Translational Pharmaceutics and Early Phase Clinical Development
Shobha N. Bhattachar
Small Molecule Design, Eli Lilly and Company, Indianapolis, IN, USA
Search for more papers by this authorJeffrey S. Tan
Small Molecule Design, Eli Lilly and Company, Indianapolis, IN, USA
Search for more papers by this authorStephen Dion Stamatis
Small Molecule Design, Eli Lilly and Company, Indianapolis, IN, USA
Search for more papers by this authorJon Hilden
Small Molecule Design, Eli Lilly and Company, Indianapolis, IN, USA
Search for more papers by this authorShobha N. Bhattachar
Small Molecule Design, Eli Lilly and Company, Indianapolis, IN, USA
Search for more papers by this authorJeffrey S. Tan
Small Molecule Design, Eli Lilly and Company, Indianapolis, IN, USA
Search for more papers by this authorStephen Dion Stamatis
Small Molecule Design, Eli Lilly and Company, Indianapolis, IN, USA
Search for more papers by this authorJon Hilden
Small Molecule Design, Eli Lilly and Company, Indianapolis, IN, USA
Search for more papers by this authorAbstract
Translational pharmaceutics is a relatively new cross-disciplinary construct that integrates discovery and development for efficiently enabling discovery activities to deliver commercially viable medicines. This article describes the role and deliverables of its various subfunctions, namely, preclinical formulations, physical chemistry, drug product prototyping and development, and biopharmaceutics. The integrated aspects of these deliverables, that enable drug discovery, while also influencing and enabling the selection of compounds for clinical testing, in a manner that embraces the principles of R&D essentialism and design thinking, are also described. This article also illustrates how cost and time savings can be achieved through the application of robust mathematical modeling where possible, to guide the generation of the most relevant experimental data, followed by risk-based decision-making to inform commercial product design. Although the article is mostly focused on traditional small molecules, the principles described here can be adapted to include other types of molecular and biologic entities, including the newer molecular modalities that are rapidly gaining importance in the industry.
References
- 1Curatolo, W. (1998). Physical chemical properties of oral drug candidates in the discovery and exploratory development settings. Pharm. Sci. Technol. Today 1 (9): 387–393.
- 2Stegemann, S. et al. (2007). When poor solubility becomes an issue: from early stage to proof of concept. Eur. J. Pharm. Sci. 31 (5): 249–261.
- 3Di, L., Fish, P.V., and Mano, T. (2012). Bridging solubility between drug discovery and development. Drug Discov. Today 17 (9–10): 486–495.
- 4Venkatesh, S. and Lipper, R.A. (2000). Role of the development scientist in compound lead selection and optimization. J. Pharm. Sci. 89 (2): 145–154.
10.1002/(SICI)1520-6017(200002)89:2<145::AID-JPS2>3.0.CO;2-6 CAS PubMed Web of Science® Google Scholar
- 5Bhattachar, S.N., Bender, D.M., Sweetana, S.A., and Wesley, J.A. (2014). Discovery formulations: approaches and practices in early preclinical development. In: Discovering and Developing Molecules with Optimal Drug-Like Properties (ed. S. Byrn, A. Templeton, R. Haskell and T. Prisinzano), 49–94. Springer.
- 6Mu, J. et al. (2006). Chronic inhibition of dipeptidyl peptidase-4 with a sitagliptin analog preserves pancreatic beta-cell mass and function in a rodent model of type 2 diabetes. Diabetes 55 (6): 1695–1704.
- 7Tan, T., Watts, S.W., and Davis, R.P. (2011). Drug delivery: enabling technology for drug discovery and development. iPRECIO micro infusion pump: programmable, refillable, and implantable. Front. Pharmacol. 2: 44.
- 8Rhyasen, G.W. et al. (2016). AZD5153: a novel bivalent BET bromodomain inhibitor highly active against hematologic malignancies. Mol. Cancer Ther. 15 (11): 2563–2574.
- 9 Harro Höfliger Capsule filling machines. https://www.hoefliger.com/en/machines-technology/dosing-filling (accessed 12 February 2020).
- 10Maas, J., Kamm, W., and Hauck, G. (2007). An integrated early formulation strategy – from hit evaluation to preclinical candidate profiling. Eur. J. Pharm. Biopharm. 66 (1): 1–10.
- 11Kesisoglou, F. and Wu, Y. (2008). Understanding the effect of API properties on bioavailability through absorption modeling. AAPS J. 10 (4): 516–525.
