1
|
Li Y, Villafuerte-Vega R, Shenoy VM, Jackson HM, Wang Y, Parrish KE, Jenkins GJ, Sarvaiya H. A novel in vitro serum stability assay for antibody therapeutics incorporating internal standards. MAbs 2025; 17:2479529. [PMID: 40097239 PMCID: PMC11917174 DOI: 10.1080/19420862.2025.2479529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/07/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025] Open
Abstract
Antibody-based therapeutics have demonstrated remarkable therapeutic benefit, but their susceptibility to biotransformation and degradation in the body can affect their safety, efficacy, and pharmacokinetic/pharmacodynamic (PK/PD) profiles. In vitro stability assessments play a pivotal role in proactively identifying potential liabilities of antibody therapeutics prior to animal studies. Liquid chromatography-mass spectrometry (LC-MS)-based in vitro stability assays has been developed and adopted in the biopharmaceutical industry for the characterization of antibody-based therapeutics. However, these methodologies often overlook operational error and random variation during sample preparation and analysis, leading to inaccurate stability estimation. To address this limitation, we have developed an LC-MS-based in vitro serum stability assessment that incorporates two internal standards (ISs), National Institute of Standards and Technology monoclonal antibody (NISTmAb) and its crystallizable fragment (Fc), to improve assay performance. Our method involves three steps: incubation of antibody therapeutics along with an IS in biological matrices, affinity purification, and LC-MS analysis. The stability of 21 monoclonal or bispecific antibodies was assessed in serums of preclinical species using this method. Our results showed improved accuracy and precision of recovery calculations with the incorporation of ISs, enabling a more confident stability assessment even in the absence of biotransformation or aggregation. In vitro stability correlated with in vivo exposure, suggesting that this in vitro assay could serve as a routine screening tool to select and advance stable antibody therapeutic candidates for subsequent in vivo studies.
Collapse
Affiliation(s)
- Yihan Li
- Department of Quantitative, Translational & ADME Sciences, AbbVie, South San Francisco, CA, USA
| | | | - Vikram M Shenoy
- Department of Quantitative, Translational & ADME Sciences, AbbVie, South San Francisco, CA, USA
| | - Heidi M Jackson
- Department of Quantitative, Translational & ADME Sciences, AbbVie, Worcester, MA, USA
| | - Yuting Wang
- Department of Quantitative, Translational & ADME Sciences, AbbVie, Worcester, MA, USA
| | - Karen E Parrish
- Department of Quantitative, Translational & ADME Sciences, AbbVie, North Chicago, IL, USA
| | - Gary J Jenkins
- Department of Quantitative, Translational & ADME Sciences, AbbVie, North Chicago, IL, USA
| | - Hetal Sarvaiya
- Department of Quantitative, Translational & ADME Sciences, AbbVie, South San Francisco, CA, USA
| |
Collapse
|
2
|
Li X, Dong S, Pan Q, Liu N, Zhang Y. Antibiotic conjugates: Using molecular Trojan Horses to overcome drug resistance. Biomed Pharmacother 2025; 186:118007. [PMID: 40268370 DOI: 10.1016/j.biopha.2025.118007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 04/25/2025] Open
Abstract
Antimicrobial resistance (AMR) has become a global health crisis due to the rapid emergence of multi-drug-resistant bacteria. The paucity of novel antibiotics in the clinical pipeline has exacerbated this issue, thereby warranting the development of new antibacterial therapies. The 'Trojan Horse' strategy entails conjugating antibiotics with bioactive components that not only facilitate the entry of antibiotic molecules into bacterial cells by circumventing the membrane barriers, but also augment the effects of conventional antibiotics against recalcitrant pathogens. These Trojan Horse elements can also serve as a promising tool for repurposing drugs with hitherto unexamined antimicrobial activity, or drugs with limited clinical utility due to considerable toxic side effects. In this review, we have discussed the current state of research on antibiotic conjugates with monoclonal antibodies (mAbs), antimicrobial peptides (AMPs) and the iron-chelating siderophores. The rationale and mechanisms of different antibiotic conjugates have been summarized, and the preclinical and clinical evidence pertaining to the activity of these conjugates against drug-resistant pathogens have been reviewed. Furthermore, the challenges associated with the clinical translation of these novel antimicrobials, and the future research directions have also been discussed. While antibiotic conjugates offer an attractive alternative to conventional antimicrobials, there are several obstacles to their clinical translation. A greater understanding of the mechanisms underlying AMR, and continuing advances in genetic engineering, synthetic biology, and bioinformatics will be crucial in designing more selective, potent, and safe antibiotic conjugates for tackling multi-drug resistant (MDR) infections.
