolabeling these drugs with short-lived positron emitters. In this mini review, principles and potential applications of PET using radiolabeled anticancer drugs will be discussed with respect to personalized treatment planning in oncology. In particular, it will be discussed how these radiolabeled anticancer drugs could help to develop strategies for improved drug delivery to solid tumors. The development and clinical implementation of PET using radiolabeled anticancer drugs will be illustrated by validation studies of carbon-11 labeled docetaxel ([(11)C]docetaxel) in lung cancer patients.
Collapse
Affiliation(s)
- Astrid A M van der Veldt
- Department of Internal Medicine, VU University Medical Center , Amsterdam , Netherlands ; Department of Radiology and Nuclear Medicine, VU University Medical Center , Amsterdam , Netherlands
| | | | | |
Collapse
|
7
|
Abstract
Cancer treatment strategies have changed considerably over the past two decades, with increasing emphasis on cancer-specific biological therapies. This situation has led to the incorporation of biomarkers, including those obtained by medical imaging, into trial designs to better understand mechanisms of action and, hopefully, to provide early evidence of treatment efficacy at a molecular or physiological level. Unlike blood tests and tissue samples, an imaging biomarker allows assessment of treatment in the whole tumor, in all tumors in the body, and at multiple time points. This situation has increased the complexity of clinical trials, as each imaging modality has issues related to cost, ease of use, patient compatibility, data analysis, and interpretation. This article reviews strengths and limitations of the current imaging methods available in clinical cancer trials, including MRI, CT, PET, and ultrasonography. The information gained by each test, and the difficulties in acquiring the data and interpreting it are also discussed in order to help researchers plan imaging in clinical trials and interpret data from such studies.
Collapse
|
8
|
Abstract
The emphasis in cancer drug development has shifted from cytotoxic, non-specific chemotherapies to molecularly targeted, rationally designed drugs promising greater efficacy and less side effects. Nevertheless, despite some successes drug development remains painfully slow. Here, we highlight the issues involved and suggest ways in which this process can be improved and expedited. We envision an increasing shift to integrated cancer research and biomarker-driven adaptive and hypothesis testing clinical trials. The goal is the development of specific cancer medicines to treat the individual patient, with treatment selection being driven by a detailed understanding of the genetics and biology of the patient and their cancer.
Collapse
Affiliation(s)
- J S de Bono
- The Institute of Cancer Research, Cotswold Road, Sutton, Surrey SM2 5NG, UK.
| | | |
Collapse
|
9
|
Abstract
The development of novel molecularly targeted cancer therapeutics remains slow and expensive with many late-stage failures. There is an urgent need to accelerate this process by improving early clinical anticancer drug evaluation through modern and rational trial designs that incorporate predictive, pharmacokinetic, pharmacodynamic, pharmacogenomic and intermediate end-point biomarkers. In this article, we discuss current approaches and propose strategies that will potentially maximize benefit to patients and expedite the regulatory approvals of new anticancer drugs.
Collapse
Affiliation(s)
- Timothy A Yap
- Drug Development Unit, Royal Marsden NHS Foundation Trust, Downs Road, Sutton, Surrey SM2 5PT, United Kingdom
| | | | | | | |
Collapse
|
10
|
Saleem A, Price PM. Early tumor drug pharmacokinetics is influenced by tumor perfusion but not plasma drug exposure. Clin Cancer Res 2009; 14:8184-90. [PMID: 19088034 DOI: 10.1158/1078-0432.ccr-08-1324] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE Pharmacokinetic parameters derived from plasma sampling are used as a surrogate of tumor pharmacokinetics. However, pharmacokinetics-modulating strategies do not always result in increased therapeutic efficacy. Nonsurrogacy of plasma kinetics may be due to tissue-specific factors such as tumor perfusion. EXPERIMENTAL DESIGN To assess the impact of tumor perfusion and plasma drug exposure on tumor pharmacokinetics, positron emission tomography studies were done with oxygen-15 radiolabeled water in 12 patients, with 6 patients undergoing positron emission tomography studies with carbon-11 radiolabeled N-[2-(dimethylamino)ethyl]acridine-4-carboxamide and the other 6 with fluorine-18 radiolabeled 5-fluorouracil. RESULTS We found that tumor blood flow (mL blood/mL tissue/minute) was significantly correlated to early tumor radiotracer uptake between 4 and 6 minutes [standard uptake value (SUV)4-6; rho = 0.79; P = 0.002], tumor radiotracer exposure over 10 minutes [area under the time-activity curve (AUC)0-10; predominantly parent drug; rho = 0.86; P < 0.001], and tumor radiotracer exposure over 60 minutes (AUC0-60; predominantly radiolabeled metabolites; rho = 0.80; P = 0.002). Similarly, fractional volume of distribution of radiolabeled water in tumor (Vd) was significantly correlated with SUV4-6 (rho = 0.80; P = 0.002), AUC0-10 (rho = 0.85; P < 0.001), and AUC0-60 (rho = 0.66; P = 0.02). In contrast, no correlation was observed between plasma drug or total radiotracer exposure over 60 minutes and tumor drug uptake or exposure. Tumor blood flow was significantly correlated to Vd (rho = 0.69; P = 0.014), underlying the interdependence of tumor perfusion and Vd. CONCLUSIONS Tumor perfusion is a key factor that influences tumor drug uptake/exposure. Tumor vasculature-targeting strategies may thus result in improved tumor drug exposure and therefore drug efficacy.
