1
|
Disha IJ, Hasan R, Bhuia S, Ansari SA, Ansari IA, Islam MT. Anxiolytic Efficacy of Indirubin: In Vivo Approach Along with Receptor Binding Profiling and Molecular Interaction with GABAergic Pathways. ChemistryOpen 2024:e202400290. [PMID: 39460441 DOI: 10.1002/open.202400290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/03/2024] [Indexed: 10/28/2024] Open
Abstract
Anxiety is a natural response to stress, characterized by feelings of worry, fear, or unease. The current research was conducted to investigate the anxiolytic effect of indirubin (IND) in different behavioral paradigms in Swiss albino mice. To observe the animal's behavioural response to assess anxiolytic activity, different tests were performed, such as the open-field (square cross, grooming, and rearing), swing, dark-light, and hole cross tests. The experimental mice were administered IND (5 and 10 mg/kg, p.o.), where diazepam (DZP) and vehicle were used as positive and negative controls, respectively. In addition, a combination treatment (DZP+IND-10) was provided to the animals to determine the modulatory effect of IND on DZP. Molecular docking approach was also conducted to determine the binding energy of IND with the GABAA receptor (α2 and α3 subunits) and pharmacokinetics were also estimated. The findings revealed that IND dose-dependently significantly (p<0.05) reduced the animal's movement exerting calming behavior like DZP. IND also demonstrated the highest docking score (-7.7 kcal/mol) against the α3 subunit, while DZP showed a lower docking value (-6.4 kcal/mol) than IND. The ADMET analysis revealed that IND has proper drug-likeness and pharmacokinetic characteristics. In conclusion, IND exerted anxiolytic effects through GABAergic Pathways.
Collapse
Affiliation(s)
- Ishrat Jahan Disha
- Biochemistry and Molecular Biology, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, Dhaka, 8100, Bangladesh
| | - Rubel Hasan
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, Dhaka, 8100, Bangladesh
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Shimul Bhuia
- Bioinformatics and Drug Innovation Laboratory, BioLuster Research Center Ltd., Gopalganj, Dhaka, 8100, Bangladesh
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, Turin, 10124, Italy
| | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, 8100, Bangladesh
- Pharmacy Discipline, Khulna University, Khulna, 9208, Bangladesh
| |
Collapse
|
2
|
Tomlinson L, Ramsden D, Leite SB, Beken S, Bonzo JA, Brown P, Candarlioglu PL, Chan TS, Chen E, Choi CK, David R, Delrue N, Devine PJ, Ford K, Garcia MI, Gosset JR, Hewitt P, Homan K, Irrechukwu O, Kopec AK, Liras JL, Mandlekar S, Raczynski A, Sadrieh N, Sakatis MZ, Siegel J, Sung K, Sunyovszki I, Van Vleet TR, Ekert JE, Hardwick RN. Considerations from an International Regulatory and Pharmaceutical Industry (IQ MPS Affiliate) Workshop on the Standardization of Complex In Vitro Models in Drug Development. Adv Biol (Weinh) 2024; 8:e2300131. [PMID: 37814378 DOI: 10.1002/adbi.202300131] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 08/08/2023] [Indexed: 10/11/2023]
Abstract
In May 2022, there is an International Regulatory and Pharmaceutical Industry (Innovation and Quality [IQ] Microphysiological Systems [MPS] Affiliate) Workshop on the standardization of complex in vitro models (CIVMs) in drug development. This manuscript summarizes the discussions and conclusions of this joint workshop organized and executed by the IQ MPS Affiliate and the United States Food and Drug Administration (FDA). A key objective of the workshop is to facilitate discussions around opportunities and/or needs for standardization of MPS and chart potential pathways to increase model utilization in the context of regulatory decision making. Participation in the workshop included 200 attendees from the FDA, IQ MPS Affiliate, and 26 global regulatory organizations and affiliated parties representing Europe, Japan, and Canada. It is agreed that understanding global perspectives regarding the readiness of CIVM/MPS models for regulatory decision making and potential pathways to gaining acceptance is useful to align on globally. The obstacles are currently too great to develop standards for every context of use (COU). Instead, it is suggested that a more tractable approach may be to think of broadly applicable standards that can be applied regardless of COU and/or organ system. Considerations and next steps for this effort are described.
Collapse
Affiliation(s)
| | | | | | - Sonja Beken
- Federal Agency for Medicines and Health Products, Brussels, 1210, Belgium
| | - Jessica A Bonzo
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Paul Brown
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | | | - Tom S Chan
- Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, 06877, USA
| | - Eugene Chen
- DMPK, Genentech, South San Francisco, CA, 94080, USA
| | - Colin K Choi
- Preclinical Safety, Biogen, Cambridge, MA, 02142, USA
| | - Rhiannon David
- Clinical Pharmacology & Safety Sciences, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Nathalie Delrue
- Organisation for Economic Co-operation and Development, Paris, 75016, France
| | - Patrick J Devine
- Discovery Toxicology, Bristol Myers Squibb, San Diego, CA, 09130, USA
| | - Kevin Ford
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Martha Iveth Garcia
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translational Science, Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | | | - Philip Hewitt
- Chemical and Preclinical Safety, Merck Healthcare KGaA, 64293, Darmstadt, Germany
| | - Kimberly Homan
- Complex in Vitro Systems Group, Genentech, South San Francisco, CA, 94080, USA
| | - Onyi Irrechukwu
- Preclinical Sciences and Translational Safety, Johnson and Johnson Innovation Medicine, Spring House, PA, 19002, USA
| | - Anna K Kopec
- Drug Safety Research & Development, Pfizer Inc., Groton, CT, 06340, USA
| | - Jennifer L Liras
- Pharmacokinetics, Dynamics & Metabolism-Medicine Design, Pfizer, Cambridge, MA, 02139, USA
| | - Sandhya Mandlekar
- Clinical Pharmacology, Genentech, South San Francisco, CA, 94080, USA
| | - Arek Raczynski
- Preclinical Safety Assessment, Vertex Pharmaceuticals Inc., Boston, MA, 02210, USA
| | - Nakissa Sadrieh
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Melanie Z Sakatis
- Non-Clinical Safety, In Vitro/In Vivo Translation, GSK R&D, Ware, SG12 9TJ, UK
| | - Jeffrey Siegel
- Center for Drug Evaluation and Research, Office of New Drugs, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Kyung Sung
- Center for Biologics Evaluation and Research, Office of Cellular Therapy and Human Tissue, Cellular and Tissue Therapy Branch, U.S. Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Ilona Sunyovszki
- Translational Cellular Sciences, Biogen, Cambridge, MA, 02142, USA
| | | | | | | |
Collapse
|
3
|
Wang C, Gu J, Li H, Zhao B, Yu T, Guo CL, Huang M, Jiang W, Ouyang Q. The Discovery of GIT1/β-Pix Inhibitors: Virtual Screening and Biological Evaluation of New Small-molecule Compounds with Anti-invasion Effect in Gastrointestinal Neoplasms. Drug Des Devel Ther 2024; 18:3075-3088. [PMID: 39050797 PMCID: PMC11268723 DOI: 10.2147/dddt.s461609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024] Open
Abstract
Background and Objective GIT1 (G-protein-coupled receptor kinase interacting protein-1) has been found to be highly related with cancer cell invasion and metastasis in many cancer types. β-Pix (p21-activated kinase-interacting exchange factor) is one of the proteins that interact with GIT1. Targeting GIT1/β-Pix complex might be a potential therapeutic strategy for interfering cancer metastasis. However, at present, no well-recognized small-molecule inhibitor targeting GIT1/β-Pix is available. Thus, we aim to discover novel GIT1/β-Pix inhibitors with simple scaffold, high activity and low toxicity to develop new therapeutic strategies to restrain cancer metastasis. Methods GIT1/β-Pix inhibitors were identified from ChemBridge by virtual screening. Briefly, the modeling of GIT1 was performed and the establishment of GIT1/β-Pix binding pocket enabled the virtual screening to identify the inhibitor. In addition, direct binding of the candidate molecules to GIT1 was detected by biolayer interferometry (BLI) to discover the hit compound. Furthermore, the inhibitory effect on invasion of stomach and colon cancer cells in vitro was carried out by the transwell assay and detection of epithelial-mesenchymal transition (EMT)-related proteins. Finally, the binding mode of hit compound to GIT1 was estimated by molecular dynamics simulation to analyze the key amino residues to guide further optimization. Results We selected the top 50 compounds from the ChemBridge library by virtual screening. Then, by skeleton similarity analysis nine compounds were selected for further study. Furthermore, the direct interaction of nine compounds to GIT1 was detected by BLI to obtain the best affinitive compound. Finally, 17302836 was successfully identified (KD = 84.1±2.0 μM). In vitro tests on 17302836 showed significant anti-invasion effect on gastric cancer and colorectal cancer. Conclusion We discovered a new GIT1/β-Pix inhibitor (17302836) against gastrointestinal cancer invasion and metastasis. This study provides a promising candidate for developing new GIT1/β-Pix inhibitors for tumor treatment.