Collapse
Affiliation(s)
- Xi Li
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyuan Dong
- Department of Thoracic surgery, The First Hospital of China Medical University, Shenyang, China
| | - Qi Pan
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China
| | - Ning Liu
- Department of Rehabilitation, the First Affiliated Hospital of China Medical University, Shenyang 110001, China.
| | - Yijie Zhang
- Department of Organ Transplantation and Hepatobiliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China; The Key Laboratory of Organ Transplantation in Liaoning Province, Shenyang, Liaoning, China.
| |
Collapse
|
3
|
Xie H, Sun L, Yao S, Tian X, Jin L, Dai Y, Li Y, Li Y, Fang J, Guo P, Zhang Y. Therapeutically targeting endometrial cancer in preclinical models by ICAM1 antibody-drug conjugates. Gynecol Oncol 2025; 196:16-27. [PMID: 40147093 DOI: 10.1016/j.ygyno.2025.03.027] [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: 01/10/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025]
Abstract
OBJECTIVE The incidence of mortality and morbidity from endometrial cancer (EC) is increasing annually, and there is a paucity of effective targeted therapies for the condition. Antibody-drug conjugates (ADCs) represent a promising approach to tumor-targeted therapy. In this study, we aim to identify a novel molecular target for the preclinical development of EC-targeted ADCs. METHODS Through quantitative and unbiased bioinformatics analyses intercellular adhesion molecule-1 (ICAM1) was identified as a potential cell membrane target. Two ADCs, ICAM1-MMAE and ICAM1-DXd, were subsequently developed by conjugating ICAM1 monoclonal antibodies with microtubule inhibitors and DNA topoisomerase inhibitors, respectively. The preclinical efficacy and biosafety of these ICAM1 ADCs were validated in both in vitro and in vivo models. Furthermore, transcriptomic analysis was conducted to elucidate the therapeutic effects of the ICAM1 ADCs. RESULTS Quantitative flow screening and bioinformatics analyses revealed significant overexpression of ICAM1 in EC. ICAM1-MMAE and ICAM1-DXd were developed using clinically effective linkers and payloads. In preclinical models, ICAM1 ADCs showed superior antitumor efficacy compared to standard chemotherapy, achieving sustained tumor regression with an excellent safety profile in both subcutaneous and orthotopic xenograft models. Transcriptomic analysis further revealed that ICAM1-DXd potently activated tumor immunity. CONCLUSIONS ICAM1 was identified as a promising cell membrane protein target for ADC development in EC. As-synthesized ICAM1 ADCs demonstrated potent antitumor activity, favorable biosafety profiles in vitro and in vivo, and the ability to activate tumor immunity. These findings support the potential of ICAM1 ADCs as a therapeutic strategy and warrant further investigation in clinical studies.
Collapse
Affiliation(s)
- Hanfei Xie
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou 310022, China
| | - Lu Sun
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China
| | - Shili Yao
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China
| | - Xuefei Tian
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; Department of Biological Medicines and Shanghai Engineering Research Center of Immunotherapeutics, Fudan University, Shanghai 201203, China
| | - Liming Jin
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China
| | - Yujie Dai
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; MOE Frontier Science Centre for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Yuanzheng Li
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China
| | - Yuxuan Li
- Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China
| | - Jianmin Fang
- School of Life Science and Technology, Tongji University, Shanghai 200092, China
| | - Peng Guo
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; School of Materials Science and Engineering, Tianjin University, Tianjin 300072, China; Department of Urology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Structural Birth Defect and Reconstruction, Chongqing 400014, China; School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, China; MOE Frontier Science Centre for Precision Oncology, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China; Eye Research Center, Hangzhou Institute of Medicine, Chinese Academy of Sciences, Eye Hospital, Wenzhou Medical University, Hangzhou, Zhejiang 310018, China.
| | - Yingli Zhang
- Department of Gynecologic Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China; Clinical and Translational Research Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310018, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou 310022, China.
