1,506
Views
25
CrossRef citations to date
0
Altmetric
Reviews

Transporter assays as useful in vitro tools in drug discovery and development

Bibliography

  • Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
  • Giacomini KM, Huang SM, Tweedie DJ, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9:215–236.

•• Consensus document from International Transporter Consortium on role of transporters in drug development.

  • Feng B, Varma MV, Costales C, et al. In vitro and in vivo approaches to characterize transporter-mediated disposition in drug discovery. Expert Opin Drug Discov. 2014;9:873–890.

• Includes a list of drugs with known transporter-based clinical drug interactions.

• Summarizes clinically observed drug–drug interactions with drug transporter involvement.

  • Lai Y, Sampson KE, Stevens JC. Evaluation of drug transporter interactions in drug discovery and development. Comb Chem High Throughput Screen. 2010;13: 112–134.

• Review article on transporter assays with screening strategies for drug discovery.

  • Sharma P, Holmes VE, Elsby R, et al. Validation of cell-based OATP1B1 assays to assess drug transport and the potential for drug-drug interaction to support regulatory submissions. Xenobiotica. 2010;40:24–37.

• Validation of OATP1B1 assay in HEK293 transfected cells.

  • Hillgren KM, Keppler D, Zur AA, et al. Emerging transporters of clinical importance: an update from the International Transporter Consortium. Clin Pharmacol Ther. 2013;94:52–63.
  • Polli JW, Wring SA, Humphreys JE, et al. Rational use of in vitro P-glycoprotein assays in drug discovery. J Pharmacol Exp Ther. 2001;299:620–628.
  • Hendrickx R, Johansson JG, Lohmann C, et al. Identification of novel substrates and structure-activity relationship of cellular uptake mediated by human organic cation transporters 1 and 2. J Med Chem. 2013;56:7232–7242.
  • Matsson P, Pederson JM, Norinder U, et al. Identification of novel specific and general inhibitors of the three major human ATP-binding cassette transporters P-gp, BCRP and MRP2 among registered drugs. Pharm Res. 2009;26:1816–1831.
  • Balimane PV, Chong S, Patel K, et al. Peptide transporter substrate identification during permeability screening in drug discovery: comparison of transfected MDCK-hPepT1 cells to Caco-2 cells. Arch Pharm Res. 2007;30:507–518.
  • Li R, Barton HA, Varma MV. Prediction of pharmacokinetics and drug-drug interactions when hepatic transporters are involved. Clin Pharmacokinet. 2014;53:659–678.
  • Varma MV, Lai Y, Kimoto E, et al. Mechanistic modeling to predict the transporter- and enzyme-mediated drug-drug interactions of repaglinide. Pharm Res. 2013;30:1188–1199.
  • Bi YA, Kimoto E, Sevidal S, et al. In vitro evaluation of hepatic transporter-mediated clinical drug-drug interactions: hepatocyte model optimization and retrospective investigation. Drug Metab Dispos. 2012;40:1085–1092.
  • Kishimoto W, Ishiguro N, Ludwig-Schwellinger E, et al. In vitro predictability of drug-drug interaction likelihood of P-glycoprotein-mediated efflux of dabigatran etexilate based on [I]2/IC50 threshold. Drug Metab Dispos. 2014;42:257–263.
  • Pedersen JM, Matsson P, Bergström CA, et al. Early identification of clinically relevant drug interactions with the human bile salt export pump (BSEP/ABCB11). Toxicol Sci. 2013;136:328–343.
  • Ciarimboli G, Holle SK, Vollenbröcker B, et al. New clues for nephrotoxicity induced by ifosfamide: preferential renal uptake via the human organic cation transporter 2. Mol Pharm. 2011;8: 270–279.
  • Huang SM, Zhang L, Giacomini KM. The International Transporter Consortium: a collaborative group of scientists from academia, industry, and the FDA. Clin Pharmacol Ther. 2010;87:32–36.
  • Pfeifer ND, Hardwick RN, Brouwer KL. Role of hepatic efflux transporters in regulating systemic and hepatocyte exposure to xenobiotics. Annu Rev Pharmacol Toxicol. 2014;54:509–535.
  • Dessilly G, Elens L, Panin N, et al. ABCB1 1199G>A genetic polymorphism (Rs2229109) influences the intracellular accumulation of tacrolimus in HEK293 and K562 recombinant cell lines. PLoS One. 2014;9:e91555.
  • Iwai M, Suzuki H, Ieiri I, et al. Functional analysis of single nucleotide polymorphisms of hepatic organic anion transporter OATP1B1 (OATP-C). Pharmacogenetics. 2004;14:749–757.
  • Tu M, Mathiowetz AM, Pfefferkorn JA, et al. Medicinal chemistry design principles for liver targeting through OATP transporters. Curr Top Med Chem. 2013;13: 857–866.
  • Varma MV, Ambler CM, Ullah M, et al. Targeting intestinal transporters for optimizing oral drug absorption. Curr Drug Metab. 2010;11:730–742.
  • Brouwer KL, Keppler D, Hoffmaster KA, et al. In vitro methods to support transporter evaluation in drug discovery and development. Clin Pharmacol Ther. 2013;94:95–112.