Collapse
Affiliation(s)
- Azeem Saleem
- Academic Department of Radiation Oncology, The Christie Hospital NHS Foundation Trust, Manchester.
| | | |
Collapse
|
11
|
Boss DS, Olmos RV, Sinaasappel M, Beijnen JH, Schellens JHM. Application of PET/CT in the development of novel anticancer drugs. Oncologist 2008; 13:25-38. [PMID: 18245010 DOI: 10.1634/theoncologist.2007-0097] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Combined positron emission tomography/computed tomography (PET/CT) is a relatively new imaging modality, combining the functional images of PET with the anatomical information of CT. Since its commercial introduction about 5 years ago, PET/CT has become an important tool in oncology. Currently, the technique is used for primary staging and restaging of cancer patients, as well as for surgery and radiation therapy planning. The abilities of PET/CT to measure early treatment response as well as drug distribution within the body make this technique very useful in the development of novel anticancer drugs. In this paper, the recent literature on the current role of PET/CT in drug development is reviewed.
Collapse
Affiliation(s)
- David S Boss
- Division of Clinical Pharmacology, Department of Medical Oncology,The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
12
|
Saleem A, Aboagye EO, Matthews JC, Price PM. Plasma pharmacokinetic evaluation of cytotoxic agents radiolabelled with positron emitting radioisotopes. Cancer Chemother Pharmacol 2008; 61:865-73. [PMID: 17639391 DOI: 10.1007/s00280-007-0552-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Accepted: 06/18/2007] [Indexed: 11/28/2022]
Abstract
PURPOSE This study aimed to evaluate the utility of plasma pharmacokinetic analyses of anti-cancer agents from data obtained during positron emission tomography (PET) oncology studies of radiolabelled anti-cancer agents. PATIENTS AND METHODS Thirteen patients were administered fluorine-18 radiolabelled 5-FU ([(18)F]5-FU) admixed with 5-FU, corresponding to a total 5-FU dose of 380-407 mg/m2 (eight patients) and 1 mg/m2 (five patients). Nine patients received 2.2-19.2 microg/m2 of carbon-11 radiolabelled N-[2-(dimethylamino)ethyl]acridine-4-carboxamide ([11C]DACA) at 1/1,000th of phase I dose, as part of phase 0 microdosing study. Radioactivity of parent drug obtained from arterial blood samples, the injected activity of the radiolabelled drug, and the total dose of injected drug were used to obtain plasma drug concentrations. Plasma pharmacokinetic parameters were estimated using model-dependent and model-independent methods. RESULTS 5-FU plasma concentrations at therapeutic doses were above the Km and a single compartment kinetic model was best used to fit the kinetics, with a mean half-life of 8.6 min. Clearance and volumes of distribution (Vd) obtained using both model-dependent and model-independent methods were similar. Mean (SE) clearance was 1,421(144), ml min(-1) and 1,319 (119) ml min(-1) and the mean (SE) Vd was 17.3 (1.8) l and 16.3 (1.9) l by the model-independent method and model-dependent methods, respectively. In contrast, with 1 mg/m2, plasma concentrations of 5-FU were less than the Km and a two-compartment model was used to best fit the kinetics, with the mean 5-FU half-life of 6.5 min. The mean (SE) clearances obtained by the model-independent method and model-dependent methods were 3,089 (314) ml min(-1) and 2,225 (200) ml min(-1), respectively and the mean (SE) Vd were 27.9 (7.0) l and 2.3 (0.4) l, by the model independent and dependent methods, respectively. Extrapolation of AUC0-Clast to AUC0-infinity was less than 3% in both these cohort of patients. A two-compartment model with a mean half-life of 42.1 min was used to best fit the kinetics of DACA; considerable extrapolation (mean 26%) was required to obtain AUC0-infinity from AUC0-Clast. Mean (SE) clearance of DACA was 1,920 (269) ml min(-1), with the model-independent method and 1,627 (287) ml min(-1) with the model-dependent method. Similarly, Vd [mean (SE)] of DACA with the model-independent and model-dependent methods were 118 (22) l and 50 (15) l, respectively. CONCLUSIONS Pharmacokinetic parameters can be estimated with confidence from PET studies for agents given at therapeutic doses, whose half-lives are significantly less than the total sampling time during the scan. Tracer studies performed alone, wherein plasma levels below the Km are expected, may also provide valuable information on drug clearance for the entire range of linear kinetics. However, drugs with half-lives longer than the sampling duration are inappropriate for PET plasma pharmacokinetic evaluation.
