1
|
Hong RS, Rojas AV, Bhardwaj RM, Wang L, Mattei A, Abraham NS, Cusack KP, Pierce MO, Mondal S, Mehio N, Bordawekar S, Kym PR, Abel R, Sheikh AY. Free Energy Perturbation Approach for Accurate Crystalline Aqueous Solubility Predictions. J Med Chem 2023; 66:15883-15893. [PMID: 38016916 DOI: 10.1021/acs.jmedchem.3c01339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Early assessment of crystalline thermodynamic solubility continues to be elusive for drug discovery and development despite its critical importance, especially for the ever-increasing fraction of poorly soluble drug candidates. Here we present a detailed evaluation of a physics-based free energy perturbation (FEP+) approach for computing the thermodynamic aqueous solubility. The predictive power of this approach is assessed across diverse chemical spaces, spanning pharmaceutically relevant literature compounds and more complex AbbVie compounds. Our approach achieves predictive (RMSE = 0.86) and differentiating power (R2 = 0.69) and therefore provides notably improved correlations to experimental solubility compared to state-of-the-art machine learning approaches that utilize quantum mechanics-based descriptors. The importance of explicit considerations of crystalline packing in predicting solubility by the FEP+ approach is also highlighted in this study. Finally, we show how computed energetics, including hydration and sublimation free energies, can provide further insights into molecule design to feed the medicinal chemistry DMTA cycle.
Collapse
Affiliation(s)
- Richard S Hong
- AbbVie Inc., Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Ana V Rojas
- Schrödinger Inc., 1540 Broadway 24th Floor, New York, New York 10036, United States
| | - Rajni Miglani Bhardwaj
- AbbVie Inc., Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Lingle Wang
- Schrödinger Inc., 1540 Broadway 24th Floor, New York, New York 10036, United States
| | - Alessandra Mattei
- AbbVie Inc., Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Nathan S Abraham
- Ventus Therapeutics 100 Beaver St, Waltham, Massachusetts 02453, United States
| | - Kevin P Cusack
- AbbVie Inc., Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - M Olivia Pierce
- Bristol Myer Squibb, 100 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Sayan Mondal
- Schrödinger Inc., 1540 Broadway 24th Floor, New York, New York 10036, United States
| | - Nada Mehio
- AbbVie Inc., Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Shailendra Bordawekar
- AbbVie Inc., Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Philip R Kym
- AbbVie Inc., Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| | - Robert Abel
- Schrödinger Inc., 1540 Broadway 24th Floor, New York, New York 10036, United States
| | - Ahmad Y Sheikh
- AbbVie Inc., Research & Development, 1 N Waukegan Road, North Chicago, Illinois 60064, United States
| |
Collapse
|
2
|
Fan Y, Castleberry S. High-throughput kinetic turbidity analysis for determination of amorphous solubility and excipient screening for amorphous solid dispersions. Int J Pharm 2023; 631:122495. [PMID: 36526147 DOI: 10.1016/j.ijpharm.2022.122495] [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: 10/08/2022] [Revised: 12/06/2022] [Accepted: 12/11/2022] [Indexed: 12/15/2022]
Abstract
Many poorly water-soluble active pharmaceutical ingredients (APIs) rely on supersaturating formulations, such as amorphous solid dispersions (ASDs), to enhance oral bioavailability. ASDs kinetically trap amorphous solid drugs within polymer excipient matrices to maintain the amorphous drug states. The maximum solution concentration of the API in these formulations is known as the amorphous solubility. In early drug development with scarce material and time, high-throughput approaches to measuring amorphous solubility and screening excipient effects on crystallization risk offer significant benefits to preclinical formulation scientists. Here, we developed a high-throughput screening (HTS) workflow to quantify amorphous solubility and screen ASD excipients by automated kinetic turbidity analysis. Testing 20 model APIs with a wide range of biorelevant solubility, we demonstrated their apparent amorphous solubility determined by the HTS approach strongly correlated with quantification results using conventional liquid chromatography; while the real-time analysis significantly saved analytical time and experimental efforts. Furthermore, kinetic turbidity profiles elucidated distinct excipient effects on the precipitation process of APIs. These results were successfully translated to dissolution and precipitation behaviors of ASD formulations composed of the tested polymers. The high-throughput kinetic turbidity workflow presents a facile and information-rich approach for amorphous solubility screenings against excipients, and helps guide enabling formulation development.
Collapse
Affiliation(s)
- Yuchen Fan
- Small Molecule Pharmaceutical Sciences, Research and Early Development, Genentech Inc. 1 DNA Way, South San Francisco, CA 94080, USA
| | - Steven Castleberry
- Small Molecule Pharmaceutical Sciences, Research and Early Development, Genentech Inc. 1 DNA Way, South San Francisco, CA 94080, USA.
| |
Collapse
|
5
|
Thakral NK, Meister E, Jankovsky C, Li L, Schwabe R, Luo L, Chen S. Prediction of in vivo supersaturation and precipitation of poorly water-soluble drugs: Achievements and aspirations. Int J Pharm 2021; 600:120505. [PMID: 33753162 DOI: 10.1016/j.ijpharm.2021.120505] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 03/10/2021] [Accepted: 03/13/2021] [Indexed: 12/14/2022]
Abstract
This review focuses on options available to a pharmaceutical scientist to predict in vivo supersaturation and precipitation of poorly water-soluble drugs. As no single device or system can simulate the complex gastrointestinal environment, a combination of appropriate in vitro tools may be utilized to get optimal predictive information. To address the empirical issues encountered during small-scale and full-scale in vitro predictive testing, theoretical background and relevant case studies are discussed. The practical considerations for selection of appropriate tools at various stages of drug development are recommended. Upcoming technologies that have potential to further reduce in vivo studies and expedite the drug development process are also discussed.