•• Review article on transporter assays and applications in drug development.

•• Review article on transporter assays including application, limitations and advantages.

  • De Bruyn T, Ye ZW, Peeters A, et al. Determination of OATP-, NTCP- and OCT-mediated substrate uptake activities in individual and pooled batches of cryopreserved human hepatocytes. Eur J Pharm Sci. 2011;43:297–307.
  • Kimoto E, Yoshida K, Balogh LM, et al. Characterization of organic anion transporting polypeptide (OATP) expression and its functional contribution to the uptake of substrates in human hepatocytes. Mol Pharm. 2012;9:3535–3542.
  • Marion TL, Perry CH, St Claire RL, et al. Endogenous bile acid disposition in rat and human sandwich-cultured hepatocytes. Toxicol Appl Pharmacol. 2012;261:1–9.
  • Ye ZW, Van Pelt J, Camus S, et al. Species-specific interaction of HIV protease inhibitors with accumulation of cholyl-glycylamido-fluorescein (CGamF) in sandwich-cultured hepatocytes. J Pharm Sci. 2010;99:2886–2898.
  • Schlatter P, Gutmann H, Drewe J. Primary porcine proximal tubular cells as a model for transepithelial drug transport in human kidney. Eur J Pharm Sci. 2006;28:141–154.
  • Lash LH, Putt DA, Cai H. Membrane transport function in primary cultures of human proximal tubular cells. Toxicology. 2006;228:200–218.
  • Brown CD, Sayer R, Windass AS, et al. Characterisation of human tubular cell monolayers as a model of proximal tubular xenobiotic handling. Toxicol Appl Pharmacol. 2008;233:428–438.
  • Miller DS, Nobmann SN, Gutmann H, et al. Xenobiotic transport across isolated brain microvessels studied by confocal microscopy. Mol Pharmacol. 2000;58:1357–1367.
  • Lee NY, Rieckmann P, Kang YS. The changes of P-glycoprotein activity by interferon-γ and tumor necrosis factor-α in primary and immortalized human brain microvascular endothelial cells. Biomol Ther. 2012;20:293–238.
  • Soars MG, Barton P, Ismair M, et al. The development, characterization, and application of an OATP1B1 inhibition assay in drug discovery. Drug Metab Dispos. 2012;40:1641–1648.

• Validation of OATP1B1 inhibition assay combined with in silico approaches.