Collapse
Affiliation(s)
- A Saleem
- Academic Department of Radiation Oncology, Christie Hospital NHS Foundation Trust, Wilmslow Road, and The University of Manchester Wolfson Molecular Imaging Centre, Manchester M20 4BX, UK.
| | | | | | | |
Collapse
|
13
|
Abstract
New surrogate end points for monitoring response to cancer treatment are needed for both current and novel therapeutic strategies. Positron emission tomography (PET) as a functional imaging technology provides rapid, reproducible, noninvasive in vivo assessment and quantification of several biological processes targeted by anticancer therapies. PET imaging with F-18 fluorodeoxyglucose (FDG), reflecting tumor glucose metabolism, provides relevant information regarding treatment response. Changes in tumor glucose metabolism precede changes in tumor size and reflect drug effects at a cellular level. FDG-PET enables the prediction of therapy response early in the course as well as determining the viability of residual masses after completion of treatment. The assessment of novel anticancer agents will increasingly depend on functional PET imaging. Assessing responses to new biological drugs using changes in tumor size is likely an inaccurate measure of efficacy. Likewise, monitoring for drug effects using surrogate (nontumor) tissues or serial invasive testing by tumor biopsies does not provide a good correlation with overall antitumor activity. Therefore, the information derived from PET using radiolabeled biological probes provides an alternative approach to conventional structural (anatomical) imaging. PET pharmacokinetic studies will allow for the rapid assessment of novel drug biodistribution, and much smaller patient number studies before decisions on whether or not to proceed with the development of a new drug are made. Summative readouts by PET, such as drug-induced changes in tumor glucose metabolism, tumor cell proliferation and tumor perfusion and, similarly, measures of specific changes will demonstrate whether drugs are having their intended biological effects.
Collapse
Affiliation(s)
- Norbert Avril
- Department of Nuclear Medicine, Queen Mary, University of London, Barts & The London School of Medicine, London, EC1A 7BE, UK.
| | | |
Collapse
|
14
|
Lammertsma AA, Hoekstra CJ, Giaccone G, Hoekstra OS. How should we analyse FDG PET studies for monitoring tumour response? Eur J Nucl Med Mol Imaging 2007; 33 Suppl 1:16-21. [PMID: 16763817 DOI: 10.1007/s00259-006-0131-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
FDG PET is a promising technique for monitoring tumour response early during anticancer therapy. Progress, however, has been limited owing to the multitude of methods currently in use. Here, the most promising techniques for multi-centre trials are discussed briefly, with emphasis on the need for standardisation. In addition, an approach is presented for response monitoring studies using newly developed drugs. This approach makes use of a large database of response monitoring studies, which defines the relationship between simplified clinical methods and full quantitative analysis for classic cytotoxic drugs. For a new drug, first a pilot study is performed to assess whether it affects this relationship. Based on this pilot, it is decided whether or not a simplified clinical method can be used in further studies.
Collapse
Affiliation(s)
- Adriaan A Lammertsma
- Department of Nuclear Medicine and PET Research, VU University Medical Centre, P.O. Box 7057, 1007 MB, Amsterdam, The Netherlands.
| | | | | | | |
Collapse
|
15
|
Saleem A, Charnley N, Price P. Clinical molecular imaging with positron emission tomography. Eur J Cancer 2006; 42:1720-7. [PMID: 16797972 DOI: 10.1016/j.ejca.2006.02.021] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Accepted: 02/10/2006] [Indexed: 11/30/2022]
Abstract
Molecular imaging allows for the in vivo evaluation of targeted molecules and biological processes in man. Positron emission tomography (PET) is a highly sensitive and quantitative molecular imaging modality, whose utility in clinical and experimental medicine is increasing by the day. In this article, the principles of PET and its currently accepted applications in oncology, such as cancer staging, treatment response assessment and as a prognostic marker are reviewed. Further, the evolving role of PET in areas of oncology such as radiotherapy treatment planning, anti-cancer drug development and the evaluation of patho-physiological processes which drive a cell into neoplastic activity is discussed.
