1
|
Lentzas A, de Gooijer MC, Zuidema S, Meurs A, Çitirikkaya CH, Venekamp N, Beijnen JH, van Tellingen O. ATP-binding cassette transporter inhibitor potency and substrate drug affinity are critical determinants of successful drug delivery enhancement to the brain. Fluids Barriers CNS 2024; 21:62. [PMID: 39103921 DOI: 10.1186/s12987-024-00562-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/28/2024] [Indexed: 08/07/2024] Open
Abstract
BACKGROUND Pharmacotherapy for brain diseases is severely compromised by the blood-brain barrier (BBB). ABCB1 and ABCG2 are drug transporters that restrict drug entry into the brain and their inhibition can be used as a strategy to boost drug delivery and pharmacotherapy for brain diseases. METHODS We employed elacridar and tariquidar in mice to explore the conditions for effective inhibition at the BBB. Abcg2;Abcb1a/b knockout (KO), Abcb1a/b KO, Abcg2 KO and wild-type (WT) mice received a 3 h i.p. infusion of a cocktail of 8 typical substrate drugs in combination with elacridar or tariquidar at a range of doses. Abcg2;Abcb1a/b KO mice were used as the reference for complete inhibition, while single KO mice were used to assess the potency to inhibit the remaining transporter. Brain and plasma drug levels were measured by LC-MS/MS. RESULTS Complete inhibition of ABCB1 at the BBB is achieved when the elacridar plasma level reaches 1200 nM, whereas tariquidar requires at least 4000 nM. Inhibition of ABCG2 is more difficult. Elacridar inhibits ABCG2-mediated efflux of weak but not strong ABCG2 substrates. Strikingly, tariquidar does not enhance the brain uptake of any ABCG2-subtrate drug. Similarly, elacridar, but not tariquidar, was able to inhibit its own brain efflux in ABCG2-proficient mice. The plasma protein binding of elacridar and tariquidar was very high but similar in mouse and human plasma, facilitating the translation of mouse data to humans. CONCLUSIONS This work shows that elacridar is an effective pharmacokinetic-enhancer for the brain delivery of ABCB1 and weaker ABCG2 substrate drugs when a plasma concentration of 1200 nM is exceeded.
Collapse
Affiliation(s)
- Aristeidis Lentzas
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Mark C de Gooijer
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M1 3WE, UK
- The Christie NHS Foundation Trust, Manchester, M20 4BX, UK
| | - Stefanie Zuidema
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Amber Meurs
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Ceren H Çitirikkaya
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
| | - Nikkie Venekamp
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands
- Department of Pharmacy, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- Division of Pharmacoepidemiology and Clinical Pharmacology, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, The Netherlands
| | - Olaf van Tellingen
- Division of Pharmacology, The Netherlands Cancer Institute, Room H3.010, Plesmanlaan 121, Amsterdam, 1066 CX, The Netherlands.
- Mouse Cancer Clinic, The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Wu CP, Hsiao SH, Wu YS. Perspectives on drug repurposing to overcome cancer multidrug resistance mediated by ABCB1 and ABCG2. Drug Resist Updat 2023; 71:101011. [PMID: 37865067 DOI: 10.1016/j.drup.2023.101011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 10/07/2023] [Accepted: 10/08/2023] [Indexed: 10/23/2023]
Abstract
The overexpression of the human ATP-binding cassette (ABC) transporters in cancer cells is a common mechanism involved in developing multidrug resistance (MDR). Unfortunately, there are currently no approved drugs specifically designed to treat multidrug-resistant cancers, making MDR a significant obstacle to successful chemotherapy. Despite over two decades of research, developing transporter-specific inhibitors for clinical use has proven to be a challenging endeavor. As an alternative approach, drug repurposing has gained traction as a more practical method to discover clinically effective modulators of drug transporters. This involves exploring new indications for already-approved drugs, bypassing the lengthy process of developing novel synthetic inhibitors. In this context, we will discuss the mechanisms of ABC drug transporters ABCB1 and ABCG2, their roles in cancer MDR, and the inhibitors that have been evaluated for their potential to reverse MDR mediated by these drug transporters. Our focus will be on providing an up-to-date report on approved drugs tested for their inhibitory activities against these drug efflux pumps. Lastly, we will explore the challenges and prospects of repurposing already approved medications for clinical use to overcome chemoresistance in patients with high tumor expression of ABCB1 and/or ABCG2.
Collapse
Affiliation(s)
- Chung-Pu Wu
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Physiology and Pharmacology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Molecular Medicine Research Center, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan; Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei 10507, Taiwan.
| | - Sung-Han Hsiao
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yu-Shan Wu
- Department of Chemistry, Tunghai University, Taichung 40704, Taiwan.
| |
Collapse
|
3
|
Schulz JA, Hartz AMS, Bauer B. ABCB1 and ABCG2 Regulation at the Blood-Brain Barrier: Potential New Targets to Improve Brain Drug Delivery. Pharmacol Rev 2023; 75:815-853. [PMID: 36973040 PMCID: PMC10441638 DOI: 10.1124/pharmrev.120.000025] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 03/29/2023] Open
Abstract
The drug efflux transporters ABCB1 and ABCG2 at the blood-brain barrier limit the delivery of drugs into the brain. Strategies to overcome ABCB1/ABCG2 have been largely unsuccessful, which poses a tremendous clinical problem to successfully treat central nervous system (CNS) diseases. Understanding basic transporter biology, including intracellular regulation mechanisms that control these transporters, is critical to solving this clinical problem.In this comprehensive review, we summarize current knowledge on signaling pathways that regulate ABCB1/ABCG2 at the blood-brain barrier. In Section I, we give a historical overview on blood-brain barrier research and introduce the role that ABCB1 and ABCG2 play in this context. In Section II, we summarize the most important strategies that have been tested to overcome the ABCB1/ABCG2 efflux system at the blood-brain barrier. In Section III, the main component of this review, we provide detailed information on the signaling pathways that have been identified to control ABCB1/ABCG2 at the blood-brain barrier and their potential clinical relevance. This is followed by Section IV, where we explain the clinical implications of ABCB1/ABCG2 regulation in the context of CNS disease. Lastly, in Section V, we conclude by highlighting examples of how transporter regulation could be targeted for therapeutic purposes in the clinic. SIGNIFICANCE STATEMENT: The ABCB1/ABCG2 drug efflux system at the blood-brain barrier poses a significant problem to successful drug delivery to the brain. The article reviews signaling pathways that regulate blood-brain barrier ABCB1/ABCG2 and could potentially be targeted for therapeutic purposes.
Collapse
Affiliation(s)
- Julia A Schulz
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| | - Anika M S Hartz
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| | - Björn Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy (J.A.S., B.B.), Sanders-Brown Center on Aging and Department of Pharmacology and Nutritional Sciences, College of Medicine (A.M.S.H.), University of Kentucky, Lexington, Kentucky
| |
Collapse
|
4
|
Lefebvre C, Pellizzari S, Bhat V, Jurcic K, Litchfield DW, Allan AL. Involvement of the AKT Pathway in Resistance to Erlotinib and Cabozantinib in Triple-Negative Breast Cancer Cell Lines. Biomedicines 2023; 11:2406. [PMID: 37760847 PMCID: PMC10525382 DOI: 10.3390/biomedicines11092406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/14/2023] [Accepted: 08/26/2023] [Indexed: 09/29/2023] Open
Abstract
Resistance to protein tyrosine kinase inhibitors (TKIs) presents a significant challenge in therapeutic target development for cancers such as triple-negative breast cancer (TNBC), where conventional therapies are ineffective at combatting systemic disease. Due to increased expression, the receptor tyrosine kinases EGFR (epidermal growth factor receptor) and c-Met are potential targets for treatment. However, targeted anti-EGFR and anti-c-Met therapies have faced mixed results in clinical trials due to acquired resistance. We hypothesize that adaptive responses in regulatory kinase networks within the EGFR and c-Met signaling axes contribute to the development of acquired erlotinib and cabozantinib resistance. To test this, we developed two separate models for cabozantinib and erlotinib resistance using the MDA-MB-231 and MDA-MB-468 cell lines, respectively. We observed that erlotinib- or cabozantinib-resistant cell lines demonstrate enhanced cell proliferation, migration, invasion, and activation of EGFR or c-Met downstream signaling (respectively). Using a SILAC (Stable Isotope Labeling of Amino acids in Cell Culture)-labeled quantitative mass spectrometry proteomics approach, we assessed the effects of erlotinib or cabozantinib resistance on the phosphoproteome, proteome, and kinome. Using this integrated proteomics approach, we identified several potential kinase mediators of cabozantinib resistance and confirmed the contribution of AKT1 to erlotinib resistance in TNBC-resistant cell lines.
Collapse
Affiliation(s)
- Cory Lefebvre
- London Regional Cancer Program, London Health Sciences Centre, London, ON N6A 5W9, Canada; (C.L.); (S.P.); (V.B.)
- Department of Anatomy & Cell Biology, Western University, London, ON N6A 3K7, Canada
| | - Sierra Pellizzari
- London Regional Cancer Program, London Health Sciences Centre, London, ON N6A 5W9, Canada; (C.L.); (S.P.); (V.B.)
- Department of Anatomy & Cell Biology, Western University, London, ON N6A 3K7, Canada
| | - Vasudeva Bhat
- London Regional Cancer Program, London Health Sciences Centre, London, ON N6A 5W9, Canada; (C.L.); (S.P.); (V.B.)
- Department of Anatomy & Cell Biology, Western University, London, ON N6A 3K7, Canada
| | - Kristina Jurcic
- Department of Biochemistry, Western University, London, ON N6A 3K7, Canada; (K.J.); (D.W.L.)
| | - David W. Litchfield
- Department of Biochemistry, Western University, London, ON N6A 3K7, Canada; (K.J.); (D.W.L.)
- Department of Oncology, Western University, London, ON N6A 3K7, Canada
| | - Alison L. Allan
- London Regional Cancer Program, London Health Sciences Centre, London, ON N6A 5W9, Canada; (C.L.); (S.P.); (V.B.)
- Department of Anatomy & Cell Biology, Western University, London, ON N6A 3K7, Canada
- Department of Oncology, Western University, London, ON N6A 3K7, Canada
- Lawson Health Research Institute, London, ON N6A 5W9, Canada
| |
Collapse
|
5
|
Shi D, Dong Y, Zhou W, Bai L, Huang J, Han Y, Sun P, Huang Y, Huang Y, Chen L, Cao M, Wu H, Huang S. Pharmacokinetic analysis of 6-O-[ 18F]FEE for PET imaging of EGFR mutation. Bioorg Med Chem Lett 2023; 85:129217. [PMID: 36889652 DOI: 10.1016/j.bmcl.2023.129217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/25/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023]
Abstract
6-O-[18F]Fluoroethylerlotinib (6-O-[18F]FEE), with a suitable half-life for commercial distribution, may be a good replacement for [11C]erlotinib to identify epidermal growth factor receptor (EGFR) positive tumors with activating mutations to tyrosine kinase inhibitors therapy. In this study, we explored the fully automated synthesis of 6-O-[18F]FEE and investigated its pharmacokinetics in tumor-bearing mice. 6-O-[18F]FEE with high specific activity (28-100 GBq/μmol) and radiochemistry purity (over 99 %) was obtained by two-step reaction and Radio-HPLC separation in PET-MF-2 V-IT-1 automated synthesizer. PET imaging of 6-O-[18F]FEE in HCC827, A431, and U87 tumor-bearing mice with different EGFR expression and mutation was performed. Uptake and blocking of PET imaging indicated that the probe specifically targeted exon 19 deleted EGFR (the quantitative analysis of tumor-to-mouse ratio for HCC827, HCC827 blocking, U87, A431 was 2.58 ± 0.24, 1.20 ± 0.15, 1.18 ± 0.19, and 1.05 ± 0.13 respectively). Dynamic imaging was used to study the pharmacokinetics of the probe in tumor-bearing mice. Logan plot graphical analysis demonstrated late linearity and a high fitting correlation coefficient (0.998), supporting reversible kinetics. According to the Akaike Information Criterion (AIC) rule, the 2-compartment reversible model was more consistent with the metabolic properties of 6-O-[18F]FEE. The automated radiosynthesis and pharmacokinetic analysis will promote clinically transformation of 6-O-[18F]FEE.
Collapse
Affiliation(s)
- Dazhi Shi
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Ye Dong
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Wenlan Zhou
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Lu Bai
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Jiawen Huang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and, Peking Union Medical College, Shenzhen 518116, China
| | - Yanjiang Han
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Penghui Sun
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Yanchao Huang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Yong Huang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and, Peking Union Medical College, Shenzhen 518116, China
| | - Li Chen
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Min Cao
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China
| | - Hubing Wu
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China.
| | - Shun Huang
- GDMPA Key Laboratory for Quality Control and Evaluation of Radiopharmaceuticals, Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, 1838 Guangzhou North Road, Guangzhou, Guangdong Province 510515, China.
| |
Collapse
|
6
|
Tran VL, Bouleau A, Nozach H, Richard M, Chevaleyre C, Dubois S, Kereselidze D, Kuhnast B, Evans MJ, Specklin S, Truillet C. Impact of Radiolabeling Strategies on the Pharmacokinetics and Distribution of an Anti-PD-L1 PET Ligand. Mol Pharm 2022; 19:3673-3680. [PMID: 35998011 DOI: 10.1021/acs.molpharmaceut.2c00497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Molecular imaging with PET offers an alternative method to quantify programmed-death-ligand 1 (PD-L1) to accurately select patients for immunotherapies. More and more clinical and preclinical trials involve radiolabeling of antibody fragments for their desirably fast clearance and high tumor penetration. As the radiolabeling strategy can significantly impact pharmacokinetics and biodistribution, we explored in this work a site-specific radiofluorination strategy on an anti-PD-L1 fragment antigen-binding (Fab) and compared the pharmacokinetic and biodistribution properties with the same Fab labeled using stochastic radiolabeling chemistry. We applied an enzymatic bioconjugation mediated by a variant of the lipoic acid ligase (LplA) that promotes the formation of an amide bond between a short peptide cloned onto the C terminus of the Fab. A synthetic analogue of the enzyme natural substrate, lipoic acid, was radiolabeled with fluorine-18 for site-specific conjugation by LplA. We compared the biodistribution of the site-specifically labeled Fab with a stochastically labeled Fab on lysine side chains in tumor-bearing mice. The two methods of fluorination demonstrate a comparable whole-body biodistribution. The 89Zr-labeled Fab had different biodistribution compared to either 18F-labeled Fab. We attribute the difference to [89Zr] metabolism. Fab-LAP-[18F]FPyOctA therefore reflects better the true pharmacokinetic profile of the Fab.
