1
|
Sabet Z, Vagiannis D, Budagaga Y, Zhang Y, Novotná E, Hanke I, Rozkoš T, Hofman J. Talazoparib Does Not Interact with ABCB1 Transporter or Cytochrome P450s, but Modulates Multidrug Resistance Mediated by ABCC1 and ABCG2: An in Vitro and Ex Vivo Study. Int J Mol Sci 2022; 23:ijms232214338. [PMID: 36430819 PMCID: PMC9697930 DOI: 10.3390/ijms232214338] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/02/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022] Open
Abstract
Talazoparib (Talzenna) is a novel poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitor that is clinically used for the therapy of breast cancer. Furthermore, the drug has shown antitumor activity against different cancer types, including non-small cell lung cancer (NSCLC). In this work, we investigated the possible inhibitory interactions of talazoparib toward selected ATP-binding cassette (ABC) drug efflux transporters and cytochrome P450 biotransformation enzymes (CYPs) and evaluated its position in multidrug resistance (MDR). In accumulation studies, talazoparib interacted with the ABCC1 and ABCG2 transporters, but there were no significant effects on ABCB1. Furthermore, incubation assays revealed a negligible capacity of the tested drug to inhibit clinically relevant CYPs. In in vitro drug combination experiments, talazoparib synergistically reversed daunorubicin and mitoxantrone resistance in cells with ABCC1 and ABCG2 expression, respectively. Importantly, the position of an effective MDR modulator was further confirmed in drug combinations performed in ex vivo NSCLC patients-derived explants, whereas the possible victim role was refuted in comparative proliferation experiments. In addition, talazoparib had no significant effects on the mRNA-level expressions of MDR-related ABC transporters in the MCF-7 cellular model. In summary, our study presents a comprehensive overview on the pharmacokinetic drug-drug interactions (DDI) profile of talazoparib. Moreover, we introduced talazoparib as an efficient MDR antagonist.
Collapse
Affiliation(s)
- Ziba Sabet
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Dimitrios Vagiannis
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Youssif Budagaga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Yu Zhang
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Eva Novotná
- Department of Biochemical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
| | - Ivo Hanke
- Department of Cardiac Surgery, Faculty of Medicine, Charles University in Hradec Králové and University Hospital Hradec Králové, Sokolská 581, 500 05 Hradec Králové, Czech Republic
| | - Tomáš Rozkoš
- The Fingerland Department of Pathology, Charles University, Faculty of Medicine and University Hospital in Hradec Králové, Czech Republic, Sokolská 581, 500 05 Hradec Králové, Czech Republic
| | - Jakub Hofman
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Králové, Charles University, Heyrovskeho 1203, 500 05 Hradec Králové, Czech Republic
- Correspondence: ; Tel.: +420-495-067-593
| |
Collapse
|
2
|
Oh JH, Power EA, Zhang W, Daniels DJ, Elmquist WF. Murine Central Nervous System and Bone Marrow Distribution of the Aurora A Kinase Inhibitor Alisertib: Pharmacokinetics and Exposure at the Sites of Efficacy and Toxicity. J Pharmacol Exp Ther 2022; 383:44-55. [PMID: 36279392 PMCID: PMC9513880 DOI: 10.1124/jpet.122.001268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022] Open
Abstract
Important challenges in developing drugs that target central nervous system (CNS) tumors include overcoming barriers for CNS delivery and reducing systemic side effects. Alisertib, an aurora A kinase inhibitor, has been examined for treatment of several CNS tumors in preclinical and clinical studies. In this study, we investigated the distribution of alisertib into the CNS, the site of efficacy for brain tumors, and into the bone marrow, the site of dose-limiting toxicity leading to myelosuppression. Mechanisms influencing site-specific distribution, such as active transport mediated by the efflux proteins, p-glycoprotein (P-gp) and breast cancer resistance protein (Bcrp), were examined. Alisertib exposure to the brain in wild-type mice was less than 1% of that in the plasma, and was evenly distributed throughout various brain regions and the spinal cord. Studies using transporter knockout mice and pharmacological inhibition show that alisertib CNS distribution is influenced by P-gp, but not Bcrp. Conversely, upon systemic administration, alisertib distribution to the bone marrow occurred rapidly, was not significantly limited by efflux transporters, and reached higher concentrations than in the CNS. This study demonstrates that, given an equivalent distributional driving force exposure in plasma, the exposure of alisertib in the brain is significantly less than that in the bone marrow, suggesting that targeted delivery may be necessary to guarantee therapeutic efficacy with minimal risk for adverse events.Therefore, these data suggest that, to improve the therapeutic index when using alisertib for brain tumors, a localized regional delivery, such as convection-enhanced delivery, may be warranted. SIGNIFICANCE STATEMENT: The CNS penetration of alisertib is limited with uniform distribution in various regions of the brain, and P-gp efflux is an important mechanism limiting that CNS distribution. Alisertib rapidly distributes into the bone marrow, a site of toxicity, with a greater exposure than in the CNS, a possible site of efficacy. These results suggest a need to design localized delivery strategies to improve the CNS exposure of alisertib and limit systemic toxicities in the treatment of brain tumors.