- 12Akimoto, M. et al. (2000). Gastric pH profiles of beagle dogs and their use as an alternative to human testing. Eur. J. Pharm. Biopharm. 49 (2): 99–102.
- 13Sagawa, K. et al. (2009). Fed and fasted gastric pH and gastric residence time in conscious beagle dogs. J. Pharm. Sci. 98 (7): 2494–2500.
- 14Bhattachar, S.N. et al. (2011). Effect of gastric pH on the pharmacokinetics of a BCS class II compound in dogs: utilization of an artificial stomach and duodenum dissolution model and GastroPlus, simulations to predict absorption. J. Pharm. Sci. 100 (11): 4756–4765.
- 15He, Y. and Ho, C. (2015). Amorphous solid dispersions: utilization and challenges in drug discovery and development. J. Pharm. Sci. 104 (10): 3237–3258.
- 16Horter, D. and Dressman, J.B. (2001). Influence of physicochemical properties on dissolution of drugs in the gastrointestinal tract. Adv. Drug Deliv. Rev. 46 (1–3): 75–87.
- 17Ishikawa, M. and Hashimoto, Y. (2011). Improvement in aqueous solubility in small molecule drug discovery programs by disruption of molecular planarity and symmetry. J. Med. Chem. 54 (6): 1539–1554.
- 18Grimbergen, R.F., Bennema, P., and Meekes, H. (1999). On the prediction of crystal morphology. III. Equilibrium and growth behaviour of crystal faces containing multiple connected nets. Acta Crystallogr. A 55 (Pt 1): 84–94.
- 19Briggner, L.E. et al. (2011). Solid-state perturbation for solubility improvement: a proof of concept. ChemMedChem 6 (1): 60–62.
- 20Williams, H.D. et al. (2013). Strategies to address low drug solubility in discovery and development. Pharmacol. Rev. 65 (1): 315–499.
- 21Serajuddin, A.T. (2007). Salt formation to improve drug solubility. Adv. Drug Deliv. Rev. 59 (7): 603–616.
- 22McNamara, D.P. et al. (2006). Use of a glutaric acid cocrystal to improve oral bioavailability of a low solubility API. Pharm. Res. 23 (8): 1888–1897.
- 23Childs, S.L., Kandi, P., and Lingireddy, S.R. (2013). Formulation of a danazol cocrystal with controlled supersaturation plays an essential role in improving bioavailability. Mol. Pharm. 10 (8): 3112–3127.
- 24Newman, A. and Wenslow, R. (2016). Solid form changes during drug development: good, bad, and ugly case studies. AAPS Open 2: 2.
10.1186/s41120-016-0003-4 Google Scholar
- 25Leuner, C. and Dressman, J. (2000). Improving drug solubility for oral delivery using solid dispersions. Eur. J. Pharm. Biopharm. 50 (1): 47–60.
- 26Engers, D. et al. (2010). A solid-state approach to enable early development compounds: selection and animal bioavailability studies of an itraconazole amorphous solid dispersion. J. Pharm. Sci. 99 (9): 3901–3922.
- 27DiNunzio, J.C. et al. (2010). Production of advanced solid dispersions for enhanced bioavailability of itraconazole using KinetiSol dispersing. Drug Dev. Ind. Pharm. 36 (9): 1064–1078.
- 28Zhao, Y. et al. (2011). Prediction of the thermal phase diagram of amorphous solid dispersions by Flory-Huggins theory. J. Pharm. Sci. 100 (8): 3196–3207.
- 29Lin, D. and Huang, Y. (2010). A thermal analysis method to predict the complete phase diagram of drug-polymer solid dispersions. Int. J. Pharm. 399 (1–2): 109–115.
- 30Sun, Y. et al. (2010). Solubilities of crystalline drugs in polymers: an improved analytical method and comparison of solubilities of indomethacin and nifedipine in PVP, PVP/VA, and PVAc. J. Pharm. Sci. 99 (9): 4023–4031.
- 31Tian, Y. et al. (2013). Construction of drug-polymer thermodynamic phase diagrams using Flory-Huggins interaction theory: identifying the relevance of temperature and drug weight fraction to phase separation within solid dispersions. Mol. Pharm. 10 (1): 236–248.