Collapse
Affiliation(s)
- Vu Long Tran
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Alizée Bouleau
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Hervé Nozach
- Université Paris-Saclay, CEA, DMTS, SIMoS, CEA-Saclay, Gif-sur-Yvette CEDEX 91191, France
| | - Mylène Richard
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Céline Chevaleyre
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Steven Dubois
- Université Paris-Saclay, CEA, DMTS, SIMoS, CEA-Saclay, Gif-sur-Yvette CEDEX 91191, France
| | - Dimitri Kereselidze
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Bertrand Kuhnast
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging, UCSF, San Francisco, California 94107, United States
| | - Simon Specklin
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| | - Charles Truillet
- Université Paris-Saclay, CEA, CNRS, Inserm, BioMaps, SHFJ, Orsay 91400, France
| |
Collapse
|
7
|
Mairinger S, Hernández-Lozano I, Filip T, Sauberer M, Löbsch M, Stanek J, Wanek T, Sake JA, Pekar T, Ehrhardt C, Langer O. Impact of P-gp and BCRP on pulmonary drug disposition assessed by PET imaging in rats. J Control Release 2022; 349:109-117. [PMID: 35798092 DOI: 10.1016/j.jconrel.2022.06.065] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/21/2022] [Accepted: 06/29/2022] [Indexed: 10/17/2022]
Abstract
P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) are two efflux transporters which are expressed in the apical (i.e. airway lumen-facing) membranes of lung epithelial cells. To assess the influence of P-gp and BCRP on the pulmonary disposition of inhaled drugs, we performed positron emission tomography (PET) imaging in rats after intratracheal aerosolization of two model P-gp/BCRP substrate radiotracers (i.e. [11C]erlotinib and [11C]tariquidar). We studied rat groups in which both transporters were active (i.e. wild-type rats), either of the two transporters was inactive (Abcb1a/b(-/-) and Abcg2(-/-) rats) or both transporters were inactive (Abcg2(-/-) rats in which pulmonary P-gp activity was inhibited by treatment with unlabeled tariquidar). PET-measured lung distribution data were compared with brain-to-plasma radioactivity concentration ratios measured in a gamma counter at the end of the PET scan. For [11C]erlotinib, lung exposure (AUClungs) was moderately but not significantly increased in Abcb1a/b(-/-) rats (1.6-fold) and Abcg2(-/-) rats (1.5-fold), and markedly (3.6-fold, p < 0.0001) increased in tariquidar-treated Abcg2(-/-) rats, compared to wild-type rats. Similarly, the brain uptake of [11C]erlotinib was substantially (4.5-fold, p < 0.0001) increased when both P-gp and BCRP activities were impaired. For [11C]tariquidar, differences in AUClungs between groups pointed into a similar direction as for [11C]erlotinib, but were less pronounced and lacked statistical significance. Our study demonstrates functional P-gp and BCRP activity in vivo in the lungs and further suggests functional redundancy between P-gp and BCRP in limiting the pulmonary uptake of a model P-gp/BCRP substrate, analogous to the blood-brain barrier. Our results suggest that pulmonary efflux transporters are important for the efficacy and safety of inhaled drugs and that their modulation may be exploited in order to improve the pharmacokinetic and pharmacodynamic performance of pulmonary delivered drugs.
Collapse
Affiliation(s)
- Severin Mairinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Thomas Filip
- Core Facility Laboratory Animal Breeding and Husbandry, Medical University of Vienna, Vienna, Austria
| | - Michael Sauberer
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Mathilde Löbsch
- Core Facility Laboratory Animal Breeding and Husbandry, Medical University of Vienna, Vienna, Austria
| | - Johann Stanek
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Thomas Wanek
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Johannes A Sake
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Thomas Pekar
- Biomedical Sciences, University of Applied Sciences Wiener Neustadt, Wiener Neustadt, Austria
| | - Carsten Ehrhardt
- School of Pharmacy and Pharmaceutical Sciences and Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
8
|
Li C, Yang Q, Chen Z, Qiu Y, Du Y, Wang R, He Q, Yang J, Zhen H, Kang L. Noninvasive Evaluation of Multidrug Resistance via Imaging of ABCG2/BCRP Multidrug Transporter in Lung Cancer Xenograft Models. Mol Pharm 2022; 19:3521-3529. [PMID: 35427142 DOI: 10.1021/acs.molpharmaceut.1c00939] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Cuicui Li
- Department of Nuclear Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing 100050, China
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Qi Yang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Zhao Chen
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Yongkang Qiu
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Yujing Du
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Rongfu Wang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Qihua He
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Jigang Yang
- Department of Nuclear Medicine, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing 100050, China
| | - Hongying Zhen
- Department of Cell Biology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
9
|
Högnäsbacka A, Poot AJ, Vugts DJ, van Dongen GAMS, Windhorst AD. The Development of Positron Emission Tomography Tracers for In Vivo Targeting the Kinase Domain of the Epidermal Growth Factor Receptor. Pharmaceuticals (Basel) 2022; 15:ph15040450. [PMID: 35455447 PMCID: PMC9033078 DOI: 10.3390/ph15040450] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/24/2022] [Accepted: 03/25/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple small molecule PET tracers have been developed for the imaging of the epidermal growth factor receptor (EGFR). These tracers target the tyrosine kinase (TK) domain of the receptor and have been used for both quantifying EGFR expression and to differentiate between EGFR mutational statuses. However, the approaches for in vivo evaluation of these tracers are diverse and have resulted in data that are hard to compare. In this review, we analyze the historical development of the in vivo evaluation approaches, starting from the first EGFR TK PET tracer [11C]PD153035 to tracers developed based on TK inhibitors used for the clinical treatment of mutated EGFR expressing non-small cell lung cancer like [11C]erlotinib and [18F]afatinib. The evaluation of each tracer has been compiled to allow for a comparison between studies and ultimately between tracers. The main challenges for each group of tracers are thereafter discussed. Finally, this review addresses the challenges that need to be overcome to be able to efficiently drive EGFR PET imaging forward.
Collapse
Affiliation(s)
- Antonia Högnäsbacka
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.J.P.); (D.J.V.); (G.A.M.S.v.D.)
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
- Correspondence: (A.H.); (A.D.W.)
| | - Alex J. Poot
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.J.P.); (D.J.V.); (G.A.M.S.v.D.)
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
| | - Danielle J. Vugts
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.J.P.); (D.J.V.); (G.A.M.S.v.D.)
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
| | - Guus A. M. S. van Dongen
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.J.P.); (D.J.V.); (G.A.M.S.v.D.)
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
| | - Albert D. Windhorst
- Department of Radiology & Nuclear Medicine, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands; (A.J.P.); (D.J.V.); (G.A.M.S.v.D.)
- Cancer Center Amsterdam, Imaging and Biomarkers, 1081 HV Amsterdam, The Netherlands
- Correspondence: (A.H.); (A.D.W.)
| |
Collapse
|
10
|
Goebel J, Chmielewski J, Hrycyna CA. The roles of the human ATP-binding cassette transporters P-glycoprotein and ABCG2 in multidrug resistance in cancer and at endogenous sites: future opportunities for structure-based drug design of inhibitors. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 4:784-804. [PMID: 34993424 PMCID: PMC8730335 DOI: 10.20517/cdr.2021.19] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ATP-binding cassette (ABC) transporters P-glycoprotein (P-gp) and ABCG2 are multidrug transporters that confer drug resistance to numerous anti-cancer therapeutics in cell culture. These findings initially created great excitement in the medical oncology community, as inhibitors of these transporters held the promise of overcoming clinical multidrug resistance in cancer patients. However, clinical trials of P-gp and ABCG2 inhibitors in combination with cancer chemotherapeutics have not been successful due, in part, to flawed clinical trial designs resulting from an incomplete molecular understanding of the multifactorial basis of multidrug resistance (MDR) in the cancers examined. The field was also stymied by the lack of high-resolution structural information for P-gp and ABCG2 for use in the rational structure-based drug design of inhibitors. Recent advances in structural biology have led to numerous structures of both ABCG2 and P-gp that elucidated more clearly the mechanism of transport and the polyspecific nature of their substrate and inhibitor binding sites. These data should prove useful helpful for developing even more potent and specific inhibitors of both transporters. As such, although possible pharmacokinetic interactions would need to be evaluated, these inhibitors may show greater effectiveness in overcoming ABC-dependent multidrug resistance in combination with chemotherapeutics in carefully selected subsets of cancers. Another perhaps even more compelling use of these inhibitors may be in reversibly inhibiting endogenously expressed P-gp and ABCG2, which serve a protective role at various blood-tissue barriers. Inhibition of these transporters at sanctuary sites such as the brain and gut could lead to increased penetration by chemotherapeutics used to treat brain cancers or other brain disorders and increased oral bioavailability of these agents, respectively.
Collapse
Affiliation(s)
- Jason Goebel
- Department of Chemistry, Purdue University West Lafayette, IN 47907, USA
| | - Jean Chmielewski
- Department of Chemistry, Purdue University West Lafayette, IN 47907, USA
| | | |
Collapse
|
11
|
Assessing the Functional Redundancy between P-gp and BCRP in Controlling the Brain Distribution and Biliary Excretion of Dual Substrates with PET Imaging in Mice. Pharmaceutics 2021; 13:pharmaceutics13081286. [PMID: 34452247 PMCID: PMC8399697 DOI: 10.3390/pharmaceutics13081286] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 12/20/2022] Open
Abstract
P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP) are co-localized at the blood–brain barrier, where they display functional redundancy to restrict the brain distribution of dual P-gp/BCRP substrate drugs. We used positron emission tomography (PET) with the metabolically stable P-gp/BCRP substrates [11C]tariquidar, [11C]erlotinib, and [11C]elacridar to assess whether a similar functional redundancy as at the BBB exists in the liver, where both transporters mediate the biliary excretion of drugs. Wild-type, Abcb1a/b(−/−), Abcg2(−/−), and Abcb1a/b(−/−)Abcg2(−/−) mice underwent dynamic whole-body PET scans after i.v. injection of either [11C]tariquidar, [11C]erlotinib, or [11C]elacridar. Brain uptake of all three radiotracers was markedly higher in Abcb1a/b(−/−)Abcg2(−/−) mice than in wild-type mice, while only moderately changed in Abcb1a/b(−/−) and Abcg2(−/−) mice. The transfer of radioactivity from liver to excreted bile was significantly lower in Abcb1a/b(−/−)Abcg2(−/−) mice and almost unchanged in Abcb1a/b(−/−) and Abcg2(−/−) mice (with the exception of [11C]erlotinib, for which biliary excretion was also significantly reduced in Abcg2(−/−) mice). Our data provide evidence for redundancy between P-gp and BCRP in controlling both the brain distribution and biliary excretion of dual P-gp/BCRP substrates and highlight the utility of PET as an upcoming tool to assess the effect of transporters on drug disposition at a whole-body level.
Collapse
|
12
|
Tran V, Lux F, Tournier N, Jego B, Maître X, Anisorac M, Comtat C, Jan S, Selmeczi K, Evans MJ, Tillement O, Kuhnast B, Truillet C. Quantitative Tissue Pharmacokinetics and EPR Effect of AGuIX Nanoparticles: A Multimodal Imaging Study in an Orthotopic Glioblastoma Rat Model and Healthy Macaque. Adv Healthc Mater 2021; 10:e2100656. [PMID: 34212539 DOI: 10.1002/adhm.202100656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/29/2021] [Indexed: 01/10/2023]
Abstract
AGuIX are emerging radiosensitizing nanoparticles (NPs) for precision radiotherapy (RT) under clinical evaluation (Phase 2). Despite being accompanied by MRI thanks to the presence of gadolinium (Gd) at its surface, more sensitive and quantifiable imaging technique should further leverage the full potential of this technology. In this study, it is shown that 89 Zr can be labeled on such NPs directly for positron emission tomography (PET) imaging with a simple and scalable method. The stability of such complexes is remarkable in vitro and in vivo. Using a glioblastoma orthotopic rat model, it is shown that injected 89 Zr-AGuIX is detectable inside the tumor for at least 1 week. Interestingly, the particles seem to efficiently infiltrate the tumor even in necrotic areas, which places great hope for the treatment of radioresistant tumor. Lastly, the first PET/MR whole-body imaging is performed in non-human primate (NHP), which further demonstrates the translational potential of these bimodal NP.
Collapse
Affiliation(s)
- Vu‐Long Tran
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - François Lux
- Institut Lumière Matière Université Claude Bernard Lyon I CNRS UMR 5306 Villeurbanne 69622 France
- Institut Universitaire de France (IUF) Paris France
| | - Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Benoit Jego
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Xavier Maître
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | | | - Claude Comtat
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Sébastien Jan
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | | | - Michael J. Evans
- Department of Radiology and Biomedical Imaging University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
- Department of Pharmaceutical Chemistry University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
- Helen Diller Family Comprehensive Cancer Center University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
| | - Olivier Tillement
- Institut Lumière Matière Université Claude Bernard Lyon I CNRS UMR 5306 Villeurbanne 69622 France
| | - Bertrand Kuhnast
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Charles Truillet
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| |
Collapse
|
13
|
Tournier N, Goutal S, Mairinger S, Hernández-Lozano I, Filip T, Sauberer M, Caillé F, Breuil L, Stanek J, Freeman AF, Novarino G, Truillet C, Wanek T, Langer O. Complete inhibition of ABCB1 and ABCG2 at the blood-brain barrier by co-infusion of erlotinib and tariquidar to improve brain delivery of the model ABCB1/ABCG2 substrate [ 11C]erlotinib. J Cereb Blood Flow Metab 2021; 41:1634-1646. [PMID: 33081568 PMCID: PMC8221757 DOI: 10.1177/0271678x20965500] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) restrict at the blood-brain barrier (BBB) the brain distribution of the majority of currently known molecularly targeted anticancer drugs. To improve brain delivery of dual ABCB1/ABCG2 substrates, both ABCB1 and ABCG2 need to be inhibited simultaneously at the BBB. We examined the feasibility of simultaneous ABCB1/ABCG2 inhibition with i.v. co-infusion of erlotinib and tariquidar by studying brain distribution of the model ABCB1/ABCG2 substrate [11C]erlotinib in mice and rhesus macaques with PET. Tolerability of the erlotinib/tariquidar combination was assessed in human embryonic stem cell-derived cerebral organoids. In mice and macaques, baseline brain distribution of [11C]erlotinib was low (brain distribution volume, VT,brain < 0.3 mL/cm3). Co-infusion of erlotinib and tariquidar increased VT,brain in mice by 3.0-fold and in macaques by 3.4- to 5.0-fold, while infusion of erlotinib alone or tariquidar alone led to less pronounced VT,brain increases in both species. Treatment of cerebral organoids with erlotinib/tariquidar led to an induction of Caspase-3-dependent apoptosis. Co-infusion of erlotinib/tariquidar may potentially allow for complete ABCB1/ABCG2 inhibition at the BBB, while simultaneously achieving brain-targeted EGFR inhibition. Our protocol may be applicable to enhance brain delivery of molecularly targeted anticancer drugs for a more effective treatment of brain tumors.