Collapse
Affiliation(s)
- Ju-Hee Oh
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (J-H.O., W.Z., W.F.E.); Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota (E.A.P., D.J.D.); and Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota (E.A.P.)
| | - Erica A Power
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (J-H.O., W.Z., W.F.E.); Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota (E.A.P., D.J.D.); and Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota (E.A.P.)
| | - Wenjuan Zhang
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (J-H.O., W.Z., W.F.E.); Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota (E.A.P., D.J.D.); and Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota (E.A.P.)
| | - David J Daniels
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (J-H.O., W.Z., W.F.E.); Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota (E.A.P., D.J.D.); and Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota (E.A.P.)
| | - William F Elmquist
- Brain Barriers Research Center, Department of Pharmaceutics, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota (J-H.O., W.Z., W.F.E.); Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota (E.A.P., D.J.D.); and Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota (E.A.P.)
| |
Collapse
|
3
|
Alisertib shows negligible potential for perpetrating pharmacokinetic drug-drug interactions on ABCB1, ABCG2 and cytochromes P450, but acts as dual-activity resistance modulator through the inhibition of ABCC1 transporter. Toxicol Appl Pharmacol 2022; 434:115823. [PMID: 34896433 DOI: 10.1016/j.taap.2021.115823] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/01/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022]
Abstract
Alisertib (MLN8237), a novel Aurora A kinase inhibitor, is currently being clinically tested in late-phase trials for the therapy of various malignancies. In the present work, we describe alisertib's potential to perpetrate pharmacokinetic drug-drug interactions (DDIs) and/or to act as an antagonist of multidrug resistance (MDR). In accumulation assays, alisertib potently inhibited ABCC1 transporter, but not ABCB1 or ABCG2. The results of molecular modeling suggested a bifunctional mechanism for interaction on ABCC1. In addition, alisertib was characterized as a low- to moderate-affinity inhibitor of recombinant CYP3A4, CYP2C8, CYP2C9, CYP2C19, and CYP2D6 isoenzymes, but without potential clinical relevance. Drug combination studies revealed the capability of alisertib to synergistically antagonize ABCC1-mediated resistance to daunorubicin. Although alisertib exhibited substrate characteristics toward ABCB1 transporter in monolayer transport assays, comparative proliferation studies showed lack of its MDR-victim behavior in cells overexpressing ABCB1 as well as ABCG2 and ABCC1. Lastly, alisertib did not affect the expression of ABCC1, ABCG2, ABCB1 transporters and CYP1A2, CYP3A4, CYP2B6 isozymes on mRNA level in various systemic and tumoral models. In conclusion, our study suggests that alisertib is a drug candidate with negligible potential for perpetrating systemic pharmacokinetic DDIs on ABCB1, ABCG2 and cytochromes P450. In addition, we introduce alisertib as an effective dual-activity chemosensitizer whose MDR-antagonistic capacities are not impaired by efflux or effect on MDR phenotype. Our in vitro findings provide important pieces of information for clinicians when introducing alisertib into the clinical area.
Collapse
|
4
|
Liewer S, Huddleston A. Alisertib: a review of pharmacokinetics, efficacy and toxicity in patients with hematologic malignancies and solid tumors. Expert Opin Investig Drugs 2018; 27:105-112. [PMID: 29260599 DOI: 10.1080/13543784.2018.1417382] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Aurora kinases are essential mediators in cell mitosis. Amplification of these kinases can lead to the development of malignancy and may be associated with inferior survival. Alisertib is an oral aurora kinase inhibitor which has been shown to induce cell-cycle arrest and apoptosis in preclinical studies. It is currently under investigation for a wide variety of malignancies including hematologic (specifically Non-Hodgkin's lymphoma) and solid tumors. Areas covered: A PubMed search was performed to identify clinical studies reporting outcomes with alisertib. Promising results are notable in patients with peripheral T cell lymphoma in particular, forming the basis for the first phase 3 randomized trial of alisertib. Although it did show encouraging response rates, it failed to demonstrate superiority over the comparator arm at an interim analysis, halting further enrollment. Expert opinion: Despite disappointing early results, alisertib remains under investigation in a number of cancer types both as monotherapy and in combination with traditional cytotoxic chemotherapy, with encouraging results. Most common toxicities in early trials include myelosuppression alopecia, mucositis and fatigue. The relatively manageable toxicity profile of alisertib along with ease of dosing may allow it to be combined with other oral agents or traditional chemotherapy across a wide variety of malignancy types.