Collapse
Affiliation(s)
- Azeem Saleem
- The University of Manchester Wolfson Molecular Imaging Centre, 27 Palatine Road, Withington, Manchester, M20 3JL, UK.
| | | | | |
Collapse
|
16
|
Bergström M, Yates R, Wall A, Kågedal M, Syvänen S, Långström B. Blood-brain barrier penetration of zolmitriptan--modelling of positron emission tomography data. J Pharmacokinet Pharmacodyn 2006; 33:75-91. [PMID: 16402287 DOI: 10.1007/s10928-005-9001-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Positron emission tomography (PET) with the drug radiolabelled allows a direct measurement of brain or other organ kinetics, information which can be essential in drug development. Usually, however, a PET-tracer is administered intravenously (i.v.), whereas the therapeutic drug is mostly given orally or by a different route to the PET-tracer. In such cases, a recalculation is needed to make the PET data representative for the alternative administration route. To investigate the blood-brain barrier penetration of a drug (zolmitriptan) using dynamic PET and by PK modelling quantify the brain concentration of the drug after the nasal administration of a therapeutic dose. [11C]Zolmitriptan at tracer dose was administered as a short i.v. infusion and the brain tissue and venous blood kinetics of [11C]zolmitriptan was measured by PET in 7 healthy volunteers. One PET study was performed before and one 30 min after the administration of 5 mg zolmitriptan as nasal spray. At each of the instances, the brain radioactivity concentration after subtraction of the vascular component was determined up to 90 min after administration and compared to venous plasma radioactivity concentration after correction for radiolabelled metabolites. Convolution methods were used to describe the relationship between arterial and venous tracer concentrations, respectively between brain and arterial tracer concentration. Finally, the impulse response functions derived from the PET studies were applied on plasma PK data to estimate the brain zolmitriptan concentration after a nasal administration of a therapeutic dose. The studies shows that the PET data on brain kinetics could well be described as the convolution of venous tracer kinetics with an impulse response including terms for arterial-to-venous plasma and arterial-to-brain impulse responses. Application of the PET derived impulse responses on the plasma PK from nasal administration demonstrated that brain PK of zolmitriptan increased with time, achieving about 0.5 mg/ml at 30 min and close to a maximum of 1.5 mg/ml after 2 hr. A significant brain concentration was observed already after 5 min. The data support the notation of a rapid brain availability of zolmitriptan after nasal administration.
Collapse
Affiliation(s)
- Mats Bergström
- Uppsala Imanet AB, University Hospital, SE-751 85, Uppsala, Sweden.
| | | | | | | | | | | |
Collapse
|
17
|
Abstract
Medical imaging technologies have undergone explosive growth over the past few decades and now play a central role in clinical oncology. But the truly transformative power of imaging in the clinical management of cancer patients lies ahead. Today, imaging is at a crossroads, with molecularly targeted imaging agents expected to broadly expand the capabilities of conventional anatomical imaging methods. Molecular imaging will allow clinicians to not only see where a tumor is located in the body, but also to visualize the expression and activity of specific molecules (e.g., proteases and protein kinases) and biological processes (e.g., apoptosis, angiogenesis, and metastasis) that influence tumor behavior and/or response to therapy. This information is expected to have a major impact on cancer detection, individualized treatment, and drug development, as well as our understanding of how cancer arises.
Collapse
Affiliation(s)
- Ralph Weissleder
- Center for Molecular Imaging Research, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
| |
Collapse
|
18
|
Bergström M, Långström B. Pharmacokinetic studies with PET. PROGRESS IN DRUG RESEARCH. FORTSCHRITTE DER ARZNEIMITTELFORSCHUNG. PROGRES DES RECHERCHES PHARMACEUTIQUES 2006; 62:279-317. [PMID: 16329260 DOI: 10.1007/3-7643-7426-8_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Affiliation(s)
- Mats Bergström
- Uppsala Imanet, GE Health Care, Box 967, SE-751 09 Uppsala, Sweden.