Collapse
Affiliation(s)
- Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Sebastien Goutal
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay, France.,MIRCen, CEA/IBFJ/DRF-JACOB/LMN, UMR CEA CNRS 9199-Université Paris Saclay, Fontenay-aux-Roses, France
| | - Severin Mairinger
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | | | - Thomas Filip
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Fabien Caillé
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Louise Breuil
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Johann Stanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Anna F Freeman
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Gaia Novarino
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria
| | - Charles Truillet
- Laboratoire d'Imagerie Biomédicale Multimodale (BioMaps), Université Paris-Saclay, CEA, CNRS, Inserm, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Oliver Langer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria.,Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
14
|
Imaging-Based Characterization of a Slco2b1(-/-) Mouse Model Using [ 11C]Erlotinib and [ 99mTc]Mebrofenin as Probe Substrates. Pharmaceutics 2021; 13:pharmaceutics13060918. [PMID: 34205780 PMCID: PMC8233734 DOI: 10.3390/pharmaceutics13060918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/09/2021] [Accepted: 06/17/2021] [Indexed: 01/19/2023] Open
Abstract
Organic anion-transporting polypeptide 2B1 (OATP2B1) is co-localized with OATP1B1 and OATP1B3 in the basolateral hepatocyte membrane, where it is thought to contribute to the hepatic uptake of drugs. We characterized a novel Slco2b1(-/-) mouse model using positron emission tomography (PET) imaging with [11C]erlotinib (a putative OATP2B1-selective substrate) and planar scintigraphic imaging with [99mTc]mebrofenin (an OATP1B1/1B3 substrate, which is not transported by OATP2B1). Dynamic 40-min scans were performed after intravenous injection of either [11C]erlotinib or [99mTc]mebrofenin in wild-type and Slco2b1(-/-) mice. A pharmacokinetic model was used to estimate the hepatic uptake clearance (CL1) and the rate constants for transfer of radioactivity from the liver to the blood (k2) and excreted bile (k3). CL1 was significantly reduced in Slco2b1(-/-) mice for both radiotracers (p < 0.05), and k2 was significantly lower (p < 0.01) in Slco2b1(-/-) mice for [11C]erlotinib, but not for [99mTc]mebrofenin. Our data support previous evidence that OATP transporters may contribute to the hepatic uptake of [11C]erlotinib. However, the decreased hepatic uptake of the OATP1B1/1B3 substrate [99mTc]mebrofenin in Slco2b1(-/-) mice questions the utility of this mouse model to assess the relative contribution of OATP2B1 to the liver uptake of drugs which are substrates of multiple OATPs.
Collapse
|
15
|
Ogihara T, Mizoi K, Kamioka H, Yano K. Physiological Roles of ERM Proteins and Transcriptional Regulators in Supporting Membrane Expression of Efflux Transporters as Factors of Drug Resistance in Cancer. Cancers (Basel) 2020; 12:E3352. [PMID: 33198344 PMCID: PMC7696277 DOI: 10.3390/cancers12113352] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/06/2020] [Accepted: 11/10/2020] [Indexed: 12/23/2022] Open
Abstract
One factor contributing to the malignancy of cancer cells is the acquisition of drug resistance during chemotherapy via increased expression of efflux transporters, such as P-glycoprotein (P-gp), multidrug resistance-associated proteins (MRPs), and breast cancer resistance protein (BCRP). These transporters operate at the cell membrane, and are anchored in place by the scaffold proteins ezrin (Ezr), radixin (Rdx), and moesin (Msn) (ERM proteins), which regulate their functional activity. The identity of the regulatory scaffold protein(s) differs depending upon the transporter, and also upon the tissue in which it is expressed, even for the same transporter. Another factor contributing to malignancy is metastatic ability. Epithelial-mesenchymal transition (EMT) is the first step in the conversion of primary epithelial cells into mesenchymal cells that can be transported to other organs via the blood. The SNAI family of transcriptional regulators triggers EMT, and SNAI expression is used is an indicator of malignancy. Furthermore, EMT has been suggested to be involved in drug resistance, since drug excretion from cancer cells is promoted during EMT. We showed recently that ERM proteins are induced by a member of the SNAI family, Snail. Here, we first review recent progress in research on the relationship between efflux transporters and scaffold proteins, including the question of tissue specificity. In the second part, we review the relationship between ERM scaffold proteins and the transcriptional regulatory factors that induce their expression.
Collapse
Affiliation(s)
- Takuo Ogihara
- Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, 60, Nakaorui-machi, Takasaki, Gunma 370-0033, Japan;
| | - Kenta Mizoi
- Faculty of Pharmacy, Takasaki University of Health and Welfare, 60, Nakaorui-machi, Takasaki, Gunma 370-0033, Japan; (K.M.); (K.Y.)
| | - Hiroki Kamioka
- Graduate School of Pharmaceutical Sciences, Takasaki University of Health and Welfare, 60, Nakaorui-machi, Takasaki, Gunma 370-0033, Japan;
| | - Kentaro Yano
- Faculty of Pharmacy, Takasaki University of Health and Welfare, 60, Nakaorui-machi, Takasaki, Gunma 370-0033, Japan; (K.M.); (K.Y.)
- Laboratory of Drug Metabolism and Pharmacokinetics, Yokohama University of Pharmacy, 601, Matano-cho, Totsuka-ku, Yokohama, Kanagawa 245-0066, Japan
| |
Collapse
|
16
|
Wanek T, Zoufal V, Brackhan M, Krohn M, Mairinger S, Filip T, Sauberer M, Stanek J, Pekar T, Pahnke J, Langer O. Brain Distribution of Dual ABCB1/ABCG2 Substrates Is Unaltered in a Beta-Amyloidosis Mouse Model. Int J Mol Sci 2020; 21:E8245. [PMID: 33153231 PMCID: PMC7663372 DOI: 10.3390/ijms21218245] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND ABCB1 (P-glycoprotein) and ABCG2 (breast cancer resistance protein) are co-localized at the blood-brain barrier (BBB), where they restrict the brain distribution of many different drugs. Moreover, ABCB1 and possibly ABCG2 play a role in Alzheimer's disease (AD) by mediating the brain clearance of beta-amyloid (Aβ) across the BBB. This study aimed to compare the abundance and activity of ABCG2 in a commonly used β-amyloidosis mouse model (APP/PS1-21) with age-matched wild-type mice. METHODS The abundance of ABCG2 was assessed by semi-quantitative immunohistochemical analysis of brain slices of APP/PS1-21 and wild-type mice aged 6 months. Moreover, the brain distribution of two dual ABCB1/ABCG2 substrate radiotracers ([11C]tariquidar and [11C]erlotinib) was assessed in APP/PS1-21 and wild-type mice with positron emission tomography (PET). [11C]Tariquidar PET scans were performed without and with partial inhibition of ABCG2 with Ko143, while [11C]erlotinib PET scans were only performed under baseline conditions. RESULTS Immunohistochemical analysis revealed a significant reduction (by 29-37%) in the number of ABCG2-stained microvessels in the brains of APP/PS1-21 mice. Partial ABCG2 inhibition significantly increased the brain distribution of [11C]tariquidar in APP/PS1-21 and wild-type mice, but the brain distribution of [11C]tariquidar did not differ under both conditions between the two mouse strains. Similar results were obtained with [11C]erlotinib. CONCLUSIONS Despite a reduction in the abundance of cerebral ABCG2 and ABCB1 in APP/PS1-21 mice, the brain distribution of two dual ABCB1/ABCG2 substrates was unaltered. Our results suggest that the brain distribution of clinically used ABCB1/ABCG2 substrate drugs may not differ between AD patients and healthy people.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism
- Amyloid beta-Peptides/metabolism
- Amyloid beta-Peptides/toxicity
- Amyloidosis/diagnostic imaging
- Amyloidosis/metabolism
- Amyloidosis/pathology
- Animals
- Blood-Brain Barrier/metabolism
- Brain/diagnostic imaging
- Brain/metabolism
- Disease Models, Animal
- Female
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Positron-Emission Tomography
- Quinolines/pharmacokinetics
- Tissue Distribution
Collapse
Affiliation(s)
- Thomas Wanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria; (V.Z.); (S.M.); (T.F.); (M.S.); (J.S.); (O.L.)
| | - Viktoria Zoufal
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria; (V.Z.); (S.M.); (T.F.); (M.S.); (J.S.); (O.L.)
| | - Mirjam Brackhan
- Department of Neuro-/Pathology, University of Oslo (UiO) and Oslo University Hospital (OUS), 0424 Oslo, Norway; (M.B.); (M.K.); (J.P.)
| | - Markus Krohn
- Department of Neuro-/Pathology, University of Oslo (UiO) and Oslo University Hospital (OUS), 0424 Oslo, Norway; (M.B.); (M.K.); (J.P.)
| | - Severin Mairinger
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria; (V.Z.); (S.M.); (T.F.); (M.S.); (J.S.); (O.L.)
| | - Thomas Filip
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria; (V.Z.); (S.M.); (T.F.); (M.S.); (J.S.); (O.L.)
| | - Michael Sauberer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria; (V.Z.); (S.M.); (T.F.); (M.S.); (J.S.); (O.L.)
| | - Johann Stanek
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria; (V.Z.); (S.M.); (T.F.); (M.S.); (J.S.); (O.L.)
| | - Thomas Pekar
- Biomedical Analytics, University of Applied Sciences Wiener Neustadt, 2700 Wiener Neustadt, Austria;
| | - Jens Pahnke
- Department of Neuro-/Pathology, University of Oslo (UiO) and Oslo University Hospital (OUS), 0424 Oslo, Norway; (M.B.); (M.K.); (J.P.)
- LIED, University of Lübeck, 23562 Lübeck, Germany
- Department of Pharmacology, Faculty of Medicine, University of Latvia, 1586 Rīga, Latvia
| | - Oliver Langer
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, 2444 Seibersdorf, Austria; (V.Z.); (S.M.); (T.F.); (M.S.); (J.S.); (O.L.)
- Department of Clinical Pharmacology, Medical University of Vienna, 1090 Vienna, Austria
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
17
|
Zoetemelk M, Ramzy GM, Rausch M, Koessler T, van Beijnum JR, Weiss A, Mieville V, Piersma SR, de Haas RR, Delucinge-Vivier C, Andres A, Toso C, Henneman AA, Ragusa S, Petrova TV, Docquier M, McKee TA, Jimenez CR, Daali Y, Griffioen AW, Rubbia-Brandt L, Dietrich PY, Nowak-Sliwinska P. Optimized low-dose combinatorial drug treatment boosts selectivity and efficacy of colorectal carcinoma treatment. Mol Oncol 2020; 14:2894-2919. [PMID: 33021054 PMCID: PMC7607171 DOI: 10.1002/1878-0261.12797] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 07/24/2020] [Accepted: 08/11/2020] [Indexed: 12/19/2022] Open
Abstract
The current standard of care for colorectal cancer (CRC) is a combination of chemotherapeutics, often supplemented with targeted biological drugs. An urgent need exists for improved drug efficacy and minimized side effects, especially at late‐stage disease. We employed the phenotypically driven therapeutically guided multidrug optimization (TGMO) technology to identify optimized drug combinations (ODCs) in CRC. We identified low‐dose synergistic and selective ODCs for a panel of six human CRC cell lines also active in heterotypic 3D co‐culture models. Transcriptome sequencing and phosphoproteome analyses showed that the mechanisms of action of these ODCs converged toward MAP kinase signaling and cell cycle inhibition. Two cell‐specific ODCs were translated to in vivo mouse models. The ODCs reduced tumor growth by ~80%, outperforming standard chemotherapy (FOLFOX). No toxicity was observed for the ODCs, while significant side effects were induced in the group treated with FOLFOX therapy. Identified ODCs demonstrated significantly enhanced bioavailability of the individual components. Finally, ODCs were also active in primary cells from CRC patient tumor tissues. Taken together, we show that the TGMO technology efficiently identifies selective and potent low‐dose drug combinations, optimized regardless of tumor mutation status, outperforming conventional chemotherapy.
Collapse
Affiliation(s)
- Marloes Zoetemelk
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland.,Translational Research Center in Oncohaematology, Geneva, Switzerland
| | - George M Ramzy
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland.,Translational Research Center in Oncohaematology, Geneva, Switzerland
| | - Magdalena Rausch
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland.,Translational Research Center in Oncohaematology, Geneva, Switzerland
| | - Thibaud Koessler
- Department of Oncology, Geneva University Hospitals and Faculty of Medicine, Switzerland
| | - Judy R van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC-location VUmc, VU University Amsterdam, The Netherlands
| | - Andrea Weiss
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| | - Valentin Mieville
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland
| | - Sander R Piersma
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands.,OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| | - Richard R de Haas
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands.,OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| | | | - Axel Andres
- Translational Department of Digestive and Transplant Surgery, Geneva University Hospitals and Faculty of Medicine, Switzerland.,Hepato-Pancreato-Biliary Centre, Geneva University Hospitals and Faculty of Medicine, Switzerland
| | - Christian Toso
- Translational Department of Digestive and Transplant Surgery, Geneva University Hospitals and Faculty of Medicine, Switzerland.,Hepato-Pancreato-Biliary Centre, Geneva University Hospitals and Faculty of Medicine, Switzerland
| | - Alexander A Henneman
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands.,OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| | - Simone Ragusa
- Department of Oncology, University of Lausanne, Switzerland.,Ludwig Institute for Cancer Research Lausanne, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne, Switzerland.,Ludwig Institute for Cancer Research Lausanne, Switzerland
| | - Mylène Docquier
- iGE3 Genomics Platform, University of Geneva, Switzerland.,Department of Genetics & Evolution, University of Geneva, Switzerland
| | - Thomas A McKee
- Division of Clinical Pathology, Diagnostic Department, University Hospitals of Geneva (HUG), Switzerland
| | - Connie R Jimenez
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands.,OncoProteomics Laboratory, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, The Netherlands
| | - Youssef Daali
- Division of Clinical Pharmacology and Toxicology, Department of Anaesthesiology, Intensive Care and Emergency Medicine, Geneva University Hospitals, Pharmacology, Switzerland
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC-location VUmc, VU University Amsterdam, The Netherlands
| | - Laura Rubbia-Brandt
- Division of Clinical Pathology, Diagnostic Department, University Hospitals of Geneva (HUG), Switzerland
| | - Pierre-Yves Dietrich
- Translational Research Center in Oncohaematology, Geneva, Switzerland.,Department of Oncology, Geneva University Hospitals and Faculty of Medicine, Switzerland
| | - Patrycja Nowak-Sliwinska
- Molecular Pharmacology Group, School of Pharmaceutical Sciences, University of Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Switzerland.,Translational Research Center in Oncohaematology, Geneva, Switzerland
| |
Collapse
|
18
|
Bongarzone S, Sementa T, Dunn J, Bordoloi J, Sunassee K, Blower PJ, Gee A. Imaging Biotin Trafficking In Vivo with Positron Emission Tomography. J Med Chem 2020; 63:8265-8275. [PMID: 32658479 PMCID: PMC7445742 DOI: 10.1021/acs.jmedchem.0c00494] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The water-soluble vitamin biotin is essential for cellular growth, development, and well-being, but its absorption, distribution, metabolism, and excretion are poorly understood. This paper describes the radiolabeling of biotin with the positron emission tomography (PET) radionuclide carbon-11 ([11C]biotin) to enable the quantitative study of biotin trafficking in vivo. We show that intravenously administered [11C]biotin is quickly distributed to the liver, kidneys, retina, heart, and brain in rodents-consistent with the known expression of the biotin transporter-and there is a surprising accumulation in the brown adipose tissue (BAT). Orally administered [11C]biotin was rapidly absorbed in the small intestine and swiftly distributed to the same organs. Preadministration of nonradioactive biotin inhibited organ uptake and increased excretion. [11C]Biotin PET imaging therefore provides a dynamic in vivo map of transporter-mediated biotin trafficking in healthy rodents. This technique will enable the exploration of biotin trafficking in humans and its use as a research tool for diagnostic imaging of obesity/diabetes, bacterial infection, and cancer.