- 32Brunsteiner, M., Khinast, J., and Paudel, A. (2018). Relative contributions of solubility and mobility to the stability of amorphous solid dispersions of poorly soluble drugs: a molecular dynamics simulation study. Pharmaceutics 10 (3): 101.
- 33Gupta, J. et al. (2011). Prediction of solubility parameters and miscibility of pharmaceutical compounds by molecular dynamics simulations. J. Phys. Chem. B 115 (9): 2014–2023.
- 34Huynh, L. et al. (2008). Predicting the solubility of the anti-cancer agent docetaxel in small molecule excipients using computational methods. Pharm. Res. 25 (1): 147–157.
- 35Xiang, T.X. and Anderson, B.D. (2013). Molecular dynamics simulation of amorphous indomethacin-poly(vinylpyrrolidone) glasses: solubility and hydrogen bonding interactions. J. Pharm. Sci. 102 (3): 876–891.
- 36Baird, J.A. and Taylor, L.S. (2012). Evaluation of amorphous solid dispersion properties using thermal analysis techniques. Adv. Drug Deliv. Rev. 64 (5): 396–421.
- 37Baird, J.A., Van Eerdenbrugh, B., and Taylor, L.S. (2010). A classification system to assess the crystallization tendency of organic molecules from undercooled melts. J. Pharm. Sci. 99 (9): 3787–3806.
- 38Yang, J., Grey, K., and Doney, J. (2010). An improved kinetics approach to describe the physical stability of amorphous solid dispersions. Int. J. Pharm. 384 (1–2): 24–31.
- 39Zhou, D. et al. (2008). Thermodynamics, molecular mobility and crystallization kinetics of amorphous griseofulvin. Mol. Pharm. 5 (6): 927–936.
- 40Newman, A., Knipp, G., and Zografi, G. (2012). Assessing the performance of amorphous solid dispersions. J. Pharm. Sci. 101 (4): 1355–1377.
- 41Ding, X., Rose, J.P., and Van Gelder, J. (2012). Developability assessment of clinical drug products with maximum absorbable doses. Int. J. Pharm. 427 (2): 260–269.
- 42Refsgaard, H.H. et al. (2005). In silico prediction of membrane permeability from calculated molecular parameters. J. Med. Chem. 48 (3): 805–811.
- 43Volpe, D.A. (2011). Drug-permeability and transporter assays in Caco-2 and MDCK cell lines. Future Med. Chem. 3 (16): 2063–2077.
- 44Bhattachar, S.N., Tan, J.S., and Bender, D.M. (2017). Developability assessment of clinical candidates. In: Translating Molecules into Medicines (ed. J. Morrison, S. Bhattachar, D. Mudra and D. Bender), 231–266. Springer.
10.1007/978-3-319-50042-3_7 Google Scholar
- 45Saeed, A. et al. (2016). 2-Chloro-4-[[(1R,2R)-2-hydroxy-2-methyl-cyclopentyl]amino]-3-methyl-benzonitrile: a transdermal selective androgen receptor modulator (SARM) for muscle atrophy. J. Med. Chem. 59 (2): 750–755.
- 46Thakral, N.K. and Kelly, R.C. (2017). Salt disproportionation: a material science perspective. Int. J. Pharm. 520 (1–2): 228–240.
- 47Thakral, N.K. et al. (2016). Salt disproportionation in the solid state: role of solubility and counterion volatility. Mol. Pharm. 13 (12): 4141–4151.
- 48Patel, M.A. et al. (2018). Effect of excipient properties, water activity, and water content on the disproportionation of a pharmaceutical salt. Int. J. Pharm. 546 (1–2): 226–234.
- 49Merritt, J.M., Viswanath, S.K., and Stephenson, G.A. (2013). Implementing quality by design in pharmaceutical salt selection: a modeling approach to understanding disproportionation. Pharm. Res. 30 (1): 203–217.
- 50Nie, H. et al. (2016). Impact of metallic stearates on disproportionation of hydrochloride salts of weak bases in solid-state formulations. Mol. Pharm. 13 (10): 3541–3552.
- 51Urabe, M. et al. (2003). Assessment of tableting properties using infinitesimal quantities of powdered medicine. Int. J. Pharm. 263 (1–2): 183–187.
- 52Tye, C.K., Sun, C.C., and Amidon, G.E. (2005). Evaluation of the effects of tableting speed on the relationships between compaction pressure, tablet tensile strength, and tablet solid fraction. J. Pharm. Sci. 94 (3): 465–472.