| |
Collapse
|
4
|
Subramaniyan S, Jayaraman M, Jeyaraman J. Exploring phytochemicals and marine natural products as alternative therapeutic agents targeting phosphotransacetylase (PTA) in Mycobacterium tuberculosis: An underexplored drug target. J Mol Graph Model 2025; 138:109025. [PMID: 40132354 DOI: 10.1016/j.jmgm.2025.109025] [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: 01/13/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 03/27/2025]
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (MTB), remains a significant global health threat due to its widespread prevalence and increasing drug resistance. This study targets phosphotransacetylase (PTA), an essential enzyme in acetate metabolism, as a potential therapeutic target. A comprehensive multi-tiered virtual screening approach was employed to identify potent phytochemicals and marine natural products (MNPs) from five databases (AMMPDB, CMNPD, MNPD, Seaweed and SPECS). Five promising bioactive molecules (AMMPDB10473, CMNPD23347, CMNPD5918, MNPD6660, and SPECS-AK-693) were identified, showing high docking scores (-8.17 to -10.83 kcal/mol) and MM-GBSA binding energy scores (-47.51 to -59.14 kcal/mol). These molecules adhered to Lipinski's rule of five (Ro5) and demonstrated acceptable pharmacokinetic profiles. Density functional theory (DFT) calculations further validated the interaction potential of these molecules through HOMO and LUMO analysis. Long-range molecular dynamics simulations (MDS) over 300 ns confirmed the structural stability and enhanced hydrogen-bonding potential of the natural products-PTA complexes. Principal component analysis (PCA) and free energy landscape (FEL) contour plots revealed a single dominant energy basin, indicating structural stability and limited conformational flexibility of the complexes. Additionally, MMPBSA analysis corroborated the strong binding affinities of the identified hit molecules with PTA. Critical 'hot spot' residues (Phe516, Cys530, Ala531, and Tyr639) were identified, contributing significantly to the structural stability and binding energy of the complexes. This computational study offers valuable insights into the potential of these lead molecules for combating TB, providing a foundation for experimental validation and innovative therapeutic development, and paving the way for future research and breakthroughs in TB treatment.
Collapse
Affiliation(s)
- Sneha Subramaniyan
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, 630 003, Tamil Nadu, India.
| | - Manikandan Jayaraman
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, 630 003, Tamil Nadu, India.
| | - Jeyakanthan Jeyaraman
- Structural Biology and Bio-Computing Lab, Department of Bioinformatics, Science Block, Alagappa University, Karaikudi, 630 003, Tamil Nadu, India.
| |
Collapse
|
5
|
DeBonis J, Veiseh O, Igoshin OA. Uncovering the interleukin-12 pharmacokinetic desensitization mechanism and its consequences with mathematical modeling. CPT Pharmacometrics Syst Pharmacol 2025; 14:217-228. [PMID: 39415353 PMCID: PMC11812943 DOI: 10.1002/psp4.13258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/25/2024] [Accepted: 10/02/2024] [Indexed: 10/18/2024] Open
Abstract
The cytokine interleukin-12 (IL-12) is a potential immunotherapy because of its ability to induce a Th1 immune response. However, success in the clinic has been limited due to a phenomenon called IL-12 desensitization - the trend where repeated exposure to IL-12 leads to reduced IL-12 concentrations (pharmacokinetics) and biological effects (pharmacodynamics). Here, we investigated IL-12 pharmacokinetic desensitization via a modeling approach to (i) validate proposed mechanisms in literature and (ii) develop a mathematical model capable of predicting IL-12 pharmacokinetic desensitization. Two potential causes of IL-12 pharmacokinetic desensitization were identified: increased clearance or reduced bioavailability of IL-12 following repeated doses. Increased IL-12 clearance was previously proposed to occur due to the upregulation of IL-12 receptor on T cells that causes increased receptor-mediated clearance in the serum. However, our model with this mechanism, the accelerated-clearance model, failed to capture trends in clinical trial data. Alternatively, our novel reduced-bioavailability model assumed that upregulation of IL-12 receptor on T cells in the lymphatic system leads to IL-12 sequestration, inhibiting the transport to the blood. This model accurately fits IL-12 pharmacokinetic data from three clinical trials, supporting its biological relevance. Using this model, we analyzed the model parameter space to illustrate that IL-12 desensitization occurs over a robust range of parameter values and to identify the conditions required for desensitization. We next simulated local, continuous IL-12 delivery and identified several methods to mitigate systemic IL-12 exposure. Ultimately, our results provide quantitative validation of our proposed mechanism and allow for accurate prediction of IL-12 pharmacokinetics over repeated doses.