Collapse
Affiliation(s)
- Salvatore Bongarzone
- School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Teresa Sementa
- School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Joel Dunn
- School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Jayanta Bordoloi
- School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Kavitha Sunassee
- School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Philip J Blower
- School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| | - Antony Gee
- School of Biomedical Engineering & Imaging Sciences, St Thomas' Hospital, King's College London, London SE1 7EH, United Kingdom
| |
Collapse
|
19
|
Hernández Lozano I, Langer O. Use of imaging to assess the activity of hepatic transporters. Expert Opin Drug Metab Toxicol 2020; 16:149-164. [PMID: 31951754 PMCID: PMC7055509 DOI: 10.1080/17425255.2020.1718107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/15/2020] [Indexed: 12/13/2022]
Abstract
Introduction: Membrane transporters of the SLC and ABC families are abundantly expressed in the liver, where they control the transfer of drugs/drug metabolites across the sinusoidal and canalicular hepatocyte membranes and play a pivotal role in hepatic drug clearance. Noninvasive imaging methods, such as PET, SPECT or MRI, allow for measuring the activity of hepatic transporters in vivo, provided that suitable transporter imaging probes are available.Areas covered: We give an overview of the working principles of imaging-based assessment of hepatic transporter activity. We discuss different currently available PET/SPECT radiotracers and MRI contrast agents and their applications to measure hepatic transporter activity in health and disease. We cover mathematical modeling approaches to obtain quantitative parameters of transporter activity and provide a critical assessment of methodological limitations and challenges associated with this approach.Expert opinion: PET in combination with pharmacokinetic modeling can be potentially applied in drug development to study the distribution of new drug candidates to the liver and their clearance mechanisms. This approach bears potential to mechanistically assess transporter-mediated drug-drug interactions, to assess the influence of disease on hepatic drug disposition and to validate and refine currently available in vitro-in vivo extrapolation methods to predict hepatic clearance of drugs.
Collapse
Affiliation(s)
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| |
Collapse
|
20
|
Gomez-Zepeda D, Taghi M, Scherrmann JM, Decleves X, Menet MC. ABC Transporters at the Blood-Brain Interfaces, Their Study Models, and Drug Delivery Implications in Gliomas. Pharmaceutics 2019; 12:pharmaceutics12010020. [PMID: 31878061 PMCID: PMC7022905 DOI: 10.3390/pharmaceutics12010020] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 12/13/2019] [Accepted: 12/20/2019] [Indexed: 12/22/2022] Open
Abstract
Drug delivery into the brain is regulated by the blood-brain interfaces. The blood-brain barrier (BBB), the blood-cerebrospinal fluid barrier (BCSFB), and the blood-arachnoid barrier (BAB) regulate the exchange of substances between the blood and brain parenchyma. These selective barriers present a high impermeability to most substances, with the selective transport of nutrients and transporters preventing the entry and accumulation of possibly toxic molecules, comprising many therapeutic drugs. Transporters of the ATP-binding cassette (ABC) superfamily have an important role in drug delivery, because they extrude a broad molecular diversity of xenobiotics, including several anticancer drugs, preventing their entry into the brain. Gliomas are the most common primary tumors diagnosed in adults, which are often characterized by a poor prognosis, notably in the case of high-grade gliomas. Therapeutic treatments frequently fail due to the difficulty of delivering drugs through the brain barriers, adding to diverse mechanisms developed by the cancer, including the overexpression or expression de novo of ABC transporters in tumoral cells and/or in the endothelial cells forming the blood-brain tumor barrier (BBTB). Many models have been developed to study the phenotype, molecular characteristics, and function of the blood-brain interfaces as well as to evaluate drug permeability into the brain. These include in vitro, in vivo, and in silico models, which together can help us to better understand their implication in drug resistance and to develop new therapeutics or delivery strategies to improve the treatment of pathologies of the central nervous system (CNS). In this review, we present the principal characteristics of the blood-brain interfaces; then, we focus on the ABC transporters present on them and their implication in drug delivery; next, we present some of the most important models used for the study of drug transport; finally, we summarize the implication of ABC transporters in glioma and the BBTB in drug resistance and the strategies to improve the delivery of CNS anticancer drugs.
Collapse
Affiliation(s)
- David Gomez-Zepeda
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| | - Méryam Taghi
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Jean-Michel Scherrmann
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
| | - Xavier Decleves
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Biologie du médicament et toxicologie, Hôpital Cochin, AP HP, 75006 Paris, France
| | - Marie-Claude Menet
- Inserm, UMR-S 1144, Optimisation Thérapeutique en Neuropsychopharmacologie, 75006 Paris, France; (M.T.); (J.-M.S.); (X.D.)
- Sorbonne Paris Cité, Université Paris Descartes, 75006 Paris, France
- Sorbonne Paris Cité, Université Paris Diderot, 75013 Paris, France
- UF Hormonologie adulte, Hôpital Cochin, AP HP, 75006 Paris, France
- Correspondence: (D.G.-Z.); (M.-C.M.)
| |
Collapse
|
21
|
Wulkersdorfer B, Bauer M, Karch R, Stefanits H, Philippe C, Weber M, Czech T, Menet MC, Declèves X, Hainfellner JA, Preusser M, Hacker M, Zeitlinger M, Müller M, Langer O. Assessment of brain delivery of a model ABCB1/ABCG2 substrate in patients with non-contrast-enhancing brain tumors with positron emission tomography. EJNMMI Res 2019; 9:110. [PMID: 31832814 PMCID: PMC6908538 DOI: 10.1186/s13550-019-0581-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/04/2019] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) are two efflux transporters expressed at the blood-brain barrier which effectively restrict the brain distribution of the majority of currently known anticancer drugs. High-grade brain tumors often possess a disrupted blood-brain tumor barrier (BBTB) leading to enhanced accumulation of magnetic resonance imaging contrast agents, and possibly anticancer drugs, as compared to normal brain. In contrast to high-grade brain tumors, considerably less information is available with respect to BBTB integrity in lower grade brain tumors. MATERIALS AND METHODS We performed positron emission tomography imaging with the radiolabeled ABCB1 inhibitor [11C]tariquidar, a prototypical ABCB1/ABCG2 substrate, in seven patients with non-contrast -enhancing brain tumors (WHO grades I-III). In addition, ABCB1 and ABCG2 levels were determined in surgically resected tumor tissue of four patients using quantitative targeted absolute proteomics. RESULTS Brain distribution of [11C]tariquidar was found to be very low across the whole brain and not significantly different between tumor and tumor-free brain tissue. Only one patient showed a small area of enhanced [11C]tariquidar uptake within the brain tumor. ABCG2/ABCB1 ratios in surgically resected tumor tissue (1.4 ± 0.2) were comparable to previously reported ABCG2/ABCB1 ratios in isolated human micro-vessels (1.3), which suggested that no overexpression of ABCB1 or ABCG2 occurred in the investigated tumors. CONCLUSIONS Our data suggest that the investigated brain tumors had an intact BBTB, which is impermeable to anticancer drugs, which are dual ABCB1/ABCG2 substrates. Therefore, effective drugs for antitumor treatment should have high passive permeability and lack ABCB1/ABCG2 substrate affinity. TRIAL REGISTRATION European Union Drug Regulating Authorities Clinical Trials Database (EUDRACT), 2011-004189-13. Registered on 23 February 2012, https://www.clinicaltrialsregister.eu/ctr-search/search?query=2011-004189-13.
Collapse
Affiliation(s)
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Rudolf Karch
- Centre for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Harald Stefanits
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Cécile Philippe
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Maria Weber
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Thomas Czech
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Marie-Claude Menet
- Inserm, U1144, Paris, France.,Université Paris Descartes, UMR-S 1144, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Xavier Declèves
- Inserm, U1144, Paris, France.,Université Paris Descartes, UMR-S 1144, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | | | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Markus Müller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria. .,Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria. .,Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria.
| |
Collapse
|
22
|
Liu Y, Zhang L, Ma Z, Tian L, Liu Y, Liu Y, Chen Q, Li Y, Ma E. Ascorbate promotes the cellular accumulation of doxorubicin and reverses the multidrug resistance in breast cancer cells by inducing ROS-dependent ATP depletion. Free Radic Res 2019; 53:758-767. [DOI: 10.1080/10715762.2019.1628957] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Yutong Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Ling Zhang
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Zitai Ma
- Shenyang No. 2 Middle School, Shenyang, China
| | - Li Tian
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Yingchi Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Yuqing Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| | - Qi Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, MO, USA
| | - Yanchun Li
- GLP Center, Shenyang Pharmaceutical University, Shenyang, China
| | - Enlong Ma
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
23
|
Linder M, Glitzner E, Srivatsa S, Bakiri L, Matsuoka K, Shahrouzi P, Dumanic M, Novoszel P, Mohr T, Langer O, Wanek T, Mitterhauser M, Wagner EF, Sibilia M. EGFR is required for FOS-dependent bone tumor development via RSK2/CREB signaling. EMBO Mol Med 2019; 10:emmm.201809408. [PMID: 30361264 PMCID: PMC6220323 DOI: 10.15252/emmm.201809408] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma (OS) is a rare tumor of the bone occurring mainly in young adults accounting for 5% of all childhood cancers. Because of the limited therapeutic options, there has been no survival improvement for OS patients in the past 40 years. The epidermal growth factor receptor (EGFR) is highly expressed in OS; however, its clinical relevance is unclear. Here, we employed an autochthonous c‐Fos‐dependent OS mouse model (H2‐c‐fosLTR) and human OS tumor biopsies for preclinical studies aimed at identifying novel biomarkers and therapeutic benefits of anti‐EGFR therapies. We show that EGFR deletion/inhibition results in reduced tumor formation in H2‐c‐fosLTR mice by directly inhibiting the proliferation of cancer‐initiating osteoblastic cells by a mechanism involving RSK2/CREB‐dependent c‐Fos expression. Furthermore, OS patients with co‐expression of EGFR and c‐Fos exhibit reduced overall survival. Preclinical studies using human OS xenografts revealed that only tumors expressing both EGFR and c‐Fos responded to anti‐EGFR therapy demonstrating that c‐Fos can be considered as a novel biomarker predicting response to anti‐EGFR treatment in OS patients.
Collapse
Affiliation(s)
- Markus Linder
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Elisabeth Glitzner
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Sriram Srivatsa
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Latifa Bakiri
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | | | - Parastoo Shahrouzi
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Monika Dumanic
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Philipp Novoszel
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Thomas Mohr
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.,Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Wanek
- Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Markus Mitterhauser
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.,LBI Applied Diagnostics, Vienna, Austria
| | - Erwin F Wagner
- Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Maria Sibilia
- Department of Medicine I, Comprehensive Cancer Center, Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
24
|
Hernández Lozano I, Karch R, Bauer M, Blaickner M, Matsuda A, Wulkersdorfer B, Hacker M, Zeitlinger M, Langer O. Towards Improved Pharmacokinetic Models for the Analysis of Transporter-Mediated Hepatic Disposition of Drug Molecules with Positron Emission Tomography. AAPS J 2019; 21:61. [PMID: 31037511 PMCID: PMC6488550 DOI: 10.1208/s12248-019-0323-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/19/2019] [Indexed: 12/19/2022] Open
Abstract
Positron emission tomography (PET) imaging with radiolabeled drugs holds great promise to assess the influence of membrane transporters on hepatobiliary clearance of drugs. To exploit the full potential of PET, quantitative pharmacokinetic models are required. In this study, we evaluated the suitability of different compartment models to describe the hepatic disposition of [11C]erlotinib as a small-molecule model drug which undergoes transporter-mediated hepatobiliary excretion. We analyzed two different, previously published data sets in healthy volunteers, in which a baseline [11C]erlotinib PET scan was followed by a second PET scan either after oral intake of unlabeled erlotinib (300 mg) or after intravenous infusion of the prototypical organic anion-transporting polypeptide inhibitor rifampicin (600 mg). We assessed a three-compartment (3C) and a four-compartment (4C) model, in which either a sampled arterial blood input function or a mathematically derived dual input function (DIF), which takes the contribution of the portal vein to the liver blood supply into account, was used. Both models provided acceptable fits of the observed PET data in the liver and extrahepatic bile duct and gall bladder. Changes in model outcome parameters between scans were consistent with the involvement of basolateral hepatocyte uptake and canalicular efflux transporters in the hepatobiliary clearance of [11C]erlotinib. Our results demonstrated that inclusion of a DIF did not lead to substantial improvements in model fits. The models developed in this work represent a step forward in applying PET as a tool to assess the impact of hepatic transporters on drug disposition and their involvement in drug-drug interactions.
Collapse
Affiliation(s)
- Irene Hernández Lozano
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Rudolf Karch
- Centre for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Matthias Blaickner
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Akihiro Matsuda
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Beatrix Wulkersdorfer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090, Vienna, Austria.
- Preclinical Molecular Imaging, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria.
- Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
25
|
Traxl A, Mairinger S, Filip T, Sauberer M, Stanek J, Poschner S, Jäger W, Zoufal V, Novarino G, Tournier N, Bauer M, Wanek T, Langer O. Inhibition of ABCB1 and ABCG2 at the Mouse Blood-Brain Barrier with Marketed Drugs To Improve Brain Delivery of the Model ABCB1/ABCG2 Substrate [ 11C]erlotinib. Mol Pharm 2019; 16:1282-1293. [PMID: 30694684 DOI: 10.1021/acs.molpharmaceut.8b01217] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
P-Glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) are two efflux transporters at the blood-brain barrier (BBB), which effectively restrict brain distribution of diverse drugs, such as tyrosine kinase inhibitors. There is a crucial need for pharmacological ABCB1 and ABCG2 inhibition protocols for a more effective treatment of brain diseases. In the present study, seven marketed drugs (osimertinib, erlotinib, nilotinib, imatinib, lapatinib, pazopanib, and cyclosporine A) and one nonmarketed drug (tariquidar), with known in vitro ABCB1/ABCG2 inhibitory properties, were screened for their inhibitory potency at the BBB in vivo. Positron emission tomography (PET) using the model ABCB1/ABCG2 substrate [11C]erlotinib was performed in mice. Tested inhibitors were administered as i.v. bolus injections at 30 min before the start of the PET scan, followed by a continuous i.v. infusion for the duration of the PET scan. Five of the tested drugs increased total distribution volume of [11C]erlotinib in the brain ( VT,brain) compared to vehicle-treated animals (tariquidar, + 69%; erlotinib, + 19% and +23% for the 21.5 mg/kg and the 43 mg/kg dose, respectively; imatinib, + 22%; lapatinib, + 25%; and cyclosporine A, + 49%). For all drugs, increases in [11C]erlotinib brain distribution were lower than in Abcb1a/b(-/-)Abcg2(-/-) mice (+149%), which suggested that only partial ABCB1/ABCG2 inhibition was reached at the mouse BBB. The plasma concentrations of the tested drugs at the time of the PET scan were higher than clinically achievable plasma concentrations. Some of the tested drugs led to significant increases in blood radioactivity concentrations measured at the end of the PET scan (erlotinib, + 103% and +113% for the 21.5 mg/kg and the 43 mg/kg dose, respectively; imatinib, + 125%; and cyclosporine A, + 101%), which was most likely caused by decreased hepatobiliary excretion of radioactivity. Taken together, our data suggest that some marketed tyrosine kinase inhibitors may be repurposed to inhibit ABCB1 and ABCG2 at the BBB. From a clinical perspective, moderate increases in brain delivery despite the administration of high i.v. doses as well as peripheral drug-drug interactions due to transporter inhibition in clearance organs question the translatability of this concept.