- 53Perumalla, S.R. and Sun, C.C. (2014). Enabling tablet product development of 5-fluorocytosine through integrated crystal and particle engineering. J. Pharm. Sci. 103 (4): 1126–1132.
- 54Roth, W.J. et al. (2017). A demonstration of mixing robustness in a direct compression continuous manufacturing process. J. Pharm. Sci. 106 (5): 1339–1346.
- 55Schaber, S.D., Gerogiorgis, D.I., Ramachandran, R., Evans, J.M.B., Barton, P.I., and Trout, B.L. (2011). Economic analysis of integrated continuous and batch pharmaceutical manufacturing: a case study. Ind. Eng. Chem. Res. 50 (11): 10083–10092.
- 56Srai, J.S. et al. (2015). Future supply chains enabled by continuous processing-opportunities challenges May 20-21 2014 continuous manufacturing symposium. J. Pharm. Sci. 104 (3): 840–849.
- 57Lee, S.L., O'Connor, T.F., Yang, X. et al. (2015). Modernizing pharmaceutical manufacturing: from batch to continuous production. J. Pharm. Innov. 10 (3): 191–199.
- 58Kushner, J.T. and Moore, F. (2010). Scale-up model describing the impact of lubrication on tablet tensile strength. Int. J. Pharm. 399 (1–2): 19–30.
- 59Kushner, J.t. and Schlack, H. (2014). Commercial scale validation of a process scale-up model for lubricant blending of pharmaceutical powders. Int. J. Pharm. 475 (1–2): 147–155.
- 60Hilden, J., Schrad, M., Reynolds, J. et al. (2017). Investigation of an intensifier-bar tumble bin scale-up model. Powder Technol. 305 (1): 723–738.
- 61Ebey, G.C. (1987). A thermodynamic model for aqueous film-coating. Pharm. Technol. 11 (4): 41–46.
- 62Strong, J.C. (2009). Psychrometric analysis of the environmental equivalency factor for aqueous tablet coating. AAPS PharmSciTech 10 (1): 303–309.
- 63am Ende, M.T. and Berchielli, A. (2005). A thermodynamic model for organic and aqueous tablet film coating. Pharm. Dev. Technol. 10 (1): 47–58.
- 64Akseli, I., Ladyzhynsky, N. et al. (2013). Development of predictive tools to assess capping tendency of tablet formulations. Powder Technol. 236 (2): 139–148.
- 65Herting, M.G. and Kleinebudde, P. (2008). Studies on the reduction of tensile strength of tablets after roll compaction/dry granulation. Eur. J. Pharm. Biopharm. 70 (1): 372–379.
- 66He, X., Secreast, P.J., and Amidon, G.E. (2007). Mechanistic study of the effect of roller compaction and lubricant on tablet mechanical strength. J. Pharm. Sci. 96 (5): 1342–1355.
- 67Patel, S. et al. (2011). Understanding size enlargement and hardening of granules on tabletability of unlubricated granules prepared by dry granulation. J. Pharm. Sci. 100 (2): 758–766.
- 68Sun, C.C. and Himmelspach, M.W. (2006). Reduced tabletability of roller compacted granules as a result of granule size enlargement. J. Pharm. Sci. 95 (1): 200–206.
- 69Mitra, B., Hilden, J., and Litster, J.D. (2015). Novel use of monodisperse granules to deconvolute impacts of granule size versus granule solid fraction on tablet tensile strength. Adv. Powder Technol. 26 (2): 553–562.
- 70Mitra, B., Hilden, J., and Litster, J.D. (2016). Effects of the granule composition on the compaction behavior of deformable dry granules. Powder Technol. 291 (4): 487–498.
- 71Mitra, B., Hilden, J., and Litster, J.D. (2016). Compaction mechanics of plastically deformable dry granules. Powder Technol. 291 (4): 328–336.
- 72Hapgood, K.P., Litster, J.D., and Smith, R. (2003). Nucleation regime map for liquid bound granules. AIChE J. 49 (2): 350–361.
- 73Kayrak-Talay, D. and Litster, J.D. (2011). A priori performance prediction in pharmaceutical wet granulation: testing the applicability of the nucleation regime map to a formulation with a broad size distribution and dry binder addition. Int. J. Pharm. 418 (2): 254–264.
- 74Knight, P.C., Johansen, R., Kristensen, H.G. et al. (2000). An investigation of the effects on agglomeration of changing the speed of a mechanical mixer. Powder Technol. 110 (3): 204–209.