Collapse
Affiliation(s)
- Susanne Liewer
- a Department of Pharmacy , Nebraska Medicine , Omaha , NE , USA.,b College of Pharmacy , University of Nebraska Medical Center , Omaha , NE , USA
| | - Ashley Huddleston
- c Department of Pharmacy , Mercy Hospital , Oklahoma City , OK , USA
| |
Collapse
|
5
|
Raaphorst RM, Savolainen H, Cantore M, van de Steeg E, van Waarde A, Colabufo NA, Elsinga PH, Lammertsma AA, Windhorst AD, Luurtsema G. Comparison of In Vitro Assays in Selecting Radiotracers for In Vivo P-Glycoprotein PET Imaging. Pharmaceuticals (Basel) 2017; 10:ph10030076. [PMID: 29036881 PMCID: PMC5620620 DOI: 10.3390/ph10030076] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 09/13/2017] [Accepted: 09/15/2017] [Indexed: 12/24/2022] Open
Abstract
Positron emission tomography (PET) imaging of P-glycoprotein (P-gp) in the blood-brain barrier can be important in neurological diseases where P-gp is affected, such as Alzheimer´s disease. Radiotracers used in the imaging studies are present at very small, nanomolar, concentration, whereas in vitro assays where these tracers are characterized, are usually performed at micromolar concentration, causing often discrepant in vivo and in vitro data. We had in vivo rodent PET data of [11C]verapamil, (R)-N-[18F]fluoroethylverapamil, (R)-O-[18F]fluoroethyl-norverapamil, [18F]MC225 and [18F]MC224 and we included also two new molecules [18F]MC198 and [18F]KE64 in this study. To improve the predictive value of in vitro assays, we labeled all the tracers with tritium and performed bidirectional substrate transport assay in MDCKII-MDR1 cells at three different concentrations (0.01, 1 and 50 µM) and also inhibition assay with P-gp inhibitors. As a comparison, we used non-radioactive molecules in transport assay in Caco-2 cells at a concentration of 10 µM and in calcein-AM inhibition assay in MDCKII-MDR1 cells. All the P-gp substrates were transported dose-dependently. At the highest concentration (50 µM), P-gp was saturated in a similar way as after treatment with P-gp inhibitors. Best in vivo correlation was obtained with the bidirectional transport assay at a concentration of 0.01 µM. One micromolar concentration in a transport assay or calcein-AM assay alone is not sufficient for correct in vivo prediction of substrate P-gp PET ligands.
Collapse
Affiliation(s)
- Renske M Raaphorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1085C, 1081 HV Amsterdam, The Netherlands.
| | - Heli Savolainen
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Mariangela Cantore
- Dipartimento di Farmacia-Scienze del Farmaco, Università Degli Studi di Bari, via Orabona 4, 70125 Bari, Italy.
- Biofordrug slr, via Orabona 4, 70125 Bari, Italy.
| | - Evita van de Steeg
- Microbiology Systems and Biology Group, Netherlands Organisation for Applied Scientific Research (TNO), Utrechtseweg 48, 3704 HE Zeist, The Netherlands.
| | - Aren van Waarde
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Nicola A Colabufo
- Dipartimento di Farmacia-Scienze del Farmaco, Università Degli Studi di Bari, via Orabona 4, 70125 Bari, Italy.
- Biofordrug slr, via Orabona 4, 70125 Bari, Italy.
| | - Philip H Elsinga
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| | - Adriaan A Lammertsma
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1085C, 1081 HV Amsterdam, The Netherlands.
| | - Albert D Windhorst
- Department of Radiology & Nuclear Medicine, VU University Medical Center, De Boelelaan 1085C, 1081 HV Amsterdam, The Netherlands.
| | - Gert Luurtsema
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands.
| |
Collapse
|
6
|
Ahamed M, Verbeek J, Funke U, Lecina J, Verbruggen A, Bormans G. Recent Progress in Metal Catalyzed Direct Carboxylation of Aryl Halides and Pseudo Halides Employing CO2: Opportunities for11C Radiochemistry. ChemCatChem 2016. [DOI: 10.1002/cctc.201600943] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
| | | | - Uta Funke
- Laboratory of Radiopharmacy; KU Leuven; Belgium
| | - Joan Lecina
- Laboratory of Radiopharmacy; KU Leuven; Belgium
| | | | - Guy Bormans
- Laboratory of Radiopharmacy; KU Leuven; Belgium
| |
Collapse
|
7
|
Durlacher CT, Li ZL, Chen XW, He ZX, Zhou SF. An update on the pharmacokinetics and pharmacodynamics of alisertib, a selective Aurora kinase A inhibitor. Clin Exp Pharmacol Physiol 2016; 43:585-601. [DOI: 10.1111/1440-1681.12571] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 03/12/2016] [Accepted: 03/15/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Cameron T Durlacher
- Department of Pharmaceutical Sciences; College of Pharmacy; University of South Florida; Tampa FL USA
| | - Zhi-Ling Li
- Department of Pharmacy; Shanghai Children's Hospital; Shanghai Jiao Tong University; Shanghai China
| | - Xiao-Wu Chen
- Department of General Surgery; The First People's Hospital of Shunde Affiliated to Southern Medical University; Shunde Foshan Guangdong
| | - Zhi-Xu He
- Guizhou Provincial Key Laboratory for Regenerative Medicine; Stem Cell and Tissue Engineering Research Centre & Sino-US Joint Laboratory for Medical Sciences; Guizhou Medical University; Guiyang Guizhou China
| | - Shu-Feng Zhou
- Department of Pharmaceutical Sciences; College of Pharmacy; University of South Florida; Tampa FL USA
| |
Collapse
|
8
|
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
|