Collapse
Affiliation(s)
- Alexander Traxl
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Severin Mairinger
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Thomas Filip
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Michael Sauberer
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Johann Stanek
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Stefan Poschner
- Department of Clinical Pharmacy and Diagnostics , University of Vienna , 1090 Vienna , Austria
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics , University of Vienna , 1090 Vienna , Austria
| | - Viktoria Zoufal
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Gaia Novarino
- Institute of Science and Technology (IST) Austria , 3400 Klosterneuburg , Austria
| | - Nicolas Tournier
- UMR 1023 IMIV, Service Hospitalier Frédéric Joliot , CEA, Inserm, Univ. Paris Sud, CNRS, Université Paris-Saclay , 91450 Orsay , France
| | - Martin Bauer
- Department of Clinical Pharmacology , Medical University of Vienna , 1090 Vienna , Austria
| | - Thomas Wanek
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | - Oliver Langer
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria.,Department of Clinical Pharmacology , Medical University of Vienna , 1090 Vienna , Austria.,Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine , Medical University of Vienna , 1090 Vienna , Austria
| |
Collapse
|
26
|
Goutal S, Gerstenmayer M, Auvity S, Caillé F, Mériaux S, Buvat I, Larrat B, Tournier N. Physical blood-brain barrier disruption induced by focused ultrasound does not overcome the transporter-mediated efflux of erlotinib. J Control Release 2018; 292:210-220. [PMID: 30415015 DOI: 10.1016/j.jconrel.2018.11.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 12/17/2022]
Abstract
Overcoming the efflux mediated by ATP-binding cassette (ABC) transporters at the blood-brain barrier (BBB) remains a challenge for the delivery of small molecule tyrosine kinase inhibitors (TKIs) such as erlotinib to the brain. Inhibition of ABCB1 and ABCG2 at the mouse BBB improved the BBB permeation of erlotinib but could not be achieved in humans. BBB disruption induced by focused ultrasound (FUS) was investigated as a strategy to overcome the efflux transport of erlotinib in vivo. In rats, FUS combined with microbubbles allowed for a large and spatially controlled disruption of the BBB in the left hemisphere. ABCB1/ABCG2 inhibition was performed using elacridar (10 mg/kg i.v). The brain kinetics of erlotinib was studied using 11C-erlotinib Positron Emission Tomography (PET) imaging in 5 groups (n = 4-5 rats per group) including a baseline group, immediately after sonication (FUS), 48 h after FUS (FUS + 48 h), elacridar (ELA) and their combination (FUS + ELA). BBB integrity was assessed using the Evan's Blue (EB) extravasation test. Brain exposure to 11C-erlotinib was measured as the area under the curve (AUC) of the brain kinetics (% injected dose (%ID) versus time (min)) in volumes corresponding to the disrupted (left) and the intact (right) hemispheres, respectively. EB extravasation highlighted BBB disruption in the left hemisphere of animals of the FUS and FUS + ELA groups but not in the control and ELA groups. EB extravasation was not observed 48 h after FUS suggesting recovery of BBB integrity. Compared with the control group (AUCBaseline = 1.4 ± 0.5%ID.min), physical BBB disruption did not impact the brain kinetics of 11C-erlotinib in the left hemisphere (p > .05) either immediately (AUCFUS = 1.2 ± 0.1%ID.min) or 48 h after FUS (AUCFUS+48h = 1.1 ± 0.3%ID.min). Elacridar similarly increased 11C-erlotinib brain exposure to the left hemisphere in the absence (AUCELA = 2.2 ± 0.5%ID.min, p < .001) and in the presence of BBB disruption (AUCFUS+ELA = 2.1 ± 0.5%ID.min, p < .001). AUCleft was never significantly different from AUCright (p > .05), in any of the tested conditions. BBB integrity is not the rate limiting step for erlotinib delivery to the brain which is mainly governed by ABC-mediated efflux. Efflux transport of erlotinib persisted despite BBB disruption.
Collapse
Affiliation(s)
- Sébastien Goutal
- Imagerie Moléculaire In Vivo, IMIV, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France; Molecular Imaging Research Center, MIRCen, Institut de Biologie François Jacob, Direction de la Recherche Fondamentale, CEA, Fontenay-Aux-Roses, France
| | - Matthieu Gerstenmayer
- Neurospin, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Université Paris Saclay, Gif sur Yvette, France
| | - Sylvain Auvity
- Imagerie Moléculaire In Vivo, IMIV, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Fabien Caillé
- Imagerie Moléculaire In Vivo, IMIV, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Sébastien Mériaux
- Neurospin, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Université Paris Saclay, Gif sur Yvette, France
| | - Irène Buvat
- Imagerie Moléculaire In Vivo, IMIV, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Benoit Larrat
- Neurospin, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Université Paris Saclay, Gif sur Yvette, France
| | - Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, Institut des sciences du vivant Frédéric Joliot, Direction de la Recherche Fondamentale, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France.
| |
Collapse
|
27
|
Bauer M, Karch R, Wulkersdorfer B, Philippe C, Nics L, Klebermass EM, Weber M, Poschner S, Haslacher H, Jäger W, Tournier N, Wadsak W, Hacker M, Zeitlinger M, Langer O. A Proof-of-Concept Study to Inhibit ABCG2- and ABCB1-Mediated Efflux Transport at the Human Blood-Brain Barrier. J Nucl Med 2018; 60:486-491. [PMID: 30237210 DOI: 10.2967/jnumed.118.216432] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 09/10/2018] [Indexed: 02/06/2023] Open
Abstract
The adenosine triphosphate-binding cassette transporters P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2) are 2 efflux transporters at the blood-brain barrier (BBB) that effectively restrict brain distribution of dual ABCB1/ABCG2 substrate drugs, such as tyrosine kinase inhibitors. Pharmacologic inhibition of ABCB1/ABCG2 may improve the efficacy of dual-substrate drugs for treatment of brain tumors, but no marketed ABCB1/ABCG2 inhibitors are currently available. In the present study, we examined the potential of supratherapeutic-dose oral erlotinib to inhibit ABCB1/ABCG2 activity at the human BBB. Methods: Healthy men underwent 2 consecutive PET scans with 11C-erlotinib: a baseline scan and a second scan either with concurrent intravenous infusion of the ABCB1 inhibitor tariquidar (3.75 mg/min, n = 5) or after oral intake of single ascending doses of erlotinib (300 mg, n = 7; 650 mg, n = 8; or 1,000 mg, n = 2). Results: Although tariquidar administration had no effect on 11C-erlotinib brain distribution, oral erlotinib led, at the 650-mg dose, to significant increases in volume of distribution (23% ± 13%, P = 0.008), influx rate constant of radioactivity from plasma into brain (58% ± 26%, P = 0.008), and area under the brain time-activity curve (78% ± 17%, P = 0.008), presumably because of combined partial saturation of ABCG2 and ABCB1 activity. Inclusion of further subjects into the 1,000-mg dose group was precluded by adverse skin events (rash). Conclusion: Supratherapeutic-dose erlotinib may be used to enhance brain delivery of ABCB1/ABCG2 substrate anticancer drugs, but its clinical applicability for continuous ABCB1/ABCG2 inhibition at the BBB may be limited by safety concerns.
Collapse
Affiliation(s)
- Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Rudolf Karch
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | | | - Cécile Philippe
- Division of Nuclear Medicine, Department of Biomedical Imaging und Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Lukas Nics
- Division of Nuclear Medicine, Department of Biomedical Imaging und Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Eva-Maria Klebermass
- Division of Nuclear Medicine, Department of Biomedical Imaging und Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Maria Weber
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Stefan Poschner
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Helmuth Haslacher
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Nicolas Tournier
- IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Wolfgang Wadsak
- Division of Nuclear Medicine, Department of Biomedical Imaging und Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.,Center for Biomarker Research in Medicine, CBmed GmbH, Graz, Austria; and
| | - Marcus Hacker
- Division of Nuclear Medicine, Department of Biomedical Imaging und Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Division of Nuclear Medicine, Department of Biomedical Imaging und Image-Guided Therapy, Medical University of Vienna, Vienna, Austria.,Center for Health and Bioresources, Austrian Institute of Technology GmbH, Seibersdorf, Austria
| |
Collapse
|
28
|
Bauer M, Traxl A, Matsuda A, Karch R, Philippe C, Nics L, Klebermass EM, Wulkersdorfer B, Weber M, Poschner S, Tournier N, Jäger W, Wadsak W, Hacker M, Wanek T, Zeitlinger M, Langer O. Effect of Rifampicin on the Distribution of [ 11C]Erlotinib to the Liver, a Translational PET Study in Humans and in Mice. Mol Pharm 2018; 15:4589-4598. [PMID: 30180590 DOI: 10.1021/acs.molpharmaceut.8b00588] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Organic anion-transporting polypeptides (OATPs) mediate the uptake of various drugs from blood into the liver in the basolateral membrane of hepatocytes. Positron emission tomography (PET) is a potentially powerful tool to assess the activity of hepatic OATPs in vivo, but its utility critically depends on the availability of transporter-selective probe substrates. We have shown before that among the three OATPs expressed in hepatocytes (OATP1B1, OATP1B3, and OATP2B1), [11C]erlotinib is selectively transported by OATP2B1. In contrast to OATP1B1 and OATP1B3, OATP2B1 has not been thoroughly explored yet, and no specific probe substrates are currently available. To assess if the prototypical OATP inhibitor rifampicin can inhibit liver uptake of [11C]erlotinib in vivo, we performed [11C]erlotinib PET scans in six healthy volunteers without and with intravenous pretreatment with rifampicin (600 mg). In addition, FVB mice underwent [11C]erlotinib PET scans without and with concurrent intravenous infusion of high-dose rifampicin (100 mg/kg). Rifampicin caused a moderate reduction in the liver distribution of [11C]erlotinib in humans, while a more pronounced effect of rifampicin was observed in mice, in which rifampicin plasma concentrations were higher than in humans. In vitro uptake experiments in an OATP2B1-overexpressing cell line indicated that rifampicin inhibited OATP2B1 transport of [11C]erlotinib in a concentration-dependent manner with a half-maximum inhibitory concentration of 72.0 ± 1.4 μM. Our results suggest that rifampicin-inhibitable uptake transporter(s) contributed to the liver distribution of [11C]erlotinib in humans and mice and that [11C]erlotinib PET in combination with rifampicin may be used to measure the activity of this/these uptake transporter(s) in vivo. Furthermore, our data suggest that a standard clinical dose of rifampicin may exert in vivo a moderate inhibitory effect on hepatic OATP2B1.
Collapse
Affiliation(s)
| | - Alexander Traxl
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | | | | | | | | | | | | | | | - Stefan Poschner
- Department of Clinical Pharmacy and Diagnostics , University of Vienna , A-1090 Vienna , Austria
| | - Nicolas Tournier
- IMIV, CEA, Inserm, CNRS , Université Paris-Sud, Université Paris Saclay, CEA-SHFJ , 91401 Orsay , France
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics , University of Vienna , A-1090 Vienna , Austria
| | - Wolfgang Wadsak
- Center for Biomarker Research in Medicine - CBmed GmbH , 8010 Graz , Austria
| | | | - Thomas Wanek
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| | | | - Oliver Langer
- Center for Health & Bioresources , AIT Austrian Institute of Technology GmbH , 2444 Seibersdorf , Austria
| |
Collapse
|
29
|
Amor D, Goutal S, Marie S, Caillé F, Bauer M, Langer O, Auvity S, Tournier N. Impact of rifampicin-inhibitable transport on the liver distribution and tissue kinetics of erlotinib assessed with PET imaging in rats. EJNMMI Res 2018; 8:81. [PMID: 30116910 PMCID: PMC6095934 DOI: 10.1186/s13550-018-0434-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023] Open
Abstract
Background Erlotinib is an epidermal growth factor receptor (EGFR)-targeting tyrosine kinase inhibitor approved for treatment of non-small cell lung cancer. The wide inter-individual pharmacokinetic (PK) variability of erlotinib may impact treatment outcome and/or toxicity. Recent in vivo studies reported a nonlinear uptake transport of erlotinib into the liver, suggesting carrier-mediated system(s) to mediate its hepatobiliary clearance. Erlotinib has been identified in vitro as a substrate of organic anion-transporting polypeptide (OATP) transporters which expression does not restrict to hepatocytes and may impact the tissue uptake of erlotinib in vivo. Results The impact of rifampicin (40 mg/kg), a potent OATP inhibitor, on the liver uptake and exposure to tissues of 11C-erlotinib was investigated in rats (4 animals per group) using positron emission tomography (PET) imaging. Tissue pharmacokinetics (PK) and corresponding exposure (area under the curve, AUC) were assessed in the liver, kidney cortex, abdominal aorta (blood pool) and the lungs. The plasma PK of parent 11C-erlotinib was also measured using arterial blood sampling to estimate the transfer rate constant (kuptake) of 11C-erlotinib from plasma into different tissues. PET images unveiled the predominant distribution of 11C-erlotinib-associated radioactivity to the liver, which gradually moved to the intestine, thus highlighting hepatobiliary clearance. 11C-erlotinib also accumulated in the kidney cortex. Rifampicin did not impact AUCaorta but reduced kuptake, liver (p < 0.001), causing a significant 27.3% decrease in liver exposure (p < 0.001). Moreover, a significant decrease in kuptake, kidney with a concomitant decrease in AUCkidney (− 30.4%, p < 0.001) were observed. Rifampicin neither affected kuptake, lung nor AUClung. Conclusions Our results suggest that 11C-erlotinib is an in vivo substrate of rOATP transporters expressed in the liver and possibly of rifampicin-inhibitable transporter(s) in the kidneys. Decreased 11C-erlotinib uptake by elimination organs did not translate into changes in systemic exposure and exposure to the lungs, which are a target tissue for erlotinib therapy.
Collapse
Affiliation(s)
- Dorra Amor
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France
| | - Sébastien Goutal
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France
| | - Solène Marie
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France
| | - Fabien Caillé
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.,Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Sylvain Auvity
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France
| | - Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, F-91400, Orsay, France. .,CEA, DRF, JOLIOT, Service Hospitalier Frédéric Joliot, F-91401, Orsay, France.
| |
Collapse
|
30
|
Robey RW, Pluchino KM, Hall MD, Fojo AT, Bates SE, Gottesman MM. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat Rev Cancer 2018; 18:452-464. [PMID: 29643473 PMCID: PMC6622180 DOI: 10.1038/s41568-018-0005-8] [Citation(s) in RCA: 1145] [Impact Index Per Article: 190.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Most patients who die of cancer have disseminated disease that has become resistant to multiple therapeutic modalities. Ample evidence suggests that the expression of ATP-binding cassette (ABC) transporters, especially the multidrug resistance protein 1 (MDR1, also known as P-glycoprotein or P-gp), which is encoded by ABC subfamily B member 1 (ABCB1), can confer resistance to cytotoxic and targeted chemotherapy. However, the development of MDR1 as a therapeutic target has been unsuccessful. At the time of its discovery, appropriate tools for the characterization and clinical development of MDR1 as a therapeutic target were lacking. Thirty years after the initial cloning and characterization of MDR1 and the implication of two additional ABC transporters, the multidrug resistance-associated protein 1 (MRP1; encoded by ABCC1)), and ABCG2, in multidrug resistance, interest in investigating these transporters as therapeutic targets has waned. However, with the emergence of new data and advanced techniques, we propose to re-evaluate whether these transporters play a clinical role in multidrug resistance. With this Opinion article, we present recent evidence indicating that it is time to revisit the investigation into the role of ABC transporters in efficient drug delivery in various cancer types and at the blood-brain barrier.