Collapse
Affiliation(s)
| | - Omid Veiseh
- Department of BioengineeringRice UniversityHoustonTexasUSA
| | - Oleg A. Igoshin
- Department of BioengineeringRice UniversityHoustonTexasUSA
- Department of ChemistryRice UniversityHoustonTexasUSA
- Department of BiosciencesRice UniversityHoustonTexasUSA
| |
Collapse
|
6
|
Debnath A, Singh RK, Mazumder R, Mazumder A, Srivastava S, Chaudhary H, Mangal S, Sanchitra J, Tyagi PK, Kumar Singh S, Singh AK. Quest for discovering novel CDK12 inhibitor. J Recept Signal Transduct Res 2025; 45:1-21. [PMID: 39697035 DOI: 10.1080/10799893.2024.2441185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/20/2024]
Abstract
CDK12 is essential for cellular processes like RNA processing, transcription, and cell cycle regulation, inhibiting cancer cell growth and facilitating macrophage invasion. CDK12 is a significant oncogenic factor in various cancers, including HER2-positive breast cancer, Anaplastic thyroid carcinoma, Hepatocellular carcinoma, prostate cancer, and Ewing sarcoma. It is also regarded as a potential biomarker, emphasizing its broader significance in oncology. Targeting CDK12 offers a promising strategy to develop therapy. Various monoclonal antibodies have drawn wide attention, but they are expensive compared to small-molecule inhibitors, limiting their accessibility and affordability for patients. Consequently, this research aims to identify effective CDK12 inhibitors using comprehensive high-throughput virtual screening. RASPD protocol has been employed to screen three different databases against the target followed by drug-likeness, molecular docking, ADME, toxicity, Consensus molecular docking, MD Simulation, and in-vitro studies MTT assay. The research conducted yielded one compound ZINC11784547 has demonstrated robust binding affinity, favorable ADME features, less toxicity, remarkable stability, and cytotoxic effect. The identified compound holds promise for promoting cancer cell death through CDK12 inhibition.
Collapse
Affiliation(s)
- Abhijit Debnath
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Rajesh Kumar Singh
- Department of Dravyaguna, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rupa Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Shikha Srivastava
- Bhaskaracharya College of Applied Sciences, University of Delhi, Delhi, India
| | - Hema Chaudhary
- School of Medical & Allied Sciences, K R Mangalam University, Gurugram, India
| | - Saloni Mangal
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | - Jahanvi Sanchitra
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, India
| | | | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Anil Kumar Singh
- Department of Dravyaguna, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
7
|
Tran YN, Chansaenroj A, Jivaphetthai A, Osathanon T, Arunmanee W. Truncated recombinant Jagged1 fused with human IgG1 Fc activates Notch target genes in human periodontal ligament cells. Arch Oral Biol 2025; 170:106138. [PMID: 39643955 DOI: 10.1016/j.archoralbio.2024.106138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/13/2024] [Accepted: 11/25/2024] [Indexed: 12/09/2024]
Abstract
OBJECTIVE Jagged1, a Notch ligand, is essential for osteogenic differentiation in human periodontal ligament cells (hPDLs) by interacting with Notch2 to induce osteogenic markers, alkaline phosphatase activity, and mineral deposition. However, its large size hampers absorption and distribution of biomaterials. This study aimed to identify the critical region of Jagged1 necessary for its interaction with Notch2 to create a truncated version that retains osteogenic activity but with improved delivery characteristics. METHODS Truncated versions of Jagged1 were designed by deleting C-terminal regions, focusing on the importance of the N-terminal domain. Both truncated and full-length Jagged1 were fused with human IgG1 Fc (Jagged1-Fc) and expressed in Chinese hamster ovary cells. hPDLs treated with these constructs were analyzed for Notch target gene expression using real-time PCR. Mineral deposition was assessed using alizarin red S staining. RESULTS Both truncated and full-length Jagged1-Fc increased the expression of Notch target genes (Hes1, Hey1, and ALP) in hPDLs, indicating successful activation of Notch signaling. However, only the full-length Jagged1-Fc enhanced mineral deposition, while the truncated version did not. CONCLUSIONS Full-length Jagged1-Fc is required for mineral deposition and complete osteogenic differentiation in hPDLs. The truncated versions, while capable of activating Notch signaling, are ineffective in promoting mineralization, underscoring the importance of the entire protein for clinical applications in bone regeneration.