Collapse
Affiliation(s)
- Chenkun Wang
- Department of Pharmacy, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, People’s Republic of China
| | - Jing Gu
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, People’s Republic of China
| | - Hongwei Li
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, People’s Republic of China
| | - Bo Zhao
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, People’s Republic of China
| | - Tao Yu
- Department of Pharmacy, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, People’s Republic of China
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, People’s Republic of China
| | - Chun-Ling Guo
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, People’s Republic of China
| | - Mouxin Huang
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, People’s Republic of China
| | - Weiwei Jiang
- Department of Pharmacy, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, People’s Republic of China
| | - Qin Ouyang
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
4
|
Chaira T, Subramani C, Barman TK. ADME, Pharmacokinetic Scaling, Pharmacodynamic and Prediction of Human Dose and Regimen of Novel Antiviral Drugs. Pharmaceutics 2023; 15:pharmaceutics15041212. [PMID: 37111697 PMCID: PMC10146820 DOI: 10.3390/pharmaceutics15041212] [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: 01/23/2023] [Revised: 03/29/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
The search for new drugs is an extremely time-consuming and expensive endeavour. Much of that time and money go into generating predictive human pharmacokinetic profiles from preclinical efficacy and safety animal data. These pharmacokinetic profiles are used to prioritize or minimize the attrition at later stages of the drug discovery process. In the area of antiviral drug research, these pharmacokinetic profiles are equally important for the optimization, estimation of half-life, determination of effective dose, and dosing regimen, in humans. In this article we have highlighted three important aspects of these profiles. First, the impact of plasma protein binding on two primary pharmacokinetic parameters-volume of distribution and clearance. Second, interdependence of primary parameters on unbound fraction of the drug. Third, the ability to extrapolate human pharmacokinetic parameters and concentration time profiles from animal profiles.
Collapse
Affiliation(s)
- Tridib Chaira
- Department of Pharmacology, SGT University, Gurugram 122505, Haryana, India
| | - Chandru Subramani
- Department of Pathology, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77550, USA
| | - Tarani Kanta Barman
- Department of Pathology, Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77550, USA
| |
Collapse
|
5
|
Tokalı FS, Demir Y, Türkeş C, Dinçer B, Beydemir Ş. Novel acetic acid derivatives containing quinazolin-4(3H)-one ring: Synthesis, in vitro, and in silico evaluation of potent aldose reductase inhibitors. Drug Dev Res 2023; 84:275-295. [PMID: 36598092 DOI: 10.1002/ddr.22031] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 12/01/2022] [Accepted: 12/23/2022] [Indexed: 01/05/2023]
Abstract
Aldose reductase (AR) is a crucial enzyme of the polyol pathway through which glucose is metabolized under conditions of hyperglycemia related to diabetes. A series of novel acetic acid derivatives containing quinazolin-4(3H)-one ring (1-22) was synthesized and tested for in vitro AR inhibitory effect. All the target compounds exhibited nanomolar activity against the target enzyme, and all compounds displayed higher activity as compared to the reference drug epalrestat. Among them, Compound 19, named 2-(4-[(2-[(4-methylpiperazin-1-yl)methyl]-4-oxoquinazolin-3(4H)-ylimino)methyl]phenoxy)acetic acid, displayed the strongest inhibitory effect with a KI value of 61.20 ± 10.18 nM. Additionally, these compounds were investigated for activity against L929, nontumoral fibroblast cells, and MCF-7, breast cancer cells using the MTT assay. Compounds 16 and 19 showed lower toxicity against the normal L929 cells. The synthesized compounds' (1-22) absorption, distribution, metabolism, and excretion properties were also evaluated. Molecular docking simulations were used to look into the possible binding mechanisms of these inhibitors against AR.
Collapse
Affiliation(s)
- Feyzi Sinan Tokalı
- Department of Material and Material Processing Technologies, Kars Vocational School, Kafkas University, Kars, Turkey
| | - Yeliz Demir
- Department of Pharmacy Services, Nihat Delibalta Göle Vocational High School, Ardahan University, Ardahan, Turkey
| | - Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Büşra Dinçer
- Department of Pharmacology, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan, Turkey
| | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
- The Rectorate of Bilecik Şeyh Edebali University, Bilecik, Turkey
| |
Collapse
|
6
|
Gu J, Peng RK, Guo CL, Zhang M, Yang J, Yan X, Zhou Q, Li H, Wang N, Zhu J, Ouyang Q. Construction of a synthetic methodology-based library and its application in identifying a GIT/PIX protein-protein interaction inhibitor. Nat Commun 2022; 13:7176. [PMID: 36418900 PMCID: PMC9684509 DOI: 10.1038/s41467-022-34598-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 10/24/2022] [Indexed: 11/24/2022] Open
Abstract
In recent years, the flourishing of synthetic methodology studies has provided concise access to numerous molecules with new chemical space. These compounds form a large library with unique scaffolds, but their application in hit discovery is not systematically evaluated. In this work, we establish a synthetic methodology-based compound library (SMBL), integrated with compounds obtained from our synthetic researches, as well as their virtual derivatives in significantly larger scale. We screen the library and identify small-molecule inhibitors to interrupt the protein-protein interaction (PPI) of GIT1/β-Pix complex, an unrevealed target involved in gastric cancer metastasis. The inhibitor 14-5-18 with a spiro[bicyclo[2.2.1]heptane-2,3'-indolin]-2'-one scaffold, considerably retards gastric cancer metastasis in vitro and in vivo. Since the PPI targets are considered undruggable as they are hard to target, the successful application illustrates the structural specificity of SMBL, demonstrating its potential to be utilized as compound source for more challenging targets.
Collapse
Affiliation(s)
- Jing Gu
- grid.410570.70000 0004 1760 6682Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, 400038 Chongqing, China
| | - Rui-Kun Peng
- grid.410570.70000 0004 1760 6682Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, 400038 Chongqing, China
| | - Chun-Ling Guo
- grid.410570.70000 0004 1760 6682Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, 400038 Chongqing, China
| | - Meng Zhang
- grid.16821.3c0000 0004 0368 8293Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Yang
- grid.410570.70000 0004 1760 6682Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, 400038 Chongqing, China
| | - Xiao Yan
- grid.410570.70000 0004 1760 6682Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, 400038 Chongqing, China
| | - Qian Zhou
- grid.410570.70000 0004 1760 6682Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, 400038 Chongqing, China
| | - Hongwei Li
- grid.410570.70000 0004 1760 6682Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, 400038 Chongqing, China
| | - Na Wang
- grid.410570.70000 0004 1760 6682Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, 400038 Chongqing, China
| | - Jinwei Zhu
- grid.16821.3c0000 0004 0368 8293Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, China
| | - Qin Ouyang
- grid.410570.70000 0004 1760 6682Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, 400038 Chongqing, China
| |
Collapse
|
7
|
Singh MB, Vishvakarma VK, Lal AA, Chandra R, Jain P, Singh P. A comparative study of 5- fluorouracil, doxorubicin, methotrexate, paclitaxel for their inhibition ability for Mpro of nCoV: Molecular docking and molecular dynamics simulations. J INDIAN CHEM SOC 2022. [PMCID: PMC9632266 DOI: 10.1016/j.jics.2022.100790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
A new corona virus (nCoV) is aetiological agent responsible for the viral pneumonia epidemic. Three is no specific therapeutic medicines available for the treatment of this condition and also effective treatment choices are few. In this work author tried to investigate some repurposing drug such as 5- fluorouracil, doxorubicin, methotrexate and paclitaxel against the main protease (Mpro) of nCoV by the computational model. Molecular docking was performed to screen out the best compound and doxorubicin was found to have minimum binding energy −121.89 kcal/mol. To further study, MD simulations were performed at 300 K and the result successfully corroborate the energy obtained by molecular docking. Temperature dependent MD simulation of the best molecule that is doxorubicin obtained from docking result was performed to check the variation in structural changes in Mpro of nCoV at 290 K, 310 K, 320 K and 325 K. It is sound that doxorubicin binds effectively with Mpro of nCoV at 290 K. Further ADME properties of the 5- fluorouracil, doxorubicin, methotrexate and paclitaxel were also evaluated to understand the bioavailability.
Collapse
|
8
|
Doolan JA, Williams GT, Hilton KLF, Chaudhari R, Fossey JS, Goult BT, Hiscock JR. Advancements in antimicrobial nanoscale materials and self-assembling systems. Chem Soc Rev 2022; 51:8696-8755. [PMID: 36190355 PMCID: PMC9575517 DOI: 10.1039/d1cs00915j] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Indexed: 11/21/2022]
Abstract
Antimicrobial resistance is directly responsible for more deaths per year than either HIV/AIDS or malaria and is predicted to incur a cumulative societal financial burden of at least $100 trillion between 2014 and 2050. Already heralded as one of the greatest threats to human health, the onset of the coronavirus pandemic has accelerated the prevalence of antimicrobial resistant bacterial infections due to factors including increased global antibiotic/antimicrobial use. Thus an urgent need for novel therapeutics to combat what some have termed the 'silent pandemic' is evident. This review acts as a repository of research and an overview of the novel therapeutic strategies being developed to overcome antimicrobial resistance, with a focus on self-assembling systems and nanoscale materials. The fundamental mechanisms of action, as well as the key advantages and disadvantages of each system are discussed, and attention is drawn to key examples within each field. As a result, this review provides a guide to the further design and development of antimicrobial systems, and outlines the interdisciplinary techniques required to translate this fundamental research towards the clinic.