  • Han YH, Busler D, Hong Y, et al. Transporter studies with the 3-O-sulfate conjugate of 17α-ethinylestradiol: assessment of human kidney drug transporters. Drug Metab Dispos. 2010;38:1064–1071.
  • Grottker J, Rosenberger A, Burckhardt G, et al. Interaction of human multidrug and toxin extrusion 1 (MATE1) transporter with antineoplastic agents. Drug Metab Drug Interact. 2011;26:181–189.
  • Kajiwara M, Masuda S, Watanabe S, et al. Renal tubular secretion of varenicline by multidrug and toxin extrusion (MATE) transporters. Drug Metab Pharmacokinet. 2012;27:563–569.
  • Sun P, Wang C, Liu Q, et al. OATP and MRP2-mediated hepatic uptake and biliary excretion of eprosartan in rat and human. Pharmacol Rep. 2014;66:311–319.
  • Taub ME, Mease K, Sane RS, et al. Digoxin is not a substrate for organic anion-transporting polypeptide transporters OATP1A2, OATP1B1, OATP1B3, and OATP2B1 but is a substrate for a sodium-dependent transporter expressed in HEK293 cells. Drug Metab Dispos. 2011;39:2093–2102.
  • Jeong HU, Kwon M, Lee Y, et al. Organic anion transporter 3- and organic anion transporting polypeptides 1B1- and 1B3-mediated transport of catalposide. Drug Des Devel Ther. 2015;9:643–653.
  • Leslie EM, Watkins PB, Kim RB, et al. Differential inhibition of rat and human Na+-dependent taurocholate cotransporting polypeptide (NTCP/SLC10A1) by bosentan: a mechanism for species differences in hepatotoxicity. J Pharmacol Exp Ther. 2007;321:1170–1178.
  • Bednarczyk D. Fluorescence-based assays for the assessment of drug interaction with the human transporters OATP1B1 and OATP1B3. Anal Biochem. 2010;405:50–58.
  • Müller F, König J, Hoier E, et al. Role of organic cation transporter OCT2 and multidrug and toxin extrusion proteins MATE1 and MATE2-K for transport and drug interactions of the antiviral lamivudine. Biochem Pharmacol. 2013;86(6):808–815.
  • Letschert K, Komatsu M, Hummel-Eisenbeiss J, et al. Vectorial transport of the peptide CCK-8 by double-transfected MDCKII cells stably expressing the organic anion transporter OATP1B3 (OATP8) and the export pump ABCC2. J Pharmacol Exp Ther. 2005;313:549–556.
  • Fahrmayr C, König J, Auge D, et al. Identification of drugs and drug metabolites as substrates of multidrug resistance protein 2 (MRP2) using triple-transfected MDCK-OATP1B1-UGT1A1-MRP2 cells. Br J Pharmacol. 2012;165:1836–1847.
  • Kopplow K, Letschert K, König J, et al. Human hepatobiliary transport of organic anions analyzed by quadruple-transfected cells. Mol Pharmacol. 2005;68:1031–1038.
  • Aschauer L, Carta G, Vogelsang N, et al. Expression of xenobiotic transporters in the human renal proximal tubule cell line RPTEC/TERT1. Toxicol In Vitro. Forthcoming 2014. DOI:10.1016/j.tiv.2014.12.003.
  • Schophuizen CM, Wilmer MJ, Jansen J, et al. Cationic uremic toxins affect human renal proximal tubule cell functioning through interaction with the organic cation transporter. Pflugers Arch. 2013;465:1701–1714.
  • Wilmer MJ, Saleem MA, Masereeuw R, et al. Novel conditionally immortalized human proximal tubule cell line expressing functional influx and efflux transporters. Cell Tissue Res. 2010;339:449–457.
  • Szabo M, Veres Z, Baranyai Z, et al. Comparison of human hepatoma HepaRG cells with human and rat hepatocytes in uptake transport assays in order to predict a risk of drug induced hepatotoxicity. PLoS One. 2013;8:e59432.
  • Le Vee M, Noel G, Jouan E, et al. Polarized expression of drug transporters in differentiated human hepatoma HepaRG cells. Toxicol In Vitro. 2013;27:1979–1986.
  • Bachour-El Azzi P, Sharanek A, Burban A, et al. Comparative localization and functional activity of the main hepatobiliary transporters in HepaRG cells and primary human hepatocytes. Toxicol Sci. 2015;145:157–168.
  • Bachmeier CJ, Trickler WJ, Miller DW. Comparison of drug efflux transport kinetics in various blood-brain barrier models. Drug Metab Dispos. 2006;34:998–1003.
  • Hakkarainen JJ, Jalkanen AJ, Kääriäinen TM, et al. Comparison of in vitro cell models in predicting in vivo brain entry of drugs. Int J Pharm. 2010;402:27–36.
  • Huls M, Brown CD, Windass AS, et al. The breast cancer resistance protein transporter ABCG2 is expressed in the human kidney proximal tubule apical membrane. Kidney Int. 2008;73:220–225.
  • Li M, Yuan H, Li N, et al. Identification of interspecies difference in efflux transporters of hepatocytes from dog, rat, monkey and human. Eur J Pharm Sci. 2008;35:114–126.