Collapse
Affiliation(s)
- Robert W Robey
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kristen M Pluchino
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Matthew D Hall
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Antonio T Fojo
- Division of Hematology/Oncology, Department of Medicine, Columbia University/New York Presbyterian Hospital, Manhattan, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Susan E Bates
- Division of Hematology/Oncology, Department of Medicine, Columbia University/New York Presbyterian Hospital, Manhattan, NY, USA
- James J. Peters VA Medical Center, Bronx, NY, USA
| | - Michael M Gottesman
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
31
|
Therapeutic Potential and Utility of Elacridar with Respect to P-glycoprotein Inhibition: An Insight from the Published In Vitro, Preclinical and Clinical Studies. Eur J Drug Metab Pharmacokinet 2018; 42:915-933. [PMID: 28374336 DOI: 10.1007/s13318-017-0411-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The occurrence of efflux mechanisms via Permeability-glycoprotein (P-gp) recognized as an important physiological process impedes drug entry or transport across membranes into tissues. In some instances, either low oral bioavailability or lack of brain penetration has been attributed to P-gp mediated efflux activity. Therefore, the objective of development of P-gp inhibitors was to facilitate the attainment of higher drug exposures in tissues. Many third-generation P-gp inhibitors such as elacridar, tariquidar, zosuquidar, etc. have entered clinical development to fulfil the promise. The body of evidence from in vitro and in vivo preclinical and clinical data reviewed in this paper provides the basis for an effective blockade of P-gp efflux mechanism by elacridar. However, clinical translation of the promise has been elusive not just for elacridar but also for other P-gp inhibitors in this class. The review provides introspection and perspectives on the lack of clinical translation of this class of drugs and a broad framework of strategies and considerations in the potential application of elacridar and other P-gp inhibitors in oncology therapeutics.
Collapse
|
32
|
Positron Emission Tomography Imaging Reveals an Importance of Saturable Liver Uptake Transport for the Pharmacokinetics of Metoclopramide. CONTRAST MEDIA & MOLECULAR IMAGING 2018; 2018:7310146. [PMID: 29853808 PMCID: PMC5964550 DOI: 10.1155/2018/7310146] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/14/2018] [Accepted: 04/02/2018] [Indexed: 12/14/2022]
Abstract
Positron emission tomography (PET) imaging using [11C]metoclopramide, a P-glycoprotein (P-gp) substrate, was used to investigate the contribution of transport processes to metoclopramide liver clearance. The liver kinetics obtained after injection of [11C]metoclopramide were measured using PET in rats (n=4‐5) in the absence (tracer dose) and the presence of a pharmacologic dose of metoclopramide (3 mg/kg), with or without P-gp inhibition using i.v. tariquidar (8 mg/kg). Corresponding [11C]metoclopramide kinetics and metabolism in plasma (n=3) were measured using radio-HPLC analysis. [11C]metoclopramide exposure to the liver and plasma was described by the area under the time-activity curve (AUC) of the radioactivity kinetics in the liver and parent [11C]metoclopramide kinetics in plasma, respectively. The pharmacologic dose of metoclopramide resulted in a ∼2.2-fold increase in [11C]metoclopramide AUCplasma, while P-gp inhibition did not. AUCliver was lower using the pharmacologic dose (42.9 ± 13.8 SUV·min) compared with the tracer dose (210.0 ± 32.4 SUV·min). P-gp inhibition enhanced the liver exposure in the pharmacologic condition only (81.0 ± 3.1 SUV·min). [11C]metoclopramide PET imaging suggests an unpredicted role for hepatocyte uptake transporter(s) in controlling metoclopramide pharmacokinetics in addition to the known contribution of the metabolic enzymes and the P-gp.
Collapse
|
33
|
Abstract
Transporter systems involved in the permeation of drugs and solutes across biological membranes are recognized as key determinants of pharmacokinetics. Typically, the action of membrane transporters on drug exposure to tissues in living organisms is inferred from invasive procedures, which cannot be applied in humans. In recent years, imaging methods have greatly progressed in terms of instruments, synthesis of novel imaging probes as well as tools for data analysis. Imaging allows pharmacokinetic parameters in different tissues and organs to be obtained in a non-invasive or minimally invasive way. The aim of this overview is to summarize the current status in the field of molecular imaging of drug transporters. The overview is focused on human studies, both for the characterization of transport systems for imaging agents as well as for the determination of drug pharmacokinetics, and makes reference to animal studies where necessary. We conclude that despite certain methodological limitations, imaging has a great potential to study transporters at work in humans and that imaging will become an important tool, not only in drug development but also in medicine. Imaging allows the mechanistic aspects of transport proteins to be studied, as well as elucidating the influence of genetic background, pathophysiological states and drug-drug interactions on the function of transporters involved in the disposition of drugs.
Collapse
Affiliation(s)
- Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
34
|
Mairinger S, Zoufal V, Wanek T, Traxl A, Filip T, Sauberer M, Stanek J, Kuntner C, Pahnke J, Müller M, Langer O. Influence of breast cancer resistance protein and P-glycoprotein on tissue distribution and excretion of Ko143 assessed with PET imaging in mice. Eur J Pharm Sci 2018; 115:212-222. [PMID: 29360507 PMCID: PMC5884419 DOI: 10.1016/j.ejps.2018.01.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/18/2018] [Accepted: 01/18/2018] [Indexed: 12/12/2022]
Abstract
Ko143 is a reference inhibitor of the adenosine triphosphate-binding cassette (ABC) transporter breast cancer resistance protein (humans: ABCG2, rodents: Abcg2) for in vitro and in vivo use. Previous in vitro data indicate that Ko143 binds specifically to ABCG2/Abcg2, suggesting a potential utility of Ko143 as a positron emission tomography (PET) tracer to assess the density (abundance) of ABCG2 in different tissues. In this work we radiolabeled Ko143 with carbon-11 (11C) and performed small-animal PET experiments with [11C]Ko143 in wild-type, Abcg2(-/-), Abcb1a/b(-/-) and Abcb1a/b(-/-)Abcg2(-/-) mice to assess the influence of Abcg2 and Abcb1a/b on tissue distribution and excretion of [11C]Ko143. [11C]Ko143 was extensively metabolized in vivo and unidentified radiolabeled metabolites were found in all investigated tissues. We detected no significant differences between wild-type and Abcg2(-/-) mice in the distribution of [11C]Ko143-derived radioactivity to Abcg2-expressing organs (brain, liver and kidney). [11C]Ko143 and possibly its radiolabeled metabolites were transported by Abcb1a and not by Abcg2 at the mouse blood-brain barrier. [11C]Ko143-derived radioactivity underwent both hepatobiliary and urinary excretion, with Abcg2 playing a possible role in mediating the transport of radiolabeled metabolites of [11C]Ko143 from the kidney into urine. Experiments in which a pharmacologic dose of unlabeled Ko143 (10 mg/kg) was co-administered with [11C]Ko143 revealed pronounced effects of the vehicle used for Ko143 formulation (containing polyethylene glycol 300 and polysorbate 80) on radioactivity distribution to the brain and the liver, as well as on hepatobiliary and urinary excretion of radioactivity. Our results highlight the challenges associated with the development of PET tracers for ABC transporters and emphasize that inhibitory effects of pharmaceutical excipients on membrane transporters need to be considered when performing in vivo drug-drug interaction studies. Finally, our study illustrates the power of small-animal PET to assess the interaction of drug molecules with membrane transporters on a whole body level.
Collapse
Affiliation(s)
- Severin Mairinger
- Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Viktoria Zoufal
- Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Wanek
- Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Alexander Traxl
- Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Filip
- Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Michael Sauberer
- Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Johann Stanek
- Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Claudia Kuntner
- Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Jens Pahnke
- Department of Neuro-/Pathology, University of Oslo (UiO) and Oslo University Hospital (OUS), Oslo, Norway; LIED, University of Lübeck, Germany; Leibniz-Institute of Plant Biochemistry, Halle, Germany
| | - Markus Müller
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Biomedical Systems, Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; Division of Nuclear Medicine, Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
35
|
Dora CP, Kushwah V, Katiyar SS, Kumar P, Pillay V, Suresh S, Jain S. Improved oral bioavailability and therapeutic efficacy of erlotinib through molecular complexation with phospholipid. Int J Pharm 2017; 534:1-13. [PMID: 28970115 DOI: 10.1016/j.ijpharm.2017.09.071] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 08/25/2017] [Accepted: 09/29/2017] [Indexed: 12/18/2022]
Abstract
The current study was aimed to prepare a molecular complex of erlotinib (ERL) with phospholipid (PC) for enhancement of solubility and thus bioavailability, therapeutic efficacy and reducing the toxicity of erlotinib. Phospholipid complex of drug was prepared by solvent evaporation method and characterized by differential scanning calorimetry (DSC), Fourier transform infra-red spectroscopy (FT-IR), proton and phosphorus nuclear magnetic resonance spectroscopy (1H NMR and 31P NMR), powder X-ray diffraction (P-XRD), scanning electron microscopy (SEM) and transmission electron microscopy (TEM), which all explained the interactions of two components, validating the complexation phenomenon. In silico study also supported the phase change and molecular interactions for the establishment of ERL-PC. Spherical shaped nanostructures with 183.37±28.61nm size, -19.52±6.94mV potential and 28.59±2.66% loading efficiency were formed following dispersion of ERL-PC in aqueous media. In vitro release study revealed the higher release of ERL-PC due to amorphization and solubilization of drug. Caco-2 cell uptake resulted in ∼2 fold higher uptake of ERL-PC than free drug. In vitro cell culture studies were performed using human pancreatic adenocarcinoma cell lines, which demonstrated the higher cytotoxicity and apoptosis in case of ERL-PC. In vivo pharmacokinetics also supported the in vitro observations and showed ∼1.7 fold higher bioavailability with ERL-PC than ERL. Finally, in vivo efficacy and toxicity studies explained the superiority of ERL-PC over the free drug. Based on the results, phospholipid complex appears to be a promising tool to enhance bioavailability, efficacy, cytotoxicity and safety of erlotinib.
Collapse
Affiliation(s)
- Chander Parkash Dora
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Varun Kushwah
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Sameer S Katiyar
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Parktown, 2193, South Africa
| | - Viness Pillay
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, Parktown, 2193, South Africa
| | - Sarasija Suresh
- Institute for Drug Delivery and Biomedical Research (IDBR), Bangalore, Karnataka, 560068, India; RGV Research & Innovations Pvt. Ltd (RGVRI), Bangalore, Karnataka, 560010, India
| | - Sanyog Jain
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Punjab, 160062, India.
| |
Collapse
|
36
|
Verheijen RB, Yaqub M, Sawicki E, van Tellingen O, Lammertsma AA, Nuijen B, Schellens JHM, Beijnen JH, Huitema ADR, Hendrikse NH, Steeghs N. Molecular Imaging of ABCB1 and ABCG2 Inhibition at the Human Blood-Brain Barrier Using Elacridar and 11C-Erlotinib PET. J Nucl Med 2017; 59:973-979. [PMID: 29175983 DOI: 10.2967/jnumed.117.195800] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 11/10/2017] [Indexed: 12/12/2022] Open
Abstract
Transporters such as ABCB1 and ABCG2 limit the exposure of several anticancer drugs to the brain, leading to suboptimal treatment in the central nervous system. The purpose of this study was to investigate the effects of the ABCB1 and ABCG2 inhibitor elacridar on brain uptake using 11C-erlotinib PET. Methods: Elacridar and cold erlotinib were administered orally to wild-type (WT) and Abcb1a/b;Abcg2 knockout mice. In addition, brain uptake was measured using 11C-erlotinib imaging and ex vivo scintillation counting in knockout and WT mice. Six patients with advanced solid tumors underwent 11C-erlotinib PET scans before and after a 1,000-mg dose of elacridar. 11C-erlotinib brain uptake was quantified by pharmacokinetic modeling using volume of distribution (VT) as the outcome parameter. In addition, 15O-H2O scans to measure cerebral blood flow were acquired before each 11C-erlotinib scan. Results: Brain uptake of 11C-erlotinib was 2.6-fold higher in Abcb1a/b;Abcg2 knockout mice than in WT mice, measured as percentage injected dose per gram of tissue (P = 0.01). In WT mice, the addition of elacridar (at systemic plasma concentrations of ≥200 ng/mL) resulted in an increased brain concentration of erlotinib, without affecting erlotinib plasma concentration. In patients, the VT of 11C-erlotinib did not increase after intake of elacridar (0.213 ± 0.12 vs. 0.205 ± 0.07, P = 0.91). 15O-H2O PET showed no significant changes in cerebral blood flow. Elacridar exposure in patients was 401 ± 154 ng/mL. No increase in VT with increased elacridar plasma exposure was found over the 271-619 ng/mL range. Conclusion: When Abcb1 and Abcg2 were disrupted in mice, brain uptake of 11C-erlotinib increased both at a tracer dose and at a pharmacologic dose. In patients, brain uptake of 11C-erlotinib was not higher after administration of elacridar. The more pronounced role that ABCG2 appears to play at the human blood-brain barrier and the lower potency of elacridar to inhibit ABCG2 may be an explanation of these interspecies differences.
Collapse
Affiliation(s)
- Remy B Verheijen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Maqsood Yaqub
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Emilia Sawicki
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Olaf van Tellingen
- Department of Bio-Pharmacology/Mouse Cancer Clinic, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Adriaan A Lammertsma
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Bastiaan Nuijen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Jan H M Schellens
- Department of Clinical Pharmacology and Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy and Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, Utrecht University Medical Center, Utrecht, The Netherlands; and
| | - N Harry Hendrikse
- Department of Radiology and Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands.,Department of Clinical Pharmacology and Pharmacy, VU University Medical Center, Amsterdam, The Netherlands
| | - Neeltje Steeghs
- Department of Clinical Pharmacology and Medical Oncology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| |
Collapse
|
37
|
Bauer M, Matsuda A, Wulkersdorfer B, Philippe C, Traxl A, Özvegy-Laczka C, Stanek J, Nics L, Klebermass EM, Poschner S, Jäger W, Patik I, Bakos É, Szakács G, Wadsak W, Hacker M, Zeitlinger M, Langer O. Influence of OATPs on Hepatic Disposition of Erlotinib Measured With Positron Emission Tomography. Clin Pharmacol Ther 2017; 104:139-147. [PMID: 28940241 PMCID: PMC6083370 DOI: 10.1002/cpt.888] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 08/09/2017] [Accepted: 09/18/2017] [Indexed: 12/19/2022]
Abstract
To assess the hepatic disposition of erlotinib, we performed positron emission tomography (PET) scans with [11 C]erlotinib in healthy volunteers without and with oral pretreatment with a therapeutic erlotinib dose (300 mg). Erlotinib pretreatment significantly decreased the liver exposure to [11 C]erlotinib with a concomitant increase in blood exposure, pointing to the involvement of a carrier-mediated hepatic uptake mechanism. Using cell lines overexpressing human organic anion-transporting polypeptides (OATPs) 1B1, 1B3, or 2B1, we show that [11 C]erlotinib is selectively transported by OATP2B1. Our data suggest that at PET microdoses hepatic uptake of [11 C]erlotinib is mediated by OATP2B1, whereas at therapeutic doses OATP2B1 transport is saturated and hepatic uptake occurs mainly by passive diffusion. We propose that [11 C]erlotinib may be used as a hepatic OATP2B1 probe substrate and erlotinib as an OATP2B1 inhibitor in clinical drug-drug interaction studies, allowing the contribution of OATP2B1 to the hepatic uptake of drugs to be revealed.