Collapse
Affiliation(s)
- Jack A Doolan
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK.
| | - George T Williams
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Kira L F Hilton
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| | - Rajas Chaudhari
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| | - John S Fossey
- School of Chemistry, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK.
| | - Benjamin T Goult
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, UK.
| | - Jennifer R Hiscock
- School of Chemistry and Forensic Science, University of Kent, Canterbury, Kent CT2 7NH, UK.
| |
Collapse
|
9
|
Xiong F, Yu M, Xu H, Zhong Z, Li Z, Guo Y, Zhang T, Zeng Z, Jin F, He X. Discovery of TIGIT inhibitors based on DEL and machine learning. Front Chem 2022; 10:982539. [PMID: 35958238 PMCID: PMC9360614 DOI: 10.3389/fchem.2022.982539] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Drug discovery has entered a new period of vigorous development with advanced technologies such as DNA-encoded library (DEL) and artificial intelligence (AI). The previous DEL-AI combination has been successfully applied in the drug discovery of classical kinase and receptor targets mainly based on the known scaffold. So far, there is no report of the DEL-AI combination on inhibitors targeting protein-protein interaction, including those undruggable targets with few or unknown active scaffolds. Here, we applied DEL technology on the T cell immunoglobulin and ITIM domain (TIGIT) target, resulting in the unique hit compound 1 (IC50 = 20.7 μM). Based on the screening data from DEL and hit derivatives a1-a34, a machine learning (ML) modeling process was established to address the challenge of poor sample distribution uniformity, which is also frequently encountered in DEL screening on new targets. In the end, the established ML model achieved a satisfactory hit rate of about 75% for derivatives in a high-scored area.
Collapse
Affiliation(s)
- Feng Xiong
- Shenzhen Innovation Center for Small Molecule Drug Discovery Co., Ltd., Shenzhen, China
- *Correspondence: Feng Xiong, ; Feng Jin, ; Xun He,
| | - Mingao Yu
- Shenzhen NewDEL Biotech Co., Ltd., Shenzhen, China
| | - Honggui Xu
- Shenzhen NewDEL Biotech Co., Ltd., Shenzhen, China
| | - Zhenmin Zhong
- Shenzhen Innovation Center for Small Molecule Drug Discovery Co., Ltd., Shenzhen, China
| | - Zhenwei Li
- Shenzhen Innovation Center for Small Molecule Drug Discovery Co., Ltd., Shenzhen, China
| | - Yuhan Guo
- Shenzhen NewDEL Biotech Co., Ltd., Shenzhen, China
| | | | - Zhixuan Zeng
- Shenzhen Innovation Center for Small Molecule Drug Discovery Co., Ltd., Shenzhen, China
| | - Feng Jin
- Shenzhen NewDEL Biotech Co., Ltd., Shenzhen, China
- *Correspondence: Feng Xiong, ; Feng Jin, ; Xun He,
| | - Xun He
- Shenzhen Innovation Center for Small Molecule Drug Discovery Co., Ltd., Shenzhen, China
- *Correspondence: Feng Xiong, ; Feng Jin, ; Xun He,
| |
Collapse
|
10
|
An approach for identifying in silico peptides against authentic metabolites: in vitro characterization of thymosin β4 metabolites. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2022. [DOI: 10.1007/s40005-022-00581-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
11
|
Griffin RJ, Avery E, Xia CQ. Predicting Approximate Clinically Effective Doses in Oncology Using Preclinical Efficacy and Body Surface Area Conversion: A Retrospective Analysis. Front Pharmacol 2022; 13:830972. [PMID: 35559235 PMCID: PMC9087189 DOI: 10.3389/fphar.2022.830972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/16/2022] [Indexed: 11/30/2022] Open
Abstract
The correlation between efficacious doses in human tumor-xenograft mouse models and the human clinical doses of approved oncology agents was assessed using published preclinical data and recommended clinical doses. For 90 approved small molecule anti-cancer drugs, body surface area (BSA) corrected mouse efficacious doses were strongly predictive of human clinical dose ranges with 85.6% of the predictions falling within three-fold (3×) of the recommended clinical doses and 63.3% within 2×. These results suggest that BSA conversion is a useful tool for estimating human doses of small molecule oncology agents from mouse xenograft models from the early discovery stage. However, the BSA based dose conversion poorly predicts for the intravenous antibody and antibody drug conjugate anti-cancer drugs. For antibody-based drugs, five out of 30 (16.7%) predicted doses were within 3× of the recommended clinical dose. The body weight-based dose projection was modestly predictive with 66.7% of drugs predicted within 3× of the recommended clinical dose. The correlation was slightly better in ADCs (77.7% in 3×). The application and limitations of such simple dose estimation methods in the early discovery stage and in the design of clinical trials are also discussed in this retrospective analysis.
Collapse
Affiliation(s)
| | - Ethan Avery
- Takeda Pharmaceuticals, Cambridge, MA, United States
| | - Cindy Q Xia
- Takeda Pharmaceuticals, Cambridge, MA, United States
| |
Collapse
|
12
|
In Silico study for acyclovir, ganciclovir and its derivatives to fight the COVID-19: Molecular docking, DFT calculations, ADME and td-Molecular dynamics simulations. J INDIAN CHEM SOC 2022. [PMCID: PMC8931996 DOI: 10.1016/j.jics.2022.100433] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In the present work, we have designed three molecules, acyclovir (A), ganciclovir (G) and derivative of hydroxymethyl derivative of ganciclovir (CH2OH of G, that is D) and investigated their biological potential against the Mpro of nCoV via in silico studies. Further, density functional theory (DFT) calculations of A, G and D were performed using Gaussian 16 on applying B3LYP under default condition to collect the information for the delocalization of electron density in their optimized geometry. Authors have also calculated various energies including free energy of A, G and D in Hartree per particle. It can be seen that D has the least free energy. As mentioned, the molecular docking of the A, G and D against the Mpro of nCoV was performed using iGemdock, an acceptable computational tool and the interaction has been studied in the form of physical data, that is, binding energy for A, G and D were calculated in kcal/mol. It can be seen the D showed effective binding, that is, maximum inhibition that A and G. For a better understanding for the inhibition of the Mpro of nCoV by A, G and D, temperature dependent molecular dynamics simulations were performed. Different trajectories like RMSD, RMSF, Rg and hydrogen bond were extracted and analyzed. The results of molecular docking of A, G and D corroborate with the td-MD simulations and hypothesized that D could be a promising candidate to inhibit the activity of Mpro of nCoV.
Collapse
|
13
|
Ball K, Bruin G, Escandon E, Funk C, Pereira JN, Yang TY, Yu H. Characterizing the pharmacokinetics and biodistribution of therapeutic proteins: an industry white paper. Drug Metab Dispos 2022; 50:858-866. [PMID: 35149542 DOI: 10.1124/dmd.121.000463] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/06/2022] [Indexed: 11/22/2022] Open
Abstract
Characterization of the pharmacokinetics (PK) and biodistribution of therapeutic proteins (TPs) is a hot topic within the pharmaceutical industry, particularly with an ever-increasing catalog of novel modality TPs. Here, we review the current practices, and provide a summary of extensive cross-company discussions as well as a survey completed by International Consortium for Innovation and Quality (IQ consortium) members on this theme. A wide variety of in vitro, in vivo and in silico techniques are currently used to assess PK and biodistribution of TPs, and we discuss the relevance of these from an industry perspective, focusing on PK/PD understanding at the preclinical stage of development, and translation to human. We consider that the 'traditional in vivo biodistribution study' is becoming insufficient as a standalone tool, and thorough characterization of the interaction of the TP with its target(s), target biology, and off-target interactions at a microscopic scale are key to understand the overall biodistribution at a full-body scale. Our summary of the current challenges and our recommendations to address these issues could provide insight into the implementation of best practices in this area of drug development, and continued cross-company collaboration will be of tremendous value. Significance Statement The Innovation & Quality Consortium (IQ) Translational and ADME Sciences Leadership Group (TALG) working group for the ADME of therapeutic proteins evaluates the current practices, recent advances, and challenges in characterizing the PK and biodistribution of therapeutic proteins during drug development, and proposes recommendations to address these issues. Incorporating the in vitro, in vivo and in silico approaches discussed herein may provide a pragmatic framework to increase early understanding of PK/PD relationships, and aid translational modelling for first-in-human dose predictions.