- 75Liu, L.X., Smith, R., and Litster, J.D. (2009). Wet granule breakage in a breakage only high-hear mixer: effect of formulation properties on breakage behaviour. Powder Technol. 189 (2): 158–164.
- 76Saleh, K., Vialatte, L., and Guigon, P. (2005). Wet granulation in a batch high shear mixer. Chem. Eng. Sci. 60 (14): 3763–3775.
- 77Wade, J.B., Martin, G.P., and Long, D.F. (2015). Controlling granule size through breakage in a novel reverse-phase wet granulation process: the effect of impeller speed and binder liquid viscosity. Int. J. Pharm. 478 (2): 439–446.
- 78Wade, J.B., Martin, G.P., and Long, D.F. (2014). Feasibility assessment for a novel reverse-phase wet granulation process: the effect of liquid saturation and binder liquid viscosity. Int. J. Pharm. 475 (1–2): 450–461.
- 79Hilden, J. et al. (2012). A first-principles model for prediction of product dose uniformity based on drug substance particle size distribution. J. Pharm. Sci. 101 (7): 2364–2371.
- 80Huang, C.Y. and Sherry Ku, M. (2010). Prediction of drug particle size and content uniformity in low-dose solid dosage forms. Int. J. Pharm. 383 (1–2): 70–80.
- 81Rohrs, B.R. et al. (2006). Particle size limits to meet USP content uniformity criteria for tablets and capsules. J. Pharm. Sci. 95 (5): 1049–1059.
- 82Huang, C.Y. and Ku, M.S. (2010). Asymmetry effect of particle size distribution on content uniformity and over-potency risk in low-dose solid drugs. J. Pharm. Sci. 99 (10): 4351–4362.
- 83Yalkowsky, S.H. and Bolton, S. (1990). Particle size and content uniformity. Pharm. Res. 7 (9): 962–966.
- 84Hilden, J., Sullivan, M., Polizzi, M., Wade, J. et al. (2018). Power consumption during oscillatory mixing of pharmaceutical powders. Powder Technol. 338 (10): 44–54.
- 85McDermott, T.S., Farrenkopf, J., Hlinak, A. et al. (2011). A material sparing method for quantitatively measuring tablet sticking. Powder Technol. 212 (1): 240–252.
- 86Waimer, F. et al. (1999). The influence of engravings on the sticking of tablets. Investigations with an instrumented upper punch. Pharm. Dev. Technol. 4 (3): 369–375.
- 87Sabir, A., Evans, B., and Jain, S. (2001). Formulation and process optimization to eliminate picking from market image tablets. Int. J. Pharm. 215 (1-2): 123–135.
- 88Roberts, M. et al. (2003). Effects of surface roughness and chrome plating of punch tips on the sticking tendencies of model ibuprofen formulations. J. Pharm. Pharmacol. 55 (9): 1223–1228.
- 89Schumann, S. and Searle, G.D. (1992). The effects of chromium nitride ION bombardment treatment of tablet tooling on tablet adherence. Drug Dev. Ind. Pharm. 18 (10): 1037–1061.
- 90Swaminathan, S., Ramey, B., Hilden, J., and Wassgren, C. (2017). Characterizing the powder punch-face adhesive interaction during the unloading phase of powder compaction. Powder Technol. 315 (June): 410–421.
- 91Swaminathan, S., Ramey, B., Hilden, J., and Wassgren, C. (2016). Modeling the formation of debossed features on a pharmaceutical tablet. J. Pharm. Innov. 11 (3): 214–230.
- 92Zettler, A., Hilden, J., Koenig, M. et al. (2016). Evaluation of small-scale powder flow characterization tests in the prediction of large-scale process failures. J. Pharm. Innov. 11 (3): 189–199.
- 93Polizzi, M.A., Franchville, J., and Hilden, J. (2016). Assessment and predictive modeling of pharmaceutical powder flow behavior in small-scale hoppers. Powder Technol. 294 (June): 30–42.
- 94Ambler, C.M., Thombre, A.G., Gokhale, M., and Morrison, J.S. (2017). Design of clinical formulations in early development. In: Translating Molecules into Medicines (ed. J. Morrison, S. Bhattachar, D. Mudra and D. Bender). Springer.