  • Lundquist P, Englund G, Skogastierna C, et al. Functional ATP-binding cassette drug efflux transporters in isolated human and rat hepatocytes significantly affect assessment of drug disposition. Drug Metab Dispos. 2014;42:448–458.
  • Takenaka T, Harada N, Kuze J, et al. Human small intestinal epithelial cells differentiated from adult intestinal stem cells as a novel system for predicting oral drug absorption in humans. Drug Metab Dispos. 2014;42:1947–1954.
  • Bentz J, O’Connor MP, Bednarczyk D, et al. Variability in P-glycoprotein inhibitory potency (IC50) using various in vitro experimental systems: implications for universal digoxin drug-drug interaction risk assessment decision criteria. Drug Metab Dispos. 2013;41:1347–1366.
  • Feng B, Mills JB, Davidson RE, et al. In vitro P-glycoprotein assays to predict the in vivo interactions of P-glycoprotein with drugs in the central nervous system. Drug Metab Dispos. 2008;36:268–275.
  • Mease K, Sane R, Podila L, et al. Differential selectivity of efflux transporter inhibitors in Caco-2 and MDCK-MDR1 monolayers: a strategy to assess the interaction of a new chemical entity with P-gp, BCRP, and MRP2. J Pharm Sci. 2012;101:1888–1897.
  • Volpe DA, Hamed SS, Zhang LK. Use of different parameters and equations for calculation of IC50 values in efflux assays: potential sources of variability in IC50 determination. AAPS J. 2014;16:172–180.
  • Poirier A, Portmann R, Cascais AC, et al. The need for human breast cancer resistance protein substrate and inhibition evaluation in drug discovery and development: why, when, and how? Drug Metab Dispos. 2014;42:1466–1477.
  • Xiao Y, Davidson R, Smith A, et al. A 96-well efflux assay to identify ABCG2 substrates using a stably transfected MDCK II cell line. Mol Pharm. 2006;3:45–54.
  • Tang F, Horie K, Borchardt RT. Are MDCK cells transfected with the human MDR1 gene a good model of the human intestinal mucosa? Pharm Res. 2002;19:765–772.
  • Tang F, Horie K, Borchardt RT. Are MDCK cells transfected with the human MRP2 gene a good model of the human intestinal mucosa? Pharm Res. 2002;19:773–779.
  • Robey RW, Lin B, Qiu J, et al. Rapid detection of ABC transporter interaction: potential utility in pharmacology. J Pharmacol Toxicol Methods. 2011;63:217–222.
  • Taur JS, Schuck EL, Wong NY. A transcellular assay to assess the P-gp inhibition in early stage of drug development. Drug Metab Lett. 2012;6:285–291.
  • van der Sandt ICJ, Blom-Roosemalen MCM, de Boer AG, et al. Specificity of doxorubicin versus rhodamine-123 in assessing P-glycoprotein functionality in the LLC-PK1, LLC-PK1:MDR1 and Caco-2 cell lines. Eur J Pharm Sci. 2000;11:207–214.
  • Shirasaka Y, Onishi Y, Sakuai A, et al. Evaluation of human P-glycoprotein (MDR1/ABCB1) ATPase activity assay method by comparing with in vitro transport measurements: Michaelis–Menten kinetic analysis to estimate the affinity of P-glycoprotein to drugs. Biol Pharm Bull. 2006;29:2465–2471.
  • Glavinas H, Kis E, Pál Á, et al. ABCG2 (breast cancer resistance protein/mitoxantrone resistance-associated protein) ATPase assay: a useful tool to detect drug-transporter interactions. Drug Metab Dispos. 2007;35:1533–1542.
  • Jin H, Di L. Permeability–in vitro assays for assessing drug transporter activity. Curr Drug Metab. 2008;9:911–920.
  • Herédi-Szabó K, Palm JE, Andersson TB, et al. A P-gp vesicular transport inhibition assay - optimization and validation for drug-drug interaction testing. Eur J Pharm Sci. 2013;49:773–781.
  • Glavinas H, Méhn D, Jani M, et al. Utilization of membrane vesicle preparations to study drug-ABC transporter interactions. Expert Opin Drug Metab Toxicol. 2008;4:721–732.
  • Leier I, Hummel-Eisenbeiss J, Cui Y, et al. ATP-dependent para-aminohippurate transport by apical multidrug resistance protein MRP2. Kidney Int. 2000;57:1636–1642.
  • Cui Y, König J, Buchholz JK, et al. Drug resistance and ATP-dependent conjugate transport mediated by the apical multidrug resistance protein, MRP2, permanently expressed in human and canine cells. Mol Pharmacol. 1999;55:929–937.
  • Elsby R, Smith V, Fox L, et al. Validation of membrane vesicle-based breast cancer resistance protein and multidrug resistance protein 2 assays to assess drug transport and the potential for drug-drug interaction to support regulatory submissions. Xenobiotica. 2011;41:764–783.