Collapse
Affiliation(s)
- Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Akihiro Matsuda
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | | | - Cécile Philippe
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Alexander Traxl
- Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Csilla Özvegy-Laczka
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Johann Stanek
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Lukas Nics
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Eva-Maria Klebermass
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Stefan Poschner
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Walter Jäger
- Department of Clinical Pharmacy and Diagnostics, University of Vienna, Vienna, Austria
| | - Izabel Patik
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Éva Bakos
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary
| | - Gergely Szakács
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Budapest, Hungary.,Institute of Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Wadsak
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria.,Center for Biomarker Research in Medicine, CBmed GmbH, Graz, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Department of Biomedical Imaging und Image-guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Vienna, Austria.,Center for Health & Bioresources, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| |
Collapse
|
38
|
Tomono T, Yano K, Ogihara T. Snail-Induced Epithelial-to-Mesenchymal Transition Enhances P-gp-Mediated Multidrug Resistance in HCC827 Cells. J Pharm Sci 2017; 106:2642-2649. [DOI: 10.1016/j.xphs.2017.03.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 03/03/2017] [Accepted: 03/09/2017] [Indexed: 12/12/2022]
|
39
|
Langer O. Use of PET Imaging to Evaluate Transporter-Mediated Drug-Drug Interactions. J Clin Pharmacol 2017; 56 Suppl 7:S143-56. [PMID: 27385172 DOI: 10.1002/jcph.722] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 02/03/2016] [Accepted: 02/11/2016] [Indexed: 12/25/2022]
Abstract
Several membrane transporters belonging to the adenosine triphosphate-binding cassette (ABC) and solute carrier (SLC) families can transport drugs and drug metabolites and thereby exert an effect on drug absorption, distribution, and excretion, which may potentially lead to transporter-mediated drug-drug interactions (DDIs). Some transporter-mediated DDIs may lead to changes in organ distribution of drugs (eg, brain, liver, kidneys) without affecting plasma concentrations. Positron emission tomography (PET) is a noninvasive imaging method that allows studying of the distribution of radiolabeled drugs to different organs and tissues and is therefore the method of choice to quantitatively assess transporter-mediated DDIs on a tissue level. There are 2 approaches to how PET can be used in transporter-mediated DDI studies. When the drug of interest is a potential perpetrator of DDIs, it may be administered in unlabeled form to assess its influence on tissue distribution of a generic transporter-specific PET tracer (probe substrate). When the drug of interest is a potential victim of DDIs, it may be radiolabeled with carbon-11 or fluorine-18 and used in combination with a prototypical transporter inhibitor (eg, rifampicin). PET has already been used both in preclinical species and in humans to assess the effects of transporter-mediated DDIs on drug disposition in different organ systems, such as brain, liver, and kidneys, for which examples are given in the present review article. Given the growing importance of membrane transporters with respect to drug safety and efficacy, PET is expected to play an increasingly important role in future drug development.
Collapse
Affiliation(s)
- Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria.,Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria.,Medical Imaging Cluster, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
40
|
Traxl A, Komposch K, Glitzner E, Wanek T, Mairinger S, Langer O, Sibilia M. Hepatocyte-Specific Deletion of EGFR in Mice Reduces Hepatic Abcg2 Transport Activity Measured by [ 11C]erlotinib and Positron Emission Tomography. Drug Metab Dispos 2017; 45:1093-1100. [PMID: 28790147 DOI: 10.1124/dmd.117.077081] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/04/2017] [Indexed: 01/03/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) regulates cellular expression levels of breast cancer resistance protein (humans: ABCG2, rodents: Abcg2) via its downstream signaling pathways. Drugs that inhibit EGFR signaling (e.g., tyrosine kinase inhibitors, antibodies) may lead to ABCG2-mediated drug-drug interactions (DDIs) by changing the disposition of concomitantly administered ABCG2 substrate drugs. In this study, we used positron emission tomography and magnetic resonance imaging to compare disposition of the model Abcg2 substrate [11C]erlotinib in a mouse model of hepatocyte-specific deletion of EGFR (EGFR∆hep mice, n = 5) with EGFRfl/fl control mice (n = 6), which have normal EGFR expression levels in all tissues. Integration plot analysis was used to estimate the rate constants for transfer of radioactivity from the liver into bile (kbile) and from the kidney into urine (kurine). EGFR∆hep mice showed significantly lower radioactivity concentrations in the intestine (1.6-fold) and higher radioactivity concentrations in the urinary bladder (3.2-fold) compared with EGFRfl/fl mice. Kbile was significantly decreased (3.0-fold) in EGFR∆hep mice, whereas kurine was by 2.2-fold increased. Western blot analysis of liver tissue confirmed deletion of EGFR and showed significant decreases in Abcg2 and increases in P-glycoprotein (Abcb1a/b) expression levels in EGFR∆hep versus EGFRfl/fl mice. Our data show that EGFR deletion in hepatocytes leads to a reduction in Abcg2-mediated hepatobiliary clearance of a probe substrate accompanied by a shift to renal excretion of the drug, which raises the possibility that EGFR-inhibiting drugs may cause ABCG2-mediated DDIs.
Collapse
Affiliation(s)
- Alexander Traxl
- Center for Health and Bioresources, Biomedical Systems, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (A.T., T.W., S.M., O.L.); Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center (K.K., E.G., M.S.); Department of Clinical Pharmacology (O.L.) and Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine (O.L.), Medical University of Vienna, Vienna, Austria
| | - Karin Komposch
- Center for Health and Bioresources, Biomedical Systems, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (A.T., T.W., S.M., O.L.); Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center (K.K., E.G., M.S.); Department of Clinical Pharmacology (O.L.) and Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine (O.L.), Medical University of Vienna, Vienna, Austria
| | - Elisabeth Glitzner
- Center for Health and Bioresources, Biomedical Systems, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (A.T., T.W., S.M., O.L.); Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center (K.K., E.G., M.S.); Department of Clinical Pharmacology (O.L.) and Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine (O.L.), Medical University of Vienna, Vienna, Austria
| | - Thomas Wanek
- Center for Health and Bioresources, Biomedical Systems, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (A.T., T.W., S.M., O.L.); Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center (K.K., E.G., M.S.); Department of Clinical Pharmacology (O.L.) and Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine (O.L.), Medical University of Vienna, Vienna, Austria
| | - Severin Mairinger
- Center for Health and Bioresources, Biomedical Systems, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (A.T., T.W., S.M., O.L.); Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center (K.K., E.G., M.S.); Department of Clinical Pharmacology (O.L.) and Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine (O.L.), Medical University of Vienna, Vienna, Austria
| | - Oliver Langer
- Center for Health and Bioresources, Biomedical Systems, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (A.T., T.W., S.M., O.L.); Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center (K.K., E.G., M.S.); Department of Clinical Pharmacology (O.L.) and Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine (O.L.), Medical University of Vienna, Vienna, Austria
| | - Maria Sibilia
- Center for Health and Bioresources, Biomedical Systems, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (A.T., T.W., S.M., O.L.); Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center (K.K., E.G., M.S.); Department of Clinical Pharmacology (O.L.) and Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine (O.L.), Medical University of Vienna, Vienna, Austria
| |
Collapse
|
41
|
Bernard-Gauthier V, Mahringer A, Vesnaver M, Fricker G, Schirrmacher R. Design and synthesis of a fluorinated quinazoline-based type-II Trk inhibitor as a scaffold for PET radiotracer development. Bioorg Med Chem Lett 2017; 27:2771-2775. [DOI: 10.1016/j.bmcl.2017.04.064] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 04/18/2017] [Accepted: 04/19/2017] [Indexed: 12/20/2022]
|
42
|
Auvity S, Chapy H, Goutal S, Caillé F, Hosten B, Smirnova M, Declèves X, Tournier N, Cisternino S. Diphenhydramine as a selective probe to study H +-antiporter function at the blood-brain barrier: Application to [ 11C]diphenhydramine positron emission tomography imaging. J Cereb Blood Flow Metab 2017; 37:2185-2195. [PMID: 27488910 PMCID: PMC5464711 DOI: 10.1177/0271678x16662042] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diphenhydramine, a sedative histamine H1-receptor (H1R) antagonist, was evaluated as a probe to measure drug/H+-antiporter function at the blood-brain barrier. In situ brain perfusion experiments in mice and rats showed that diphenhydramine transport at the blood-brain barrier was saturable, following Michaelis-Menten kinetics with a Km = 2.99 mM and Vmax = 179.5 nmol s-1 g-1. In the pharmacological plasma concentration range the carrier-mediated component accounted for 77% of diphenhydramine influx while passive diffusion accounted for only 23%. [14C]Diphenhydramine blood-brain barrier transport was proton and clonidine sensitive but was influenced by neither tetraethylammonium, a MATE1 (SLC47A1), and OCT/OCTN (SLC22A1-5) modulator, nor P-gp/Bcrp (ABCB1a/1b/ABCG2) deficiency. Brain and plasma kinetics of [11C]diphenhydramine were measured by positron emission tomography imaging in rats. [11C]Diphenhydramine kinetics in different brain regions were not influenced by displacement with 1 mg kg-1 unlabeled diphenhydramine, indicating the specificity of the brain positron emission tomography signal for blood-brain barrier transport activity over binding to any central nervous system target in vivo. [11C]Diphenhydramine radiometabolites were not detected in the brain 15 min after injection, allowing for the reliable calculation of [11C]diphenhydramine brain uptake clearance (Clup = 0.99 ± 0.18 mL min-1 cm-3). Diphenhydramine is a selective and specific H+-antiporter substrate. [11C]Diphenhydramine positron emission tomography imaging offers a reliable and noninvasive method to evaluate H+-antiporter function at the blood-brain barrier.
Collapse
Affiliation(s)
- Sylvain Auvity
- 1 Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France.,2 Variabilité de réponse aux psychotropes, INSERM, U1144, Paris, France; Université Paris Descartes, Faculté de pharmacie, UMR-S 1144, Paris, F-75006, France. Université Paris Diderot, UMR-S 1144, Paris, F-75013, France
| | - Hélène Chapy
- 2 Variabilité de réponse aux psychotropes, INSERM, U1144, Paris, France; Université Paris Descartes, Faculté de pharmacie, UMR-S 1144, Paris, F-75006, France. Université Paris Diderot, UMR-S 1144, Paris, F-75013, France
| | - Sébastien Goutal
- 1 Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Fabien Caillé
- 1 Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Benoit Hosten
- 2 Variabilité de réponse aux psychotropes, INSERM, U1144, Paris, France; Université Paris Descartes, Faculté de pharmacie, UMR-S 1144, Paris, F-75006, France. Université Paris Diderot, UMR-S 1144, Paris, F-75013, France
| | - Maria Smirnova
- 2 Variabilité de réponse aux psychotropes, INSERM, U1144, Paris, France; Université Paris Descartes, Faculté de pharmacie, UMR-S 1144, Paris, F-75006, France. Université Paris Diderot, UMR-S 1144, Paris, F-75013, France
| | - Xavier Declèves
- 2 Variabilité de réponse aux psychotropes, INSERM, U1144, Paris, France; Université Paris Descartes, Faculté de pharmacie, UMR-S 1144, Paris, F-75006, France. Université Paris Diderot, UMR-S 1144, Paris, F-75013, France
| | - Nicolas Tournier
- 1 Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Salvatore Cisternino
- 1 Imagerie Moléculaire In Vivo, IMIV, CEA, Inserm, CNRS, Univ. Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France.,2 Variabilité de réponse aux psychotropes, INSERM, U1144, Paris, France; Université Paris Descartes, Faculté de pharmacie, UMR-S 1144, Paris, F-75006, France. Université Paris Diderot, UMR-S 1144, Paris, F-75013, France
| |
Collapse
|
43
|
[ 11C]Erlotinib PET cannot detect acquired erlotinib resistance in NSCLC tumor xenografts in mice. Nucl Med Biol 2017; 52:7-15. [PMID: 28575795 DOI: 10.1016/j.nucmedbio.2017.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/02/2017] [Accepted: 05/10/2017] [Indexed: 12/24/2022]
Abstract
INTRODUCTION [11C]Erlotinib PET has shown promise to distinguish non-small cell lung cancer (NSCLC) tumors harboring the activating epidermal growth factor receptor (EGFR) mutation delE746-A750 from tumors with wild-type EGFR. To assess the suitability of [11C]erlotinib PET to detect the emergence of acquired erlotinib resistance in initially erlotinib-responsive tumors, we performed in vitro binding and PET experiments in mice bearing tumor xenografts using a range of different cancer cells, which were erlotinib-sensitive or exhibited clinically relevant resistance mechanisms to erlotinib. METHODS The following cell lines were used for in vitro binding and PET experiments: the epidermoid carcinoma cell line A-431 (erlotinib-sensitive, wild-type EGFR) and the three NSCLC cell lines HCC827 (erlotinib-sensitive, delE746-A750), HCC827EPR (erlotinib-resistant, delE746-A750 and T790M) and HCC827ERLO (erlotinib-resistant, delE746-A750 and MET amplification). BALB/c nude mice with subcutaneous tumor xenografts underwent two consecutive [11C]erlotinib PET scans, a baseline scan and a second scan in which unlabeled erlotinib (10mg/kg) was co-injected. Logan graphical analysis was used to estimate total distribution volume (VT) of [11C]erlotinib in tumors. RESULTS In vitro experiments revealed significantly higher uptake of [11C]erlotinib (5.2-fold) in the three NSCLC cell lines as compared to A-431 cells. In all four cell lines co-incubation with unlabeled erlotinib (1μM) led to significant reductions in [11C]erlotinib uptake (-19% to -66%). In both PET scans and for all four studied cell lines there were no significant differences in tumoral [11C]erlotinib VT values. For all three NSCLC cell lines, but not for the A-431 cell line, tumoral VT was significantly reduced following co-injection of unlabeled erlotinib (-20% to -35%). CONCLUSIONS We found no significant differences in the in vitro and in vivo binding of [11C]erlotinib between erlotinib-sensitive and erlotinib-resistant NSCLC cells. Our findings suggest that [11C]erlotinib PET will not be suitable to distinguish erlotinib-sensitive NSCLC tumors from tumors with acquired resistance to erlotinib.