10.1007/978-3-319-50042-3_10 Google Scholar
- 95Agoram, B., Woltosz, W.S., and Bolger, M.B. (2001). Predicting the impact of physiological and biochemical processes on oral drug bioavailability. Adv. Drug Deliv. Rev. 50 (Suppl 1): S41–S67.
- 96Stillhart, C. et al. (2019). PBPK absorption modeling: establishing the in vitro-in vivo link-industry perspective. AAPS J. 21 (2): 19.
- 97Pepin, X.J. et al. (2016). Justification of drug product dissolution rate and drug substance particle size specifications based on absorption PBPK modeling for Lesinurad immediate release tablets. Mol. Pharm. 13 (9): 3256–3269.
- 98Wagner, C. et al. (2012). Predicting the oral absorption of a poorly soluble, poorly permeable weak base using biorelevant dissolution and transfer model tests coupled with a physiologically based pharmacokinetic model. Eur. J. Pharm. Biopharm. 82 (1): 127–138.
- 99Noyes, A.A. and Whitney, W.R. (1897). The rate of solution of solid substances in their own solutions. J. Am. Chem. Soc. 19 (12): 930–934.
10.1021/ja02086a003 Google Scholar
- 100Dokoumetzidis, A. and Macheras, P. (2006). A century of dissolution research: from Noyes and Whitney to the biopharmaceutics classification system. Int. J. Pharm. 321 (1–2): 1–11.
- 101Sun, R. et al. (2016). Transition-tempered metadynamics is a promising tool for studying the permeation of drug-like molecules through membranes. J. Chem. Theory Comput. 12 (10): 5157–5169.
- 102Sun, R. et al. (2018). Molecular transport through membranes: accurate permeability coefficients from multidimensional potentials of mean force and local diffusion constants. J. Chem. Phys. 149 (7): 072310.
- 103Miller, N.A. et al. (2019). Physiologically based pharmacokinetic modelling for first-in-human predictions: an updated model building strategy illustrated with challenging industry case studies. Clin. Pharmacokinet. 58 (6): 727–746.
- 104Johnson, K.C. and Swindell, A.C. (1996). Guidance in the setting of drug particle size specifications to minimize variability in absorption. Pharm. Res. 13 (12): 1795–1798.
- 105Agoram, B., Woltosz, W.S., and Bolger, M.B. (2001). Predicting the impact of physiological and biochemical processes on oral drug bioavailability. Adv. Drug Deliv. Rev. 50: S41–S67.
- 106Sugano, K. (2009). Introduction to computational oral absorption simulation. Expert Opin. Drug Metab. Toxicol. 5 (3): 259–293.
- 107Kostewicz, E.S. et al. (2014). PBPK models for the prediction of in vivo performance of oral dosage forms. Eur. J. Pharm. Sci. 57: 300–321.
- 108Skvara, H. et al. (2011). Topical treatment of Basal cell carcinomas in nevoid Basal cell carcinoma syndrome with a smoothened inhibitor. J. Invest. Dermatol. 131 (8): 1735–1744.
- 109Epstein, E.H. et al. (2018). Hedgehog pathway inhibition by topical patidegib to reduce BCC burden in patients with basal cell nevus (Gorlin) syndrome. J. Clin. Oncol. 36 (15_suppl): e21626.
- 110Arrighi, I. et al. (2009). Bone healing induced by local delivery of an engineered parathyroid hormone prodrug. Biomaterials 30 (9): 1763–1771.
- 111Kyllönen, L. et al. (2015). Local drug delivery for enhancing fracture healing in osteoporotic bone. Acta Biomater. 11: 412–434.
- 112Rowland, M. and Tozer, T.N. (2011). Clinical pharmacokinetics and pharmacodynamics: concepts and applications, 4e. Philadelphia: Wolters Kluwer Health/Lippincott William & Wilkins.
- 113Kaufmann, A.M. and Krise, J.P. (2007). Lysosomal sequestration of amine-containing drugs: analysis and therapeutic implications. J. Pharm. Sci. 96 (4): 729–746.
- 114Bolger, M.B. et al. (2019). The irrelevance of in vitro dissolution in setting product specifications for drugs like dextromethorphan that are subject to lysosomal trapping. J. Pharm. Sci. 108 (1): 268–278.
- 115Molloy, C. (2018). Pharma R&D productivity: discovering new medicines, in Open Access Government.
- 116Morrison, J., Squibb, B.M., and Louie, S. (2018). A small molecule renaissance? Teaching an old modality new tricks. AAPS Newsmagazine.