@phdthesis{Jede2019, author = {Christian Jede}, title = {Development of translational biopharmaceutical assays for biorelevant dissolution and precipitation testing of poorly soluble drugs}, journal = {Entwicklung translationaler biopharmazeutischer Assays zur biorelevanten Untersuchung des L{\"o}slichkeits- und Pr{\"a}zipitationsverhaltens schwerl{\"o}slicher Wirkstoffe}, url = {https://nbn-resolving.org/urn:nbn:de:gbv:9-opus-33949}, pages = {109}, year = {2019}, abstract = {From a biopharmaceutical point of view, poor oral bioavailability of a drug is one of the greatest challenges for formulation scientists. The majority of new chemical entities (NCEs) are weakly basic drugs. Consequently, these drugs exhibit pH-dependent solubility, being higher under acidic conditions in the fasted stomach and lower under neutral conditions in the small intestine, the main site of drug absorption. For theses compounds, pH-dependent precipitation testing represents a key parameter during early development stages. In this development phase, the amount of drug available is limited, and fast and detailed investigations of simulated drug solubility are desired. Therefore, an automated small-scale in vitro transfer model, simulating drug transfer from a donor (stomach; simulated gastric fluid, SGF pH 2.0) to an acceptor (small intestine; fasted state simulated intestinal fluid, FaSSIF-phosphate pH 6.5) compartment, has been developed. In contrast to the originally published transfer model, this model allowed a detailed investigation of drug supersaturation and precipitation in a small-scale, feasible for pre-formulation purposes, through miniaturization and automation in an in-line analytical set-up. In-line drug concentration analysis in turbid samples, due to pH-dependent drug precipitation, was achieved by a pre-filtration step, the use of flow-through cuvettes and the application of UV derivative spectroscopy. Compared to the common procedure of manual sampling followed by HPLC-UV analysis for concentration determination, the supersaturation and precipitation of the model drug ketoconazole was more accurately captured by the newly developed in-line analytical set-up. In addition, the newly developed small-scale model was compared to a USP II-based transfer model, representing an established scale of the transfer model. Using a physiologically relevant simulated gastric emptying rate of 5 min half-time, supersaturation and precipitation of the model drugs ketoconazole and a new chemical entity from the research laboratories of Merck Healthcare KGaA, MSC-A, were observed to be highly comparable. Following miniaturization and automation, the developed small-scale model was used to establish eight physiologically relevant test-sets. These test-sets were used to assess the impact of gastrointestinal (GI) variability, i.e. gastric pH, gastric emptying, and GI fluid volumes, on supersaturation and precipitation of two weakly basic model compounds, ketoconazole and MSC-A. The experiments revealed that variations in all GI parameters investigated affected the in vitro supersaturation and precipitation of ketoconazole. For example, faster gastric emptying yielded higher supersaturation and faster precipitation of ketoconazole. In contrast, MSC-A supersaturation and precipitation was only affected by variability in gastric pH. Consequently, the effect of varying GI parameters was found to be drug-specific. Elevated gastric pH, as it can result from co-medication with acid-reducing drugs, resulted in lower degrees of supersaturation for both substances. For ketoconazole, this result is in agreement with the observation that the oral bioavailability of ketoconazole is lowered when proton pump inhibitors are co-administered. In addition to the physiological considerations, the small-scale model developed herein was used to establish an in vitro screening assay for precipitation inhibitors (PIs). The use of PIs represents one option of reducing the process of pH-dependent drug precipitation during simulated GI transfer. For this purpose, ketoconazole and five orally administered kinase inhibitors (i.e. pazopanib, gefitinib, lapatinib, vemurafenib, and MSC-A) were analyzed with and without the polymeric PIs HPMC, HPMCAS, PVPK17 and K30, PEG6000, and Soluplus® in the small-scale transfer model. This screening revealed that at least one effective PI could be identified for each model drug. Moreover, HPMCAS and Soluplus® were the most effective PIs. Another outcome of these studies was that gefitinib expressed highly variable amorphous precipitation which was confirmed by powder X-ray diffraction (PXRD). During the transfer model experiments, the intermediate amorphous and supersaturated state of gefitinib was stabilized using HPMCAS and Soluplus®. After the polymer investigations, the impact of the buffer species in the simulated intestinal medium on drug supersaturation and precipitation was assessed. Since luminal fluids are mainly buffered by hydrogen carbonate ions, a USP II-based transfer model equipped with the pHysio-grad® device was proposed. This allowed the use of a complex bicarbonate buffer for the preparation of FaSSIF-bicarbonate in an in vitro transfer model. Results of transfer model experiments using standard phosphate-based FaSSIF and a more physiologically relevant bicarbonate-based FaSSIF were compared. Therefore, ketoconazole, pazopanib, and lapatinib were analyzed with and without the precipitation inhibitor HPMCAS. While HPMCAS was found to be an effective precipitation inhibitor for all drugs in FaSSIF-phosphate, the effect in FaSSIF-bicarbonate was much less pronounced. Additionally, performed rat PK studies revealed that HPMCAS did not increase the exposure of any of the model compounds significantly, indicating that the transfer model employing bicarbonate-buffered FaSSIF was more predictive compared to the model using phosphate-buffered FaSSIF. The in vitro and in vivo results of these studies demonstrated that the supersaturation precipitation of poorly soluble weakly basic drugs can be significantly affected by GI variability. Furthermore, the use of the automated small-scale transfer model enabled the identification of effective precipitation inhibitors for the model drugs involved in these studies. At the same time the buffer species has been observed to be especially important to reliably predict the in vivo solubility/dissolution behavior of HPMCAS and the weakly basic model drugs.}, language = {en} }