Collapse
Affiliation(s)
| | - Gerard Bruin
- Novartis Institutes for Biomedical Research, Switzerland
| | | | - Christoph Funk
- Dept. of Drug Metabolism and Pharmacokinetics, F. Hoffmann-La Roche Ltd., Switzerland
| | | | | | - Hongbin Yu
- Boehringer Ingelheim Pharmaceuticals, Inc, United States
| |
Collapse
|
14
|
Anti-Angiogenic Property of Free Human Oligosaccharides. Biomolecules 2021; 11:biom11060775. [PMID: 34064180 PMCID: PMC8224327 DOI: 10.3390/biom11060775] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 12/29/2022] Open
Abstract
Angiogenesis, a fundamental process in human physiology and pathology, has attracted considerable attention owing to its potential as a therapeutic strategy. Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) are deemed major mediators of angiogenesis. To date, inhibition of the VEGF-A/VEGFR-2 axis has been an effective strategy employed in the development of anticancer drugs. However, some limitations, such as low efficacy and side effects, need to be addressed. Several drug candidates have been discovered, including small molecule compounds, recombinant proteins, and oligosaccharides. In this review, we focus on human oligosaccharides as modulators of angiogenesis. In particular, sialylated human milk oligosaccharides (HMOs) play a significant role in the inhibition of VEGFR-2-mediated angiogenesis. We discuss the structural features concerning the interaction between sialylated HMOs and VEGFR-2 as a molecular mechanism of anti-angiogenesis modulation and its effectiveness in vivo experiments. In the current state, extensive clinical trials are required to develop a novel VEGFR-2 inhibitor from sialylated HMOs.
Collapse
|
15
|
Rogers H, Adeniyi O, Ramamoorthy A, Bailey S, Pacanowski M. Clinical Pharmacology Studies Supporting Oligonucleotide Therapy Development: An Assessment of Therapies Approved and in Development Between 2012 and 2018. Clin Transl Sci 2021; 14:468-475. [PMID: 33278337 PMCID: PMC7993268 DOI: 10.1111/cts.12945] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 11/18/2020] [Indexed: 11/26/2022] Open
Abstract
Synthetic nucleotides that utilize RNA-centric pharmacology can target diseases at the RNA level, thus altering protein expression in ways previously inaccessible to small molecules and therapeutic biologics. Recognizing that the unique pharmacology of oligonucleotides may require specific considerations in pre-approval assessment, clinical and nonclinical pharmacology studies being conducted for a selected set of oligonucleotide therapies in a 6-year period were assessed. This investigation focused primarily on the four following areas: (i) drug-drug interaction (DDI) potential, (ii) organ impairment (i.e., renal and hepatic impairment), (iii) immunogenicity, and (iv) cardiac safety. Data were summarized and assessed from 14 Investigational New Drug programs and 7 New Drug Applications submitted to the US Food and Drug Administration (FDA) from the period of January 2012 to August 2018, encompassing 152 unique studies. The assessment of DDI potential was largely consistent with the recommendations of current DDI-relevant guidances. Limited data were available to provide recommendations across organ impairment categories. Limited data on immunogenicity indicate impact on pharmacokinetic, the impact on safety and efficacy, although not extensively evaluated, appeared negligible. Cardiac safety evaluation indicated a potential for discordant translation of risk from nonclinical studies to clinical findings. Continued experience with synthetic oligonucleotide therapies will help inform the development of best practices to support their development and regulatory approval.
Collapse
Affiliation(s)
- Hobart Rogers
- Center for Drug Evaluation and ResearchOffice of Translational SciencesOffice of Clinical PharmacologyUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Oluseyi Adeniyi
- Center for Drug Evaluation and ResearchOffice of Translational SciencesOffice of Clinical PharmacologyUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Anuradha Ramamoorthy
- Center for Drug Evaluation and ResearchOffice of Translational SciencesOffice of Clinical PharmacologyUS Food and Drug AdministrationSilver SpringMarylandUSA
| | - Samantha Bailey
- School of PharmacyUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Michael Pacanowski
- Center for Drug Evaluation and ResearchOffice of Translational SciencesOffice of Clinical PharmacologyUS Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
16
|
Pearman NA, Ronander E, Smith AM, Morris GA. The identification and characterisation of novel bioactive peptides derived from porcine liver. Curr Res Food Sci 2020; 3:314-321. [PMID: 33336193 PMCID: PMC7733001 DOI: 10.1016/j.crfs.2020.11.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Bioactive peptides (BAPs) can be derived from a variety of sources; these could be from dietary proteins which are then broken down in the gastrointestinal tract to release BAPs, or they can be isolated from various sources ex vivo. Sources include plant-based proteins such as soy, and chickpeas, and animal proteins from waste from the meat industry and from fish skin. Bioinformatics is also a useful approach to assess the peptides released from digests due to the great number of possible sequences that can be isolated from proteins. Therefore, an in silico analysis of peptides could potentially lead to a more rapid discovery of BAPs. This article investigates a "crude" liver peptide mixture derived from papain hydrolysis of porcine liver and purified peptides derived from the hydrolysates following HPLC fractionation and in silico digestion of the host proteins identified using LC-MS/MS. This allowed the identification of two proteins (cytosol aminopeptidase and haemoglobin subunit alpha) present in the "crude" mixture after LC-MS/MS. In silico hydrolysis of these proteins identified that several peptides were predicted to be both present in the crude mixture using the BIOPEP database and to have potential bioactivity using the Peptide Ranker tool. Peptides (FWG, MFLG and SDPPLVFVG) with the greatest potential bioactivity and which had not previously been reported in the literature were then synthesised. The results indicated that the predicted bioactivity of the synthetic peptides would likely include antioxidant activity. FWG and MFLG derived from the in silico papain hydrolysis of cytosol aminopeptidase showed activity better or comparable to Trolox in the Oxygen Radical Absorbance Capacity (ORAC) assay. The use of these in silico tools, alongside a robust range of biochemical assays which cover a wider range of bioactivities would be a way of improving the discovery of novel bioactive peptides.
Collapse
Affiliation(s)
- Nicholas A. Pearman
- Department of Chemical Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, HD1 3DH, UK
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, HD1 3DH, UK
| | - Elena Ronander
- Biofac A/S, Englandsvej 350-356, DK-2770, Kastrup, Denmark
| | - Alan M. Smith
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Huddersfield, HD1 3DH, UK
| | - Gordon A. Morris
- Department of Chemical Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, HD1 3DH, UK
| |
Collapse
|
17
|
Yu X, Fridman A, Bagchi A, Xu S, Kwasnjuk KA, Lu P, Cancilla MT. Metabolite Identification of Therapeutic Peptides and Proteins by Top-down Differential Mass Spectrometry and Metabolite Database Matching. Anal Chem 2020; 92:8298-8305. [PMID: 32402188 DOI: 10.1021/acs.analchem.0c00652] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
As metabolism impacts the efficacy and safety of therapeutic peptides and proteins (TPPs), understanding of the metabolic fate of TPPs is critical for their preclinical and clinical development. Despite the continued increase of new TPPs entering clinical trials, the metabolite identification (MetID) of these emerging modalities remains challenging. In the present study, we report an analytical workflow for MetID of TPPs. Using insulin detemir as an example, we demonstrated that top-down differential mass spectrometry (dMS) was able to distinguish and discover metabolites from complex biological matrices. For structural interpretation, we developed an algorithm to generate a complete and nonredundant theoretical metabolite database for a TPP of any topology (e.g., branched, multicyclic, etc.). Candidate structures of a metabolite were obtained by matching the monoisotopic mass of a dMS feature to the theoretical metabolite database. Finally, the MS/MS sequence tags enabled unambiguous characterization of metabolite structures when isobaric/isomeric candidates were present. This platform is widely applicable to TPPs with complex structures and will ultimately guide the structural optimization of TPPs in pharmaceutical development.
Collapse
|
18
|
Shou WZ. Current status and future directions of high-throughput ADME screening in drug discovery. J Pharm Anal 2020; 10:201-208. [PMID: 32612866 PMCID: PMC7322755 DOI: 10.1016/j.jpha.2020.05.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023] Open
Abstract
During the last decade high-throughput in vitro absorption, distribution, metabolism and excretion (HT-ADME) screening has become an essential part of any drug discovery effort of synthetic molecules. The conduct of HT-ADME screening has been "industrialized" due to the extensive development of software and automation tools in cell culture, assay incubation, sample analysis and data analysis. The HT-ADME assay portfolio continues to expand in emerging areas such as drug-transporter interactions, early soft spot identification, and ADME screening of peptide drug candidates. Additionally, thanks to the very large and high-quality HT-ADME data sets available in many biopharma companies, in silico prediction of ADME properties using machine learning has also gained much momentum in recent years. In this review, we discuss the current state-of-the-art practices in HT-ADME screening including assay portfolio, assay automation, sample analysis, data processing, and prediction model building. In addition, we also offer perspectives in future development of this exciting field.