• Validation of vesicle MRP2 and BCRP assays for substrates and inhibitors.

  • Kidron H, Wissel G, Manevski N, et al. Impact of probe compound in MRP2 vesicular transport assays. Eur J Pharm Sci. 2012;46:100–105.
  • Glavinas H, von Richter O, Vojnits K, et al. Calcein assay: a high-throughput method to assess P-gp inhibition. Xenobiotica. 2011;41:712–719.
  • Balimane PV, Han YH, Chong S. Current industrial practices of assessing permeability and P-glycoprotein interaction. AAPS J. 2006;8:E1–13.
  • Li J, Volpe DA, Wang Y, et al. Use of transporter knockdown Caco-2 cells to investigate the in vitro efflux of statin drugs. Drug Metab Dispos. 2011;39:1196–1202.
  • Graber-Maier A, Gutmann H, Drewe J. A new intestinal cell culture model to discriminate the relative contribution of P-gp and BCRP on transport of substrates such as imatinib. Mol Pharm. 2010;7:1618–1628.
  • Elsby R, Surry DD, Smith VN, et al. Validation and application of Caco-2 assays for the in vitro evaluation of development candidate drugs as substrates or inhibitors of P-glycoprotein to support regulatory submissions. Xenobiotica. 2008;38:1140–1164.

• Validation of a Caco-2 P-gp assay for substrates and inhibitors.

  • Keogh JP, Kunta JR. Development, validation and utility of an in vitro technique for assessment of potential clinical drug-drug interactions involving P-glycoprotein. Eur J Pharm Sci. 2006;27:543–554.

• Validation of MDCK P-gp inhibition assay.