| | | |
Collapse
|
19
|
|
20
|
|
21
|
West CML, Jones T, Price P. The potential of positron-emission tomography to study anticancer-drug resistance. Nat Rev Cancer 2004; 4:457-69. [PMID: 15170448 DOI: 10.1038/nrc1368] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Catharine M L West
- Academic Department of Radiation Oncology and Manchester Molecular Imaging Centre, University of Manchester, Christie NHS Trust Hospital, Wilmslow Road, Manchester, M20 4BX, United Kingdom.
| | | | | |
Collapse
|
22
|
Han C, Braybrooke JP, Deplanque G, Taylor M, Mackintosh D, Kaur K, Samouri K, Ganesan TS, Harris AL, Talbot DC. Comparison of prognostic factors in patients in phase I trials of cytotoxic drugs vs new noncytotoxic agents. Br J Cancer 2003; 89:1166-71. [PMID: 14520440 PMCID: PMC2394292 DOI: 10.1038/sj.bjc.6601218] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The aims of this study were to identify prognostic variables for toxicity and survival in patients with cancer participating in phase I clinical trials and compare characteristics of those treated with cytotoxic chemotherapy (CT) and non-cytotoxic drugs (non-CT). Data were collected from 420 (114 CT, 306 non-CT) patients enrolled in 16 phase I trials (five CT and 11 non-CT trials) in one cancer centre. Analyses of all patients were used to compare treatment groups, identify predictive variables for toxicity and to estimate prognostic factors in overall survival (OS). These were used to develop a prognostic index (PI). Multivariate analysis found those patients with better performance status, fewer sites of metastases, baseline Hb>12 g dl−1 and WBC or LDH in the normal range had significantly better OS. Male gender, platelet count <450 × 109 l−1, high WBC or treatment with a non-CT phase I agent significantly reduced the chance of grade 3/4 toxicity. Overall survival was not significantly different between the CT and non-CT groups (260 vs 192 days, P=0.47) except for those with liver metastases (228 vs 137 days, P=0.02). Overall tumour response was 4.9% (95% CI: 2.7–7.0%). The PI identified three distinct patient groups with median survival of 321, 257 and 117 days. In conclusion, entry into a phase I trial of a non-CT drug is a safe option for heavily pretreated patients with cancer. The PI generated from these data can estimate the survival probability for patients entering phase I studies.
Collapse
Affiliation(s)
- C Han
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - J P Braybrooke
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - G Deplanque
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - M Taylor
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - D Mackintosh
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - K Kaur
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - K Samouri
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - T S Ganesan
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - A L Harris
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
| | - D C Talbot
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK
- Cancer Research UK Medical Oncology Unit, Churchill Hospital, Headington, Oxford OX3 7LJ, UK. E-mail:
| |
Collapse
|
23
|
Bergström M, Grahnén A, Långström B. Positron emission tomography microdosing: a new concept with application in tracer and early clinical drug development. Eur J Clin Pharmacol 2003; 59:357-66. [PMID: 12937873 DOI: 10.1007/s00228-003-0643-x] [Citation(s) in RCA: 206] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2002] [Accepted: 06/25/2003] [Indexed: 10/26/2022]
Abstract
The realisation that new chemical entities under development as drug candidates fail in three of four cases in clinical trials, together with increased costs and increased demands of reducing preclinical animal experiments, have promoted concepts for improvement of early screening procedures in humans. Positron emission tomography (PET) is a non-invasive imaging technology, which makes it possible to determine drug distribution and concentration in vivo in man with the drug labelled with a positron-emitting radionuclide that does not change the biochemical properties. Recently, developments in the field of rapid synthesis of organic compounds labelled with positron-emitting radionuclides have allowed a substantial number of new drug candidates to be labelled and potentially used as probes in PET studies. Together, these factors led to the logical conclusion that early PET studies, performed with very low drug doses-PET-microdosing-could be included in the drug development process as one means for selection or rejection of compounds based on performance in vivo in man. Another important option of PET, to evaluate drug interaction with a target, utilising a PET tracer specific for this target, necessitates a more rapid development of such PET methodology and validations in humans. Since only very low amounts of drugs are used in PET-microdosing studies, the safety requirements should be reduced relative to the safety requirements needed for therapeutic doses. In the following, a methodological scrutinising of the concept is presented. A complete pre-clinical package including limited toxicity assessment is proposed as a base for the regulatory framework of the PET-microdosing concept.
Collapse
Affiliation(s)
- Mats Bergström
- Department of Pharmaceutical Biosciences, Faculty of Pharmacy, Uppsala University, Sweden,
| | | | | |
Collapse
|
24
|
|