Collapse
Affiliation(s)
- Wilson Z. Shou
- Bristol-Myers Squibb, PO Box 4000, Princeton, NJ, 08540, USA
| |
Collapse
|
19
|
Ligand-based pharmacophore filtering, atom based 3D-QSAR, virtual screening and ADME studies for the discovery of potential ck2 inhibitors. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2019.127670] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
20
|
Chahar DS, Ravindran S, Pisal S. Monoclonal antibody purification and its progression to commercial scale. Biologicals 2020; 63:1-13. [DOI: 10.1016/j.biologicals.2019.09.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/24/2019] [Accepted: 09/17/2019] [Indexed: 11/17/2022] Open
|
21
|
Chung TW, Kim EY, Choi HJ, Han CW, Jang SB, Kim KJ, Jin L, Koh YJ, Ha KT. 6'-Sialylgalactose inhibits vascular endothelial growth factor receptor 2-mediated angiogenesis. Exp Mol Med 2019; 51:1-13. [PMID: 31604908 PMCID: PMC6802645 DOI: 10.1038/s12276-019-0311-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 05/03/2019] [Accepted: 06/03/2019] [Indexed: 01/29/2023] Open
Abstract
Angiogenesis should be precisely regulated because disordered neovascularization is involved in the aggravation of multiple diseases. The vascular endothelial growth factor (VEGF)-A/VEGF receptor 2 (VEGFR-2) axis is crucial for controlling angiogenic responses in vascular endothelial cells (ECs). Therefore, inactivating VEGFR-2 signaling may effectively suppress aberrant angiogenesis and alleviate related symptoms. In this study, we performed virtual screening, identified the synthetic disaccharide 6′-sialylgalactose (6SG) as a potent VEGFR-2-binding compound and verified its high binding affinity by Biacore assay. 6SG effectively suppressed VEGF-A-induced VEGFR-2 phosphorylation and subsequent in vitro angiogenesis in HUVECs without inducing cytotoxicity. 6SG also inhibited VEGF-A-induced extracellular-regulated kinase (ERK)/Akt activation and actin stress fiber formation in HUVECs. We demonstrated that 6SG inhibited retinal angiogenesis in a mouse model of retinopathy of prematurity and tumor angiogenesis in a xenograft mouse model. Our results suggest a potential therapeutic benefit of 6SG in inhibiting angiogenesis in proangiogenic diseases, such as retinopathy and cancer. Therapy based on a synthetic molecule can block abnormal blood vessel formation, limiting the progression of diabetic eye conditions and tumor growth in mice. The growth of new blood vessels from existing vessels, called angiogenesis, is critical to wound healing and embryonic development. The main angiogenesis signalling pathway involves growth factors, including one called VEGFR-2. Disruption to this pathway plays a significant part in the development of multiple diseases. A South Korean team led by Ki-Tae Ha at Pusan National University, Yangsan, and Young Jun Koh at Dongguk University, Seoul, identified and trialed a synthetic disaccharide capable of binding to and limiting the activity of VEGFR-2 during faulty signaling. Trials on mice with the diabetic eye condition retinopathy, and mice with implanted tumors, showed that the compound inhibited excessive angiogenesis and limited disease progression.
Collapse
Affiliation(s)
- Tae-Wook Chung
- Department of Korean Medical Science, School of Korean Medicine and Healthy Aging Korean Medical Research Center, Pusan National University, Yangsan, Gyeongnam, 50612, Korea
| | - Eun-Yeong Kim
- Department of Korean Medical Science, School of Korean Medicine and Healthy Aging Korean Medical Research Center, Pusan National University, Yangsan, Gyeongnam, 50612, Korea
| | - Hee-Jung Choi
- Department of Korean Medical Science, School of Korean Medicine and Healthy Aging Korean Medical Research Center, Pusan National University, Yangsan, Gyeongnam, 50612, Korea
| | - Chang Woo Han
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Geumjeong-gu, Busan, 46241, Korea
| | - Se Bok Jang
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Geumjeong-gu, Busan, 46241, Korea
| | - Keuk-Jun Kim
- Department of Clinical Pathology, TaeKyeung University, Gyeongsan, Gyeongbuk, 38547, Korea
| | - Ling Jin
- Department of Korean Medical Science, School of Korean Medicine and Healthy Aging Korean Medical Research Center, Pusan National University, Yangsan, Gyeongnam, 50612, Korea
| | - Young Jun Koh
- Department of Pathology, College of Korean Medicine, Dongguk University, Goyang, Gyeonggi-do, 10326, Korea. .,GI Innovation, Inc., A-1116, Tera Tower, Songpa-daero 167, Songpa-gu, Seoul, 05855, Korea.
| | - Ki-Tae Ha
- Department of Korean Medical Science, School of Korean Medicine and Healthy Aging Korean Medical Research Center, Pusan National University, Yangsan, Gyeongnam, 50612, Korea.
| |
Collapse
|
22
|
Vishvakarma VK, Singh P, Kumar V, Kumari K, Patel R, Chandra R. Pyrrolothiazolones as Potential Inhibitors for the nsP2B‐nsP3 Protease of Dengue Virus and Their Mechanism of Synthesis. ChemistrySelect 2019. [DOI: 10.1002/slct.201901119] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Vijay Kumar Vishvakarma
- Department of ChemistryA.R.S.D. CollegeUniversity of Delhi, Delhi India
- Department of ChemistryUniversity of Delhi, Delhi India
| | - Prashant Singh
- Department of ChemistryA.R.S.D. CollegeUniversity of Delhi, Delhi India
| | - Vinod Kumar
- Department of ChemistryKirori Mal College, University of Delhi India
| | - Kamlesh Kumari
- Department of ZoologyDDU College, University of Delhi, Delhi India
| | | | | |
Collapse
|
23
|
Logotheti S, Valmas A, Trampari S, Fili S, Saslis S, Spiliopoulou M, Beckers D, Degen T, Nénert G, Fitch AN, Karavassili F, Margiolaki I. Unit-cell response of tetragonal hen egg white lysozyme upon controlled relative humidity variation. J Appl Crystallogr 2019. [DOI: 10.1107/s1600576719009919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Variation of relative humidity (rH) greatly affects the internal order of solvent-based protein crystals, and the rearrangement of molecules can be efficiently recorded in distinct diffraction patterns. This study focuses on this topic, reporting the effect of rH variation experiments on hen egg white lysozyme (HEWL) polycrystalline precipitates of tetragonal symmetry using X-ray powder diffraction (XRPD). In situ XRPD data were collected on HEWL specimens during dehydration and rehydration processes using laboratory instrumentation. A known polymorph [space group P43212, a = 79.07181 (1), c = 38.0776 (1) Å] was identified during gradual dehydration from 95 to 63% rH and vice versa. Pawley analysis of collected data sets and accurate extraction of unit-cell parameters indicated a characteristic evolution of the tetragonal axes with rH. In addition, there is a low humidity level below which samples do not retain their crystallinity. This work illustrates the accuracy of laboratory XRPD as a probe for time-resolved studies of proteins and in situ investigations of gradual structural modifications upon rH variation. These experiments provide essential information for improving production and post-production practices of microcrystalline protein-based pharmaceuticals.
Collapse
|
24
|
Bathula R, Lanka G, Muddagoni N, Dasari M, Nakkala S, Bhargavi M, Somadi G, Sivan SK, Rajender Potlapally S. Identification of potential Aurora kinase-C protein inhibitors: an amalgamation of energy minimization, virtual screening, prime MMGBSA and AutoDock. J Biomol Struct Dyn 2019; 38:2314-2325. [DOI: 10.1080/07391102.2019.1630318] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Revanth Bathula
- Molecular Modeling Laboratory, Department of Chemistry, Nizam College, Osmania University, Hyderabad, India
| | - Goverdhan Lanka
- Molecular Modeling Laboratory, Department of Chemistry, Nizam College, Osmania University, Hyderabad, India
| | - Narasimha Muddagoni
- Molecular Modeling Laboratory, Department of Chemistry, Nizam College, Osmania University, Hyderabad, India
| | - Mahendar Dasari
- Molecular Modeling Laboratory, Department of Chemistry, Nizam College, Osmania University, Hyderabad, India
| | - Sravanthi Nakkala
- Molecular Modeling Laboratory, Department of Chemistry, Nizam College, Osmania University, Hyderabad, India
| | - Manan Bhargavi
- Molecular Modeling Laboratory, Department of Chemistry, Nizam College, Osmania University, Hyderabad, India
| | - Gururaj Somadi
- Molecular Modeling Laboratory, Department of Chemistry, Nizam College, Osmania University, Hyderabad, India
| | - Sree Kanth Sivan
- Molecular Modeling Laboratory, Department of Chemistry, Nizam College, Osmania University, Hyderabad, India
| | - Sarita Rajender Potlapally
- Molecular Modeling Laboratory, Department of Chemistry, Nizam College, Osmania University, Hyderabad, India
| |
Collapse
|
25
|
Fili S, Valmas A, Spiliopoulou M, Kontou P, Fitch A, Beckers D, Degen T, Barlos K, Barlos KK, Karavassili F, Margiolaki I. Revisiting the structure of a synthetic somatostatin analogue for peptide drug design. ACTA CRYSTALLOGRAPHICA SECTION B-STRUCTURAL SCIENCE CRYSTAL ENGINEERING AND MATERIALS 2019; 75:611-620. [DOI: 10.1107/s2052520619006012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 04/30/2019] [Indexed: 11/10/2022]
Abstract
Natural or artificially manufactured peptides attract scientific interest worldwide owing to their wide array of pharmaceutical and biological activities. X-ray structural studies are used to provide a precise extraction of information, which can be used to enable a better understanding of the function and physicochemical characteristics of peptides. Although it is vulnerable to disassociation, one of the most vital human peptide hormones, somatostatin, plays a regulatory role in the endocrine system as well as in the release of numerous secondary hormones. This study reports the successful crystallization and complete structural model of octreotide, a stable octapeptide analogue of somatostatin. Common obstacles in crystallographic studies arising from the intrinsic difficulties of obtaining a suitable single-crystal specimen were efficiently overcome as polycrystalline material was employed for synchrotron and laboratory X-ray powder diffraction (XPD) measurements. Data collection and preliminary analysis led to the identification of unit-cell symmetry [orthorhombic, P212121, a = 18.5453 (15), b = 30.1766 (25), c = 39.798 (4) Å], a process which was later followed by complete structure characterization and refinement, underlying the efficacy of the suggested (XPD) approach.