  • Giri N, Agarwal S, Shaik N, et al. Substrate-dependent breast cancer resistance protein (Bcrp1/Abcg2)-mediated interactions: consideration of multiple binding sites in in vitro assay design. Drug Metab Dispos. 2009;37:560–570.
  • Siissalo S, Hannukainen J, Kolehmainen J, et al. A Caco-2 cell based screening method for compounds interacting with MRP2 efflux protein. Eur J Pharm Biopharm. 2009;71:332–338.
  • Volpe DA. Drug-permeability and transporter assays in Caco-2 and MDCK cell lines. Future Med Chem. 2011;3:2063–2077.
  • Di L, Whitney-Pickett C, Umland JP, et al. Development of a new permeability assay using low-efflux MDCKII cells. J Pharm Sci. 2011;100:4974–4985.
  • Li J, Wang Y, Hidalgo IJ. Kinetic analysis of human and canine P-glycoprotein-mediated drug transport in MDR1-MDCK cell model: approaches to reduce false-negative substrate classification. J Pharm Sci. 2013;102:3436–3446.
  • Sampson KE, Brinker A, Pratt J, et al. Zinc finger nuclease-mediated gene knockout results in loss of transport activity for P-glycoprotein, BCRP, and MRP2 in Caco-2 cells. Drug Metab Dispos. 2015;43:199–207.
  • Sugimoto H, Matsumoto S, Tachibana M, et al. Establishment of in vitro P-glycoprotein inhibition assay and its exclusion criteria to assess the risk of drug-drug interaction at the drug discovery stage. J Pharm Sci. 2011;100:4013–4023.
  • Volpe DA. Variability in Caco-2 and MDCK cell-based intestinal permeability assays. J Pharm Sci. 2008;97:712–725.
  • Soars MG, Barton P, Elkin LL, et al. Application of an in vitro OAT assay in drug design and optimization of renal clearance. Xenobiotica. 2014;44:657–665.
  • Yamaguchi H, Takeuchi T, Okada M, et al. Screening of antibiotics that interact with organic anion-transporting polypeptides 1B1 and 1B3 using fluorescent probes. Biol. Pharm. Bull. 2011;34:389–395.
  • Ahlin G, Karlsson J, Pederson JM, et al. Structural requirements for drug inhibition of the liver specific human organic cation transport protein 1. J Med Chem. 2008;51:5932–5942.
  • Shin HHJ, Lee CH, Park SJ, et al. Establishment and characterization of Mardin-Darby canine kidney cells stably expressing human organic anion transporters. Arch Pharm Res. 2010;33:709–716.
  • Liu X, LeCluyse EL, Brouwer KR, et al. Use of Ca2+ modulation to evaluate biliary excretion in sandwich-cultured rat hepatocytes. J Pharmacol Exp Ther. 1999;289:1592–1599.
  • Swift B, Pfeifer ND, Brouwer KLR. Sandwich-cultured hepatocytes: an in vitro model to evaluate hepatobiliary transporter-based drug interactions and hepatotoxicity. Drug Metab Rev. 2010;42:446–471.
  • Bhoopathy S, Bode C, Naageshwaran V, et al. Principles and experimental considerations for in vitro transporter interaction assays. Methods Mol Biol. 2014;1113:229–252.

• Discussion of parameters influencing outcome of assays}.

  • Patil AG, D’Souza R, Dixit N, et al. Validation of quinidine as a probe substrate for the in vitro P-gp inhibition assay in Caco-2 cell monolayer. Eur J Drug Metab Pharmacokinet. 2011;36:115–119.
  • Siissalo S, Laitinen L, Koljonen M, et al. Effect of cell differentiation and passage number on the expression of efflux proteins in wild type and vinblastine-induced Caco-2 cell lines. Eur J Pharm Biopharm. 2007;67:548–554.
  • Badolo L, Trancart MM, Gustavsson L, et al. Effect of cryopreservation on the activity of OATP1B1/3 and OCT1 in isolated human hepatocytes. Chem Biol Interact. 2011;190:165–170.
  • Shirasaka Y, Sakane T, Yamashita S. Effect of P-glycoprotein expression levels on the concentration-dependent permeability of drugs to the cell membrane. J Pharm Sci. 2008;97:553–565.
  • Kis E, Ioja E, Nagy T, et al. Effect of membrane cholesterol on BSEP/Bsep activity: species specificity studies for substrates and inhibitors. Drug Metab Dispos. 2009;37:1878–1886.
  • Suzuyama N, Katoh M, Takeuchi T, et al. Species differences of inhibitory effects on P-glycoprotein-mediated drug transport. J Pharm Sci. 2007;96:1609–1618.
  • Chu X, Bleasby K, Evers R. Species differences in drug transporters and implications for translating preclinical findings to humans. Expert Opin Drug Metab Toxicol. 2013;9:237–252.
  • Katoh M, Suzuyama N, Takeuchi T, et al. Kinetic analyses for species differences in P-glycoprotein-mediated drug transport. J Pharm Sci. 2006;95:2673–2683.
  • Zimmermann C, Van De Wetering K, Van De Steeg E, et al. Species-dependent transport and modulation properties of human and mouse multidrug resistance protein 2 (MRP2/Mrp2, ABCC2/Abcc2). Drug Metab Dispos. 2008;36:631–640.
  • Balimane PV, Marino A, Chong S. P-gp inhibition potential in cell-based models: which “calculation” method is the most accurate? AAPS J. 2008;10:577–586.
  • Bjornsson TD, Callaghan JT, Einolf HJ, et al. The conduct of in vitro and in vivo drug-drug interaction studies: A PhRMA perspective. J Clin Pharmacol. 2003;43:443–469.
  • Volpe DA. Application of method suitability for drug permeability classification. AAPS J. 2010;12:670–678.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.