Collapse
Affiliation(s)
- Naveen K Thakral
- Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, United States.
| | - Eva Meister
- Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, United States
| | - Corinne Jankovsky
- Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, United States
| | - Li Li
- Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, United States; Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 4849 Calhoun Road, Houston, TX 77204, United States
| | - Robert Schwabe
- Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, United States
| | - Laibin Luo
- Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, United States
| | - Shirlynn Chen
- Boehringer Ingelheim Pharmaceuticals, Inc., 900 Ridgebury Road, Ridgefield, CT 06877, United States
| |
Collapse
|
6
|
Nagle A, Biggart A, Be C, Srinivas H, Hein A, Caridha D, Sciotti RJ, Pybus B, Kreishman-Deitrick M, Bursulaya B, Lai YH, Gao MY, Liang F, Mathison CJN, Liu X, Yeh V, Smith J, Lerario I, Xie Y, Chianelli D, Gibney M, Berman A, Chen YL, Jiricek J, Davis LC, Liu X, Ballard J, Khare S, Eggimann FK, Luneau A, Groessl T, Shapiro M, Richmond W, Johnson K, Rudewicz PJ, Rao SPS, Thompson C, Tuntland T, Spraggon G, Glynne RJ, Supek F, Wiesmann C, Molteni V. Discovery and Characterization of Clinical Candidate LXE408 as a Kinetoplastid-Selective Proteasome Inhibitor for the Treatment of Leishmaniases. J Med Chem 2020; 63:10773-10781. [PMID: 32667203 PMCID: PMC7549094 DOI: 10.1021/acs.jmedchem.0c00499] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
![]()
Visceral
leishmaniasis is responsible for up to 30,000 deaths every
year. Current treatments have shortcomings that include toxicity and
variable efficacy across endemic regions. Previously, we reported
the discovery of GNF6702, a selective inhibitor of the kinetoplastid
proteasome, which cleared parasites in murine models of leishmaniasis,
Chagas disease, and human African trypanosomiasis. Here, we describe
the discovery and characterization of LXE408, a structurally related
kinetoplastid-selective proteasome inhibitor currently in Phase 1
human clinical trials. Furthermore, we present high-resolution cryo-EM
structures of the Leishmania tarentolae proteasome
in complex with LXE408, which provides a compelling explanation for
the noncompetitive mode of binding of this novel class of inhibitors
of the kinetoplastid proteasome.
Collapse
Affiliation(s)
- Advait Nagle
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Agnes Biggart
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Celine Be
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | - Honnappa Srinivas
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | - Andreas Hein
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | - Diana Caridha
- Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Richard J Sciotti
- Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Brandon Pybus
- Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Mara Kreishman-Deitrick
- Experimental Therapeutics, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, United States
| | - Badry Bursulaya
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Yin H Lai
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Mu-Yun Gao
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Fang Liang
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Casey J N Mathison
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Xiaodong Liu
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Vince Yeh
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Jeffrey Smith
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Isabelle Lerario
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Yongping Xie
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Donatella Chianelli
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Michael Gibney
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Ashley Berman
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Yen-Liang Chen
- Novartis Institute of Tropical Diseases, Emeryville, California 94608, United States
| | - Jan Jiricek
- Novartis Institute of Tropical Diseases, Emeryville, California 94608, United States
| | - Lauren C Davis
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Xianzhong Liu
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Jaime Ballard
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Shilpi Khare
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | | | - Alexandre Luneau
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | - Todd Groessl
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Michael Shapiro
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Wendy Richmond
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Kevin Johnson
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Patrick J Rudewicz
- Novartis Institute of Tropical Diseases, Emeryville, California 94608, United States
| | - Srinivasa P S Rao
- Novartis Institute of Tropical Diseases, Emeryville, California 94608, United States
| | - Christopher Thompson
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Tove Tuntland
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Glen Spraggon
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Richard J Glynne
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | - Frantisek Supek
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| | | | - Valentina Molteni
- Genomics Institute of the Novartis Research Foundation (GNF), San Diego, California 92121, United States
| |
Collapse
|
10
|
Könczöl Á, Dargó G. Brief overview of solubility methods: Recent trends in equilibrium solubility measurement and predictive models. DRUG DISCOVERY TODAY. TECHNOLOGIES 2018; 27:3-10. [PMID: 30103861 DOI: 10.1016/j.ddtec.2018.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 05/31/2018] [Accepted: 06/03/2018] [Indexed: 06/08/2023]
Abstract
Solubility is a crucial physicochemical parameter affecting the whole process of drug discovery and development. Thus, understanding of the methods and concepts to measure and predict this propensity are of utmost importance for the pharmaceutical scientist. Despite their inherent limitations, kinetic solubility screening methods became routine assays by mimicking bioassay conditions and guiding the lead optimization process. In contrast, thermodynamic solubility methods show a clear evolution: miniaturized high throughput assays coupled to analytical techniques such as solid-state characterization, ultra performance liquid chromatography, or polychromatic turbidimetry, have been developed, thereby enabling a more complex physicochemical profiling at the early discovery stage. Solubility prediction still poses a significant challenge at the industrial level. Classification and critical evaluation of recent in silico models are provided. Discussion of experimental and computational methods: was based on relevant industrial references.
Collapse
Affiliation(s)
- Árpád Könczöl
- Compound Profiling Laboratory, Gedeon Richter Plc., H-1475 Budapest, Hungary.
| | - Gergő Dargó
- Compound Profiling Laboratory, Gedeon Richter Plc., H-1475 Budapest, Hungary; Department of Organic Chemistry and Technology, Budapest University of Technology and Economics, Műegyetem rakpart 3, H-1111 Budapest, Hungary
| |
Collapse
|