Collapse
|
26
|
Datta-Mannan A. Mechanisms Influencing the Pharmacokinetics and Disposition of Monoclonal Antibodies and Peptides. Drug Metab Dispos 2019; 47:1100-1110. [PMID: 31043438 DOI: 10.1124/dmd.119.086488] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/22/2019] [Indexed: 12/15/2022] Open
Abstract
Monoclonal antibodies (mAbs) and peptides are an important class of therapeutic modalities that have brought improved health outcomes in areas with limited therapeutic optionality. Presently, there more than 90 mAb and peptide therapeutics on the United States market, with over 600 more in various clinical stages of development in a broad array of therapeutic areas, including diabetes, autoimmune disorders, oncology, neuroscience, and cardiovascular and infectious diseases. Notwithstanding this potential, there is high clinical rate of attrition, with approximately 10% reaching patients. A major contributor to the failure of the molecules is often times an incomplete or poor understanding of the pharmacokinetics (PK) and disposition profiles leading to limited or diminished efficacy. Increased and thorough characterization efforts directed at disseminating mechanisms influencing the PK and disposition of mAbs and peptides can aid in improving the design for their intended pharmacological activity, and thereby their clinical success. The PK and disposition factors for mAbs and peptides are broadly influenced by target-mediated drug disposition and nontarget-related clearance mechanisms related to the interplay between the relationship of the structure and physiochemical properties of mAbs and peptides with physiologic processes. This review focuses on nontarget-related factors influencing the disposition and PK of mAbs and peptides. Contemporary considerations around the increasing in silico approaches to identify nontarget-related molecule limitations and enhancing the druggability of mAbs and peptides, including parenteral and nonparenteral delivery strategies that are geared toward improving patient experience and compliance, are also discussed.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Department of Experimental Medicine and Pharmacology, Lilly Research Laboratories, Lilly Corporate Center, Indianapolis, Indiana
| |
Collapse
|
27
|
Trampari S, Valmas A, Logotheti S, Saslis S, Fili S, Spiliopoulou M, Beckers D, Degen T, Nénert G, Fitch AN, Calamiotou M, Karavassili F, Margiolaki I. In situ detection of a novel lysozyme monoclinic crystal form upon controlled relative humidity variation. J Appl Crystallogr 2018. [DOI: 10.1107/s1600576718013936] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
The effect of relative humidity (rH) on protein crystal structures, an area that has attracted high scientific interest during the past decade, is investigated in this study on hen egg-white lysozyme (HEWL) polycrystalline precipitates via in situ laboratory X-ray powder diffraction (XRPD) measurements. For this purpose, HEWL was crystallized at room temperature and pH 4.5, leading to a novel monoclinic HEWL phase which, to our knowledge, has not been reported before. Analysis of XRPD data collected upon rH variation revealed several structural modifications. These observations, on a well-studied molecule like HEWL, underline not only the high impact of humidity levels on biological crystal structures, but also the significance of in-house XRPD as an analytical tool in industrial drug development and its potential to provide information for enhancing manufacturing of pharmaceuticals.
Collapse
|
28
|
A liquid chromatography high-resolution mass spectrometry in vitro assay to assess metabolism at the injection site of subcutaneously administered therapeutic peptides. J Pharm Biomed Anal 2018; 159:449-458. [DOI: 10.1016/j.jpba.2018.07.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/06/2018] [Accepted: 07/09/2018] [Indexed: 01/09/2023]
|
29
|
Wang W, Leu J, Watson R, Xu Z, Zhou H. Investigation of the Mechanism of Therapeutic Protein-Drug Interaction Between Methotrexate and Golimumab, an Anti-TNFα Monoclonal Antibody. AAPS JOURNAL 2018; 20:63. [PMID: 29667047 DOI: 10.1208/s12248-018-0219-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/12/2018] [Indexed: 12/31/2022]
Abstract
A prominent example of human therapeutic protein-drug interaction (TP-DI) is between methotrexate (MTX) and anti-TNFα mAbs. One plausible mechanism for this TP-DI is through the pharmacodynamic effect of MTX on immunogenicity. However, there is no definitive evidence to substantiate this mechanism, and other competing hypotheses, such as MTX suppressing FcγRI expression thereby affecting mAb PK, have also been proposed. In order to understand this mechanism, a cynomolgus monkey study was conducted using golimumab as a model compound. Golimumab elicited high incidences of immunogenicity in healthy cynomolgus monkeys. Concomitant dosing of MTX delayed the onset and reduced the magnitude of anti-drug antibody (ADA) formation. The impact of MTX on golimumab PK correlated with the ADA status. Prior to ADA formation, MTX has no discernable effect on golimumab PK. Additionally, no alteration in FcγRI expression was observed following MTX treatment. The impact of MTX on golimumab immunogenicity and PK has been observed in patients with rheumatoid arthritis, psoriatic arthritis (PsA), and ankylosing spondylitis. In a representative phase 3 study of golimumab in patients with PsA, patients not receiving concomitant MTX was reported to have ~ 30% lower steady-state trough golimumab levels compared to those who received MTX. However, further analysis showed that PsA patients who were negative for ADA in both treatment groups had comparable trough levels of golimumab. Taken together, our results suggest that the mechanism of TP-DI between MTX and golimumab can mostly be attributed to the pharmacodynamic effect of MTX, i.e., the lowering of immunogenicity and immunogenicity-mediated clearance of mAbs.
Collapse
Affiliation(s)
- Weirong Wang
- Biologics Development Sciences, Janssen R&D, LLC, Spring House, Pennsylvania, 19477, USA
| | - Jocelyn Leu
- Global Clinical Pharmacology, Janssen R&D, LLC, Spring House, Pennsylvania, 19477, USA
| | - Rebecca Watson
- Biologics Development Sciences, Janssen R&D, LLC, Spring House, Pennsylvania, 19477, USA
| | - Zhenhua Xu
- Global Clinical Pharmacology, Janssen R&D, LLC, Spring House, Pennsylvania, 19477, USA
| | - Honghui Zhou
- Global Clinical Pharmacology, Janssen R&D, LLC, Spring House, Pennsylvania, 19477, USA.
| |
Collapse
|
30
|
Yao M, Chen B, Zhao W, Mehl JT, Li L, Zhu M. LC-MS Differential Analysis for Fast and Sensitive Determination of Biotransformation of Therapeutic Proteins. Drug Metab Dispos 2018; 46:451-457. [PMID: 29386233 DOI: 10.1124/dmd.117.077792] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 01/29/2018] [Indexed: 11/22/2022] Open
Abstract
Therapeutic biologics have become a fast-growing segment within the pharmaceutical industry during the past 3 decades. Although the metabolism of biologics is more predictable than small molecule drugs, biotransformation can significantly affect the activity of biologics. Unfortunately, there are only a limited number of published studies on the biotransformation of biologics, most of which are focused on one or a few types of modifications. In this study, an untargeted LC-MS-based differential analysis approach was developed to rapidly and precisely determine the universal biotransformation profile of biologics with the assistance of bioinformatic tools. A human monoclonal antibody (mAb) was treated with t-butyl hydroperoxide and compared with control mAb using a bottom-up proteomics approach. Thirty-seven types of post-translational modifications were identified, and 38 peptides were significantly changed. Moreover, although all modifications were screened and detected, only the ones related to the treatment process were revealed by differential analysis. Other modifications that coexist in both groups were filtered out. This novel analytical strategy can be effectively applied to study biotransformation-mediated protein modifications, which will streamline the process of biologic drug discovery and development.