Collapse
|
44
|
Matsuda A, Karch R, Bauer M, Traxl A, Zeitlinger M, Langer O. A Prediction Method for P-glycoprotein-Mediated Drug-Drug Interactions at the Human Blood-Brain Barrier From Blood Concentration-Time Profiles, Validated With PET Data. J Pharm Sci 2017; 106:2780-2786. [PMID: 28385544 DOI: 10.1016/j.xphs.2017.03.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 03/22/2017] [Accepted: 03/27/2017] [Indexed: 12/11/2022]
Abstract
The purpose of this study was to establish physiologically based pharmacokinetic models to predict in humans the brain concentration-time profiles and P-glycoprotein (Pgp)-mediated brain drug-drug interactions between the model Pgp substrate (R)-[11C]verapamil (VPM), the model dual Pgp/breast cancer resistance protein (BCRP) substrate [11C]tariquidar (TQD), and the Pgp inhibitor tariquidar. The model predictions were validated with results from positron emission tomography studies in humans. Using these physiologically based pharmacokinetic models, the differences between predicted and observed areas under the concentration-time curves (AUC) of VPM and TQD in the brain were within a 1.2-fold and 2.5-fold range, respectively. Also, brain AUC increases of VPM and TQD after Pgp inhibitor administration were predicted with 2.5-fold accuracy when in vitro inhibition constant or half-maximum inhibitory concentration values of tariquidar were used. The predicted rank order of the magnitude of AUC increases reflected the results of the clinical positron emission tomography studies. Our results suggest that the established models can predict brain exposure from the respective blood concentration-time profiles and rank the magnitude of the Pgp-mediated brain drug-drug interaction potential for both Pgp and Pgp/BCRP substrates in humans.
Collapse
Affiliation(s)
- Akihiro Matsuda
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Rudolf Karch
- Center for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, A-1090 Vienna, Austria
| | - Martin Bauer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Alexander Traxl
- Center for Health and Bioresources, AIT Austrian Institute of Technology GmbH, A-2444 Seibersdorf, Austria
| | - Markus Zeitlinger
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria
| | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, A-1090 Vienna, Austria; Center for Health and Bioresources, AIT Austrian Institute of Technology GmbH, A-2444 Seibersdorf, Austria; Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, A-1090 Vienna, Austria.
| |
Collapse
|
45
|
Zhu L, Wu J, Zhao M, Song W, Qi X, Wang Y, Lu L, Liu Z. Mdr1a plays a crucial role in regulating the analgesic effect and toxicity of aconitine by altering its pharmacokinetic characteristics. Toxicol Appl Pharmacol 2017; 320:32-39. [PMID: 28193520 DOI: 10.1016/j.taap.2017.02.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 02/06/2017] [Accepted: 02/10/2017] [Indexed: 02/06/2023]
Abstract
Aconitine (AC) is the primary bioactive/toxic alkaloid in plants of the Aconitum species. Our previous study demonstrated that Mdr1 was involved in efflux of AC. However, the mechanism by which Mdr1 regulates the efficacy/toxicity of AC in vivo remains unclear. The present study aimed to determine the effects of Mdr1a on the efficacy/toxicity and pharmacokinetics of AC in wild-type and Mdr1a-/- FVB mice. After oral administration of AC, significantly higher analgesic effect was observed in Mdr1a-/- mice (49% to 105%) compared to wild-type mice (P<0.05). The levels of s100-β protein and creatine kinase, which indicate cerebral and myocardial damage, respectively, were also significantly increased (P<0.05) in Mdr1a-/- mice. Histopathological examination revealed that the Mdr1a-/- mice suffered from evident cerebral and myocardial damages, but the wild-type mice did not. These findings suggested that Mdr1a deficiency significantly promoted the analgesic effect of AC and exacerbated its toxicity. Pharmacokinetic experiments showed that T1/2 of AC in the Mdr1a-/- mice was significantly higher (from 87% to 300%) than that in wild-type mice (P<0.05). The distribution of AC in the brain of Mdr1a-/- mice was 2- to 32-fold higher than that in the brains of wild-type mice (P<0.05). Toxic reactions were more severe in Mdr1a-/- mice compared to wild-type mice. In conclusion, Mdr1a deficiency significantly enhanced the analgesic effect of AC and exacerbated its toxicity by upregulating its distribution to the brain and decreasing its plasma elimination rate. Thus, Mdr1a dysfunction may cause severe AC poisoning.
Collapse
Affiliation(s)
- Lijun Zhu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China
| | - Jinjun Wu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China
| | - Min Zhao
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China
| | - Wenjie Song
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China
| | - Xiaoxiao Qi
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China
| | - Ying Wang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China
| | - Linlin Lu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, PR China
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510006, PR China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR) 999078, PR China.
| |
Collapse
|
46
|
Tournier N, Goutal S, Auvity S, Traxl A, Mairinger S, Wanek T, Helal OB, Buvat I, Soussan M, Caillé F, Langer O. Strategies to Inhibit ABCB1- and ABCG2-Mediated Efflux Transport of Erlotinib at the Blood-Brain Barrier: A PET Study on Nonhuman Primates. J Nucl Med 2016; 58:117-122. [PMID: 27493269 DOI: 10.2967/jnumed.116.178665] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/12/2016] [Indexed: 12/12/2022] Open
Abstract
The tyrosine kinase inhibitor erlotinib poorly penetrates the blood-brain barrier (BBB) because of efflux transport by P-glycoprotein (ABCB1) and breast cancer resistance protein (ABCG2), thereby limiting its utility in the treatment of non-small cell lung cancer metastases in the brain. Pharmacologic strategies to inhibit ABCB1/ABCG2-mediated efflux transport at the BBB have been successfully developed in rodents, but it remains unclear whether these can be translated to humans given the pronounced species differences in ABCG2/ABCB1 expression ratios at the BBB. We assessed the efficacy of two different ABCB1/ABCG2 inhibitors to enhance brain distribution of 11C-erlotinib in nonhuman primates as a model of the human BBB. METHODS Papio anubis baboons underwent PET scans of the brain after intravenous injection of 11C-erlotinib under baseline conditions (n = 4) and during intravenous infusion of high-dose erlotinib (10 mg/kg/h, n = 4) or elacridar (12 mg/kg/h, n = 3). RESULTS Under baseline conditions, 11C-erlotinib distribution to the brain (total volume of distribution [VT], 0.22 ± 0.015 mL/cm3) was markedly lower than its distribution to muscle tissue surrounding the skull (VT, 0.86 ± 0.10 mL/cm3). Elacridar infusion resulted in a 3.5 ± 0.9-fold increase in 11C-erlotinib distribution to the brain (VT, 0.81 ± 0.21 mL/cm3, P < 0.01), reaching levels comparable to those in muscle tissue, without changing 11C-erlotinib plasma pharmacokinetics. During high-dose erlotinib infusion, 11C-erlotinib brain distribution was also significantly (1.7 ± 0.2-fold) increased (VT, 0.38 ± 0.033 mL/cm3, P < 0.05), with a concomitant increase in 11C-erlotinib plasma exposure. CONCLUSION We successfully implemented ABCB1/ABCG2 inhibition protocols in nonhuman primates resulting in pronounced increases in brain distribution of 11C-erlotinib. For patients with brain tumors, such inhibition protocols may ultimately be applied to create more effective treatments using drugs that undergo efflux transport at the BBB.
Collapse
Affiliation(s)
- Nicolas Tournier
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Sebastien Goutal
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Sylvain Auvity
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Alexander Traxl
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Severin Mairinger
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Thomas Wanek
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Ourkia-Badia Helal
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Irène Buvat
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Michael Soussan
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Fabien Caillé
- Imagerie Moléculaire In Vivo, IMIV, CEA, INSERM, CNRS, Université Paris-Sud, Université Paris Saclay, CEA-SHFJ, Orsay, France
| | - Oliver Langer
- Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria.,Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria; and.,Division of Nuclear Medicine, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
47
|
Wanek T, Halilbasic E, Visentin M, Mairinger S, Römermann K, Stieger B, Kuntner C, Müller M, Langer O, Trauner M. Influence of 24-Nor-Ursodeoxycholic Acid on Hepatic Disposition of [(18)F]Ciprofloxacin, a Positron Emission Tomography Study in Mice. J Pharm Sci 2016; 105:106-12. [PMID: 26852845 DOI: 10.1016/j.xphs.2015.11.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2015] [Revised: 10/28/2015] [Accepted: 11/05/2015] [Indexed: 01/10/2023]
Abstract
24-nor-ursodeoxycholic acid (norUDCA) is a novel therapeutic approach to cholestatic liver diseases. In mouse models of cholestasis, norUDCA induces basolateral multidrug resistance-associated proteins 4 (Mrp4) and 3 in hepatocytes, which provide alternative escape routes for bile acids accumulating during cholestasis but could also result in altered hepatic disposition of concomitantly administered substrate drugs. We used positron emission tomography imaging to study the influence of norUDCA on hepatic disposition of the model Mrp4 substrate [(18)F]ciprofloxacin in wild-type and Mdr2((-/-)) mice, a model of cholestasis. Animals underwent [(18)F]ciprofloxacin positron emission tomography at baseline and after norUDCA treatment. After norUDCA treatment, liver-to-blood area under the curve ratio of [(18)F]ciprofloxacin was significantly decreased compared to baseline, both in wild-type (-34.0 ± 2.1%) and Mdr2((-/-)) mice (-20.5 ± 6.0%). [(18)F]Ciprofloxacin uptake clearance from blood into liver was reduced by -17.1 ± 9.0% in wild-type and by -20.1 ± 7.3% in Mdr2((-/-)) mice. Real-time PCR analysis showed significant increases in hepatic Mrp4 and multidrug resistance-associated protein 3 mRNA after norUDCA. Transport experiments in organic anion transporting polypeptide (OATP)1B1-, OATP1B3-, and OATP2B1-transfected cells revealed weak transport of [(14)C]ciprofloxacin by OATP1B3 and OATP2B1 and no inhibition by norUDCA. In conclusion, our data suggest that changes in hepatic [(18)F]ciprofloxacin disposition in mice after norUDCA treatment were caused by induction of basolateral Mrp4 in hepatocytes.
Collapse
Affiliation(s)
- Thomas Wanek
- Biomedical Systems, Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Emina Halilbasic
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Michele Visentin
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Severin Mairinger
- Biomedical Systems, Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Kerstin Römermann
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine, Center for Systems Neuroscience, Hannover, Germany
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland
| | - Claudia Kuntner
- Biomedical Systems, Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria
| | - Markus Müller
- Department of Clinical Pharmacology, Medical University of Vienna, Austria
| | - Oliver Langer
- Biomedical Systems, Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria; Department of Clinical Pharmacology, Medical University of Vienna, Austria.
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
48
|
Recent Advances in the Development and Application of Radiolabeled Kinase Inhibitors for PET Imaging. Molecules 2015; 20:22000-27. [PMID: 26690113 PMCID: PMC6332294 DOI: 10.3390/molecules201219816] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/18/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022] Open
Abstract
Over the last 20 years, intensive investigation and multiple clinical successes targeting protein kinases, mostly for cancer treatment, have identified small molecule kinase inhibitors as a prominent therapeutic class. In the course of those investigations, radiolabeled kinase inhibitors for positron emission tomography (PET) imaging have been synthesized and evaluated as diagnostic imaging probes for cancer characterization. Given that inhibitor coverage of the kinome is continuously expanding, in vivo PET imaging will likely find increasing applications for therapy monitoring and receptor density studies both in- and outside of oncological conditions. Early investigated radiolabeled inhibitors, which are mostly based on clinically approved tyrosine kinase inhibitor (TKI) isotopologues, have now entered clinical trials. Novel radioligands for cancer and PET neuroimaging originating from novel but relevant target kinases are currently being explored in preclinical studies. This article reviews the literature involving radiotracer design, radiochemistry approaches, biological tracer evaluation and nuclear imaging results of radiolabeled kinase inhibitors for PET reported between 2010 and mid-2015. Aspects regarding the usefulness of pursuing selective vs. promiscuous inhibitor scaffolds and the inherent challenges associated with intracellular enzyme imaging will be discussed.
Collapse
|
49
|
Pottier G, Marie S, Goutal S, Auvity S, Peyronneau MA, Stute S, Boisgard R, Dollé F, Buvat I, Caillé F, Tournier N. Imaging the Impact of the P-Glycoprotein (ABCB1) Function on the Brain Kinetics of Metoclopramide. J Nucl Med 2015; 57:309-14. [PMID: 26585058 DOI: 10.2967/jnumed.115.164350] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Accepted: 11/02/2015] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED The effects of metoclopramide on the central nervous system (CNS) in patients suggest substantial brain distribution. Previous data suggest that metoclopramide brain kinetics may nonetheless be controlled by ATP-binding cassette (ABC) transporters expressed at the blood-brain barrier. We used (11)C-metoclopramide PET imaging to elucidate the kinetic impact of transporter function on metoclopramide exposure to the brain. METHODS (11)C-metoclopramide transport by P-glycoprotein (P-gp; ABCB1) and the breast cancer resistance protein (BCRP; ABCG2) was tested using uptake assays in cells overexpressing P-gp and BCRP. (11)C-metoclopramide brain kinetics were compared using PET in rats (n = 4-5) in the absence and presence of a pharmacologic dose of metoclopramide (3 mg/kg), with or without P-gp inhibition using intravenous tariquidar (8 mg/kg). The (11)C-metoclopramide brain distribution (VT based on Logan plot analysis) and brain kinetics (2-tissue-compartment model) were characterized with either a measured or an imaged-derived input function. Plasma and brain radiometabolites were studied using radio-high-performance liquid chromatography analysis. RESULTS (11)C-metoclopramide transport was selective for P-gp over BCRP. Pharmacologic dose did not affect baseline (11)C-metoclopramide brain kinetics (VT = 2.28 ± 0.32 and 2.04 ± 0.19 mL⋅cm(-3) using microdose and pharmacologic dose, respectively). Tariquidar significantly enhanced microdose (11)C-metoclopramide VT (7.80 ± 1.43 mL⋅cm(-3)) with a 4.4-fold increase in K1 (influx rate constant) and a 2.3-fold increase in binding potential (k3/k4) in the 2-tissue-compartment model. In the pharmacologic situation, P-gp inhibition significantly increased metoclopramide brain distribution (VT = 6.28 ± 0.48 mL⋅cm(-3)) with a 2.0-fold increase in K1 and a 2.2-fold decrease in k2 (efflux rate), with no significant impact on binding potential. In this situation, only parent (11)C-metoclopramide could be detected in the brains of P-gp-inhibited rats. CONCLUSION (11)C-metoclopramide benefits from favorable pharmacokinetic properties that offer reliable quantification of P-gp function at the blood-brain barrier in a pharmacologic situation. Using metoclopramide as a model of CNS drug, we demonstrated that P-gp function not only reduces influx but also mediates the efflux from the brain back to the blood compartment, with additional impact on brain distribution. This PET-based strategy of P-gp function investigation may provide new insight on the contribution of P-gp to the variability of response to CNS drugs between patients.
Collapse
Affiliation(s)
- Géraldine Pottier
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Solène Marie
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Sébastien Goutal
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Sylvain Auvity
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Marie-Anne Peyronneau
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Simon Stute
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Raphaël Boisgard
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Frédéric Dollé
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Irène Buvat
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Fabien Caillé
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| | - Nicolas Tournier
- Inserm/CEA/Université Paris Sud, UMR 1023 - ERL 9218 CNRS, IMIV, Orsay, France; and CEA, DSV, I2BM, Service Hospitalier Frédéric Joliot, Orsay, France
| |
Collapse
|