Collapse
Affiliation(s)
- Ming Yao
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (M.Y., W.Z., J.T.M., M.Z.); School of Pharmacy and Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin (B.C., L.L.); School of Life Sciences, Tianjin University, Nankai, Tianjin, People's Republic of China (L.L.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Bingming Chen
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (M.Y., W.Z., J.T.M., M.Z.); School of Pharmacy and Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin (B.C., L.L.); School of Life Sciences, Tianjin University, Nankai, Tianjin, People's Republic of China (L.L.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Weiping Zhao
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (M.Y., W.Z., J.T.M., M.Z.); School of Pharmacy and Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin (B.C., L.L.); School of Life Sciences, Tianjin University, Nankai, Tianjin, People's Republic of China (L.L.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - John T Mehl
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (M.Y., W.Z., J.T.M., M.Z.); School of Pharmacy and Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin (B.C., L.L.); School of Life Sciences, Tianjin University, Nankai, Tianjin, People's Republic of China (L.L.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Lingjun Li
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (M.Y., W.Z., J.T.M., M.Z.); School of Pharmacy and Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin (B.C., L.L.); School of Life Sciences, Tianjin University, Nankai, Tianjin, People's Republic of China (L.L.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| | - Mingshe Zhu
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey (M.Y., W.Z., J.T.M., M.Z.); School of Pharmacy and Department of Chemistry, University of Wisconsin-Madison, Madison, Wisconsin (B.C., L.L.); School of Life Sciences, Tianjin University, Nankai, Tianjin, People's Republic of China (L.L.); and MassDefect Technologies, Princeton, New Jersey (M.Z.)
| |
Collapse
|
31
|
A short review of the pharmacokinetic behavior of biological medicinal agents for the clinical practice. Microchem J 2018. [DOI: 10.1016/j.microc.2017.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
32
|
Dostalek M, Prueksaritanont T, Kelley RF. Pharmacokinetic de-risking tools for selection of monoclonal antibody lead candidates. MAbs 2017; 9:756-766. [PMID: 28463063 DOI: 10.1080/19420862.2017.1323160] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Pharmacokinetic studies play an important role in all stages of drug discovery and development. Recent advancements in the tools for discovery and optimization of therapeutic proteins have created an abundance of candidates that may fulfill target product profile criteria. Implementing a set of in silico, small scale in vitro and in vivo tools can help to identify a clinical lead molecule with promising properties at the early stages of drug discovery, thus reducing the labor and cost in advancing multiple candidates toward clinical development. In this review, we describe tools that should be considered during drug discovery, and discuss approaches that could be included in the pharmacokinetic screening part of the lead candidate generation process to de-risk unexpected pharmacokinetic behaviors of Fc-based therapeutic proteins, with an emphasis on monoclonal antibodies.
Collapse
Affiliation(s)
- Miroslav Dostalek
- a Drug Metabolism and Pharmacokinetics, Global Nonclinical Development, Shire , Lexington , MA , USA
| | | | - Robert F Kelley
- c Department of Drug Delivery , Genentech Inc. , South San Francisco , CA , USA
| |
Collapse
|
33
|
Geldenhuys WJ, Khayat MT, Yun J, Nayeem MA. Drug Delivery and Nanoformulations for the Cardiovascular System. RESEARCH & REVIEWS. DRUG DELIVERY 2017; 1:32-40. [PMID: 28713881 PMCID: PMC5507069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Therapeutic delivery to the cardiovascular system may play an important role in the successful treatment of a variety of disease state, including atherosclerosis, ischemic-reperfusion injury and other types of microvascular diseases including hypertension. In this review we evaluate the different options available for the development of suitable delivery systems that include the delivery of small organic compounds [adenosin A2A receptor agonist (CGS 21680), CYP-epoxygenases inhibitor (N-(methylsulfonyl)-2-(2-propynyloxy)-benzenehexanamide, trans-4-[4-(3-adamantan-1-ylureido)cyclohexyloxy] benzoic acid), soluble epoxide hydrolase inhibitor (N-methylsulfonyl-12,12-dibromododec-11-enamide), PPARγ agonist (rosiglitazone) and PPARγ antagonist (T0070907)], nanoparticles, peptides, and siRNA to the cardiovascular system. Effective formulations of nanoproducts have significant potential to overcome physiological barriers and improve therapeutic outcomes in patients. As per the literature covering targeted delivery to the cardiovascular system, we found that this area is still at infancy stage, as compare to the more mature fields of tumor cancer or brain delivery (e.g. blood-brain barrier permeability) with fewer publications focused on the targeted drug delivery technologies. Additionally, we show how pharmacology needs to be well understood when considering the cardiovascular system. Therefore, we discussed in this review various receptors agonists, antagonists, activators and inhibitors which will have effects on cardiovascular system.
Collapse
Affiliation(s)
- WJ Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University, School of Pharmacy, Morgantown WV 26506 USA
| | - MT Khayat
- Department of Pharmaceutical Sciences, West Virginia University, School of Pharmacy, Morgantown WV 26506 USA
- Deparment of Pharmaceutical Chemistry, King Abdulaziz University, School of Pharmacy, Jeddah, Saudi Arabia
| | - J Yun
- Department of Integrative Medical Sciences, Northeast Ohio Medical University College of Medicine, Rootstown OH 44272 USA
| | - MA Nayeem
- Department of Pharmaceutical Sciences, West Virginia University, School of Pharmacy, Morgantown WV 26506 USA
| |
Collapse
|
34
|
Schad F, Axtner J, Kröz M, Matthes H, Steele ML. Safety of Combined Treatment With Monoclonal Antibodies and Viscum album L Preparations. Integr Cancer Ther 2016; 17:41-51. [PMID: 29444603 PMCID: PMC5950938 DOI: 10.1177/1534735416681641] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Combination strategies involving chemotherapy and monoclonal antibodies (mAb) are
commonly used in attempts to produce better clinical outcomes. This practice has
led to new and ongoing toxicities that may lead to reductions in dose or
noncompliance, limiting the effectiveness of treatment. Viscum
album L (VA) preparations are widely used in Europe as additive
therapy and have been associated with reduced chemotherapy-related adverse
reactions and increased health-related quality of life. Concomitant VA therapy
might also reduce toxicity related to mAb. This retrospective study investigated
the safety of combined treatment with VA and mAb in cancer patients. A total of
43 patients had combined therapy (474 exposures); 12 had VA without mAb (129
exposures), and 8 had mAb without VA (68 exposures). Most patients (89.3%)
received concomitant chemotherapy or supportive therapies. A total of 34
patients (60.7%) experienced 142 adverse events (AEs). Leucopenia (14.1% of all
events), acneiform rash (8.5%), and stomatitis (6.3%) occurred most frequently.
Longitudinal logistic regression analysis suggested a nearly 5 times higher odds
of experiencing an AE following treatment with mAb compared with mAb plus VA
(95% CI = 1.53-16.14). Our results, together with theoretical consideration of
potential botanical-drug interactions, suggest that combined treatment with VA
and mAb is safe.
Collapse
Affiliation(s)
- Friedemann Schad
- 1 Research Institute Havelhoehe, Berlin, Germany.,2 Hospital Havelhoehe, Berlin, Germany
| | - Jan Axtner
- 1 Research Institute Havelhoehe, Berlin, Germany
| | - Matthias Kröz
- 1 Research Institute Havelhoehe, Berlin, Germany.,2 Hospital Havelhoehe, Berlin, Germany.,3 Charité University Medical Center, Berlin, Germany.,4 University of Witten/Herdecke, Herdecke, Germany
| | - Harald Matthes
- 1 Research Institute Havelhoehe, Berlin, Germany.,2 Hospital Havelhoehe, Berlin, Germany
| | | |
Collapse
|
35
|
Li Y, Monine M, Huang Y, Swann P, Nestorov I, Lyubarskaya Y. Quantitation and pharmacokinetic modeling of therapeutic antibody quality attributes in human studies. MAbs 2016; 8:1079-87. [PMID: 27216574 PMCID: PMC4968108 DOI: 10.1080/19420862.2016.1186322] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
A thorough understanding of drug metabolism and disposition can aid in the assessment of efficacy and safety. However, analytical methods used in pharmacokinetics (PK) studies of protein therapeutics are usually based on ELISA, and therefore can provide a limited perspective on the quality of the drug in concentration measurements. Individual post-translational modifications (PTMs) of protein therapeutics are rarely considered for PK analysis, partly because it is technically difficult to recover and quantify individual protein variants from biological fluids. Meanwhile, PTMs may be directly linked to variations in drug efficacy and safety, and therefore understanding of clearance and metabolism of biopharmaceutical protein variants during clinical studies is an important consideration. To address such challenges, we developed an affinity-purification procedure followed by peptide mapping with mass spectrometric detection, which can profile multiple quality attributes of therapeutic antibodies recovered from patient sera. The obtained data enable quantitative modeling, which allows for simulation of the PK of different individual PTMs or attribute levels in vivo and thus facilitate the assessment of quality attributes impact in vivo. Such information can contribute to the product quality attribute risk assessment during manufacturing process development and inform appropriate process control strategy.
Collapse
|
36
|
Abstract
This article provides an overview of the information and factors relevant to designing bioanalytical strategies in support of in vivo nonclinical and clinical studies of protein therapeutics. The summarized information includes representative types of the therapeutic proteins, their key structural characteristics, the relationship between post-translational modifications and function, issues during purification and formulation, PK of therapeutic proteins and immunogenicity. The effect of each of those on bioanalysis strategy has been pointed out. The impacts of structural variant and ‘free’/‘bound’ forms on PK assessment have been discussed.
Collapse
|
37
|
Tibbitts J, Canter D, Graff R, Smith A, Khawli LA. Key factors influencing ADME properties of therapeutic proteins: A need for ADME characterization in drug discovery and development. MAbs 2015; 8:229-45. [PMID: 26636901 PMCID: PMC4966629 DOI: 10.1080/19420862.2015.1115937] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Protein therapeutics represent a diverse array of biologics including antibodies, fusion proteins, and therapeutic replacement enzymes. Since their inception, they have revolutionized the treatment of a wide range of diseases including respiratory, vascular, autoimmune, inflammatory, infectious, and neurodegenerative diseases, as well as cancer. While in vivo pharmacokinetic, pharmacodynamic, and efficacy studies are routinely carried out for protein therapeutics, studies that identify key factors governing their absorption, distribution, metabolism, and excretion (ADME) properties have not been fully investigated. Thorough characterization and in-depth study of their ADME properties are critical in order to support drug discovery and development processes for the production of safer and more effective biotherapeutics. In this review, we discuss the main factors affecting the ADME characteristics of these large macromolecular therapies. We also give an overview of the current tools, technologies, and approaches available to investigate key factors that influence the ADME of recombinant biotherapeutic drugs, and demonstrate how ADME studies will facilitate their future development.
Collapse
|
38
|
Bioanalytical approaches to assess the proteolytic stability of therapeutic fusion proteins. Bioanalysis 2015; 7:3035-51. [DOI: 10.4155/bio.15.217] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Therapeutic fusion proteins (TFPs) are designed to improve the therapeutic profile of an endogenous protein or protein fragment with a limited dose frequency providing the desired pharmacological activity in vivo. Fusion of a therapeutic protein to a half-life extension or targeting domain can improve the disposition of the molecule or introduce a novel mechanism of action. Prolonged exposure and altered biodistribution of an endogenous protein through fusion technology increases the potential for local protein unfolding during circulation increasing the chance for partial proteolysis of the therapeutic domain. Characterizing the proteolytic liabilities of a TFP can guide engineering efforts to inhibit or hinder partial proteolysis. This review focuses on considerations and techniques for evaluating the stability of a TFP both in vivo and in vitro.
Collapse
|
39
|
Stratton CF, Newman DJ, Tan DS. Cheminformatic comparison of approved drugs from natural product versus synthetic origins. Bioorg Med Chem Lett 2015; 25:4802-4807. [PMID: 26254944 PMCID: PMC4607632 DOI: 10.1016/j.bmcl.2015.07.014] [Citation(s) in RCA: 121] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 07/08/2015] [Indexed: 11/27/2022]
Abstract
Despite the recent decline of natural product discovery programs in the pharmaceutical industry, approximately half of all new drug approvals still trace their structural origins to a natural product. Herein, we use principal component analysis to compare the structural and physicochemical features of drugs from natural product-based versus completely synthetic origins that were approved between 1981 and 2010. Drugs based on natural product structures display greater chemical diversity and occupy larger regions of chemical space than drugs from completely synthetic origins. Notably, synthetic drugs based on natural product pharmacophores also exhibit lower hydrophobicity and greater stereochemical content than drugs from completely synthetic origins. These results illustrate that structural features found in natural products can be successfully incorporated into synthetic drugs, thereby increasing the chemical diversity available for small-molecule drug discovery.
Collapse
Affiliation(s)
- Christopher F Stratton
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 422, New York, NY 10065, USA
| | - David J Newman
- Natural Products Branch, Developmental Therapeutics Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Frederick, PO Box B, Frederick, MD 21702, USA
| | - Derek S Tan
- Tri-Institutional PhD Program in Chemical Biology, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 422, New York, NY 10065, USA; Chemical Biology Program and Tri-Institutional Research Program, Memorial Sloan Kettering Cancer Center, 1275 York Ave, Box 422, New York, NY 10065, USA
| |
Collapse
|
40
|
Visk D. Will Advances in Preclinical In Vitro Models Lower the Costs of Drug Development? ACTA ACUST UNITED AC 2015. [DOI: 10.1089/aivt.2015.1503] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
41
|
Hojjat-Farsangi M. Small-molecule inhibitors of the receptor tyrosine kinases: promising tools for targeted cancer therapies. Int J Mol Sci 2014; 15:13768-801. [PMID: 25110867 PMCID: PMC4159824 DOI: 10.3390/ijms150813768] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 07/31/2014] [Accepted: 08/05/2014] [Indexed: 12/20/2022] Open
Abstract
Chemotherapeutic and cytotoxic drugs are widely used in the treatment of cancer. In spite of the improvements in the life quality of patients, their effectiveness is compromised by several disadvantages. This represents a demand for developing new effective strategies with focusing on tumor cells and minimum side effects. Targeted cancer therapies and personalized medicine have been defined as a new type of emerging treatments. Small molecule inhibitors (SMIs) are among the most effective drugs for targeted cancer therapy. The growing number of approved SMIs of receptor tyrosine kinases (RTKs) i.e., tyrosine kinase inhibitors (TKIs) in the clinical oncology imply the increasing attention and application of these therapeutic tools. Most of the current approved RTK-TKIs in preclinical and clinical settings are multi-targeted inhibitors with several side effects. Only a few specific/selective RTK-TKIs have been developed for the treatment of cancer patients. Specific/selective RTK-TKIs have shown less deleterious effects compared to multi-targeted inhibitors. This review intends to highlight the importance of specific/selective TKIs for future development with less side effects and more manageable agents. This article provides an overview of: (1) the characteristics and function of RTKs and TKIs; (2) the recent advances in the improvement of specific/selective RTK-TKIs in preclinical or clinical settings; and (3) emerging RTKs for targeted cancer therapies by TKIs.
Collapse
Affiliation(s)
- Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene Therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm 17176, Sweden.
| |
Collapse
|
42
|
Hall MP. Biotransformation and in vivo stability of protein biotherapeutics: impact on candidate selection and pharmacokinetic profiling. Drug Metab Dispos 2014; 42:1873-80. [PMID: 24947971 DOI: 10.1124/dmd.114.058347] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Historically, since the metabolism of administered peptide/protein drugs ("biotherapeutics") has been expected to undergo predictable pathways similar to endogenous proteins, comprehensive biotherapeutic metabolism studies have not been widely reported in the literature. However, since biotherapeutics have rapidly evolved into an impressive array of eclectic modalities, there has been a shift toward understanding the impact of metabolism on biotherapeutic development. For biotherapeutics containing non-native chemical linkers and other moieties besides natural amino acids, metabolism studies are critical as these moieties may impart undesired toxicology. For biotherapeutics that are composed solely of natural amino acids, where end-stage peptide and amino acid catabolites do not generally pose toxicity concerns, the understanding of biotherapeutic biotransformation, defined as in vivo modifications such as peripherally generated intermediate circulating catabolites prior to end-stage degradation or elimination, may impact in vivo stability and potency/clearance. As of yet, there are no harmonized methodologies for understanding biotherapeutic biotransformation and its impact on drug development, nor is there clear guidance from regulatory agencies on how and when these studies should be conducted. This review provides an update on biotherapeutic biotransformation studies and an overview of lessons learned, tools that have been developed, and suggestions of approaches to address issues. Biotherapeutic biotransformation studies, especially for certain modalities, should be implemented at an early stage of development to 1) understand the impact on potency/clearance, 2) select the most stable candidates or direct protein re-engineering efforts, and 3) select the best bioanalytical technique(s) for proper drug quantification and subsequent pharmacokinetic profiling and exposure/response assessment.
Collapse
Affiliation(s)
- Michael P Hall
- Department of Pharmacokinetics & Drug Metabolism, Amgen Inc., Thousand Oaks, California
| |
Collapse
|
43
|
Chen N, Wang W, Fauty S, Fang Y, Hamuro L, Hussain A, Prueksaritanont T. The effect of the neonatal Fc receptor on human IgG biodistribution in mice. MAbs 2014; 6:502-8. [PMID: 24492305 DOI: 10.4161/mabs.27765] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The neonatal Fc receptor (FcRn) plays a pivotal role in IgG homeostasis, i.e., it salvages IgG antibodies from lysosomal degradation following fluid-phase pinocytosis, thus preventing rapid systemic elimination of IgG. Recombinant therapeutic antibodies are typically composed of human or humanized sequences, and their biodistribution, or tissue distribution, is often studied in murine models, although, the effect of FcRn on tissue distribution of human IgG in rodents has not been investigated. In this report, an (125)I-labeled human IgG1 antibody was studied in both wild type C57BL/6 (WT) and FcRn knockout (KO) mice. Total radioactivity in both plasma and tissues (0-96hr post-dose) was measured by gamma-counting. Plasma exposure of human IgG1 were significantly lower in FcRn KO mice, which is consistent with the primary function of FcRn. Differences in biodistribution of human IgG to selected tissues were also observed. Among the tissue examined, the fat, skin and muscle showed a decrease in tissue-to-blood (T/B) exposure ratio of human IgG1 in FcRn KO mice comparing to the WT mice, while the liver, spleen, kidney, and lung showed an increase in the T/B exposure ratio in FcRn KO mice. A time-dependent change in the T/B ratios of human IgG1 was also observed for many tissues in FcRn KO mice. These results suggest that, in addition to its role in IgG elimination, FcRn may also play a role in antibody biodistribution.
Collapse
Affiliation(s)
- Nancy Chen
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism; Merck Research Laboratories; West Point, PA USA
| | - Weirong Wang
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism; Merck Research Laboratories; West Point, PA USA
| | - Scott Fauty
- Department of Laboratory Animal Resources; Merck Research Laboratories; West Point, PA USA
| | - Yulin Fang
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism; Merck Research Laboratories; West Point, PA USA
| | - Lora Hamuro
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism; Merck Research Laboratories; West Point, PA USA
| | - Azher Hussain
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism; Merck Research Laboratories; West Point, PA USA
| | - Thomayant Prueksaritanont
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism; Merck Research Laboratories; West Point, PA USA
| |
Collapse
|