Collapse
Affiliation(s)
- Y Nhu Tran
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ajjima Chansaenroj
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; Department of Animal Husbandry, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Araya Jivaphetthai
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Thanaphum Osathanon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wanatchaporn Arunmanee
- Department of Biochemistry and Microbiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Center of Excellence in Cancer Cell and Molecular Biology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
8
|
Mulaudzi VE, Adeosun IJ, Adewumi AT, Soliman MES, Cosa S. Helichrysum populifolium Compounds Inhibit MtrCDE Efflux Pump Transport Protein for the Potential Management of Gonorrhoea Infection. Int J Mol Sci 2024; 25:13310. [PMID: 39769078 PMCID: PMC11677219 DOI: 10.3390/ijms252413310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
The progressive development of resistance in Neisseria gonorrhoeae to almost all available antibiotics has made it crucial to develop novel approaches to tackling multi-drug resistance (MDR). One of the primary causes of antibiotic resistance is the over-expression of the MtrCDE efflux pump protein, making this protein a vital target for fighting against antimicrobial resistance (AMR) in N. gonorrhoeae. This study was aimed at evaluating the potential MtrCDE efflux pump inhibitors (EPIs) and their stability in treating gonorrhoea infection. This is significant because finding novel EPIs would allow for the longer maintenance of antibiotics at therapeutic levels, thereby prolonging the susceptibility of currently available antibiotics. A virtual screening of the selected Helichrysum populifolium compounds (4,5-dicaffeoylquinic acid, apigeninin-7-glucoside, and carvacrol) was conducted to evaluate their potential EPI activity. An integrated computational framework consisting of molecular docking (MD), molecular mechanics generalized born, and surface area solvation (MMGBSA) analysis, molecular dynamics simulations (MDS), and absorption, distribution, metabolism, and excretion (ADME) properties calculations were conducted. Of the tested compounds, 4,5-dicaffeoylquinic acid revealed the highest molecular docking binding energies (-8.8 kcal/mol), equivalent MMGBSA binding free energy (-54.82 kcal/mol), indicative of consistent binding affinity with the MtrD protein, reduced deviations and flexibility (root mean square deviation (RMSD) of 5.65 Å) and, given by root mean square fluctuation (RMSF) of 1.877 Å. Carvacrol revealed a docking score of -6.0 kcal/mol and a MMGBSA computed BFE of -16.69 kcal/mol, demonstrating the lowest binding affinity to the MtrD efflux pump compared to the remaining test compounds. However, the average RMSD (4.45 Å) and RMSF (1.638 Å) of carvacrol-bound MtrD protein showed no significant difference from the unbound MtrD protein, except for the reference compounds, implying consistent MtrD conformation throughout simulations and indicates a desirable feature during drug design. Additionally, carvacrol obeyed the Lipinski rule of five which confirmed the compound's drug-likeness properties making it the most promising EPI candidate based on its combined attributes of a reasonable binding affinity, sustained stability during MDS, its obedience to the Lipinski rule of five and compliance with drug-likeness criteria. An in vitro validation of the potential EPI activities of H. populifolium compounds confirmed that 4,5-dicaffeoylquinic acid reduced the expulsion of the bis-benzimide dye by MtrCDE pump, while carvacrol showed low accumulation compared to other compounds. While 4,5-dicaffeoylquinic acid demonstrated the highest binding affinity in computational analysis and an EPI activity in vitro, it showed lower stability compared to the other compounds, as indicated in MDS. This leaves carvacrol, as a better EPI candidate for the management of gonorrhoea infection.
Collapse
Affiliation(s)
- Vhangani E. Mulaudzi
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa; (V.E.M.); (I.J.A.)
| | - Idowu J. Adeosun
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa; (V.E.M.); (I.J.A.)
| | - Adeniyi T. Adewumi
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (A.T.A.); (M.E.S.S.)
| | - Mahmoud E. S. Soliman
- Molecular Bio-Computation and Drug Design Laboratory, School of Health Sciences, University of KwaZulu-Natal, Durban 4000, South Africa; (A.T.A.); (M.E.S.S.)
| | - Sekelwa Cosa
- Department of Biochemistry, Genetics and Microbiology, University of Pretoria, Private Bag X20, Hatfield, Pretoria 0028, South Africa; (V.E.M.); (I.J.A.)
| |
Collapse
|
9
|
Ndarawit W, Ochieng CO, Angwenyi D, Cruz JN, Santos CBR, Kimani NM. Discovery of α-amylase and α-glucosidase dual inhibitors from NPASS database for management of Type 2 Diabetes Mellitus: A chemoinformatic approach. PLoS One 2024; 19:e0313758. [PMID: 39541274 PMCID: PMC11563405 DOI: 10.1371/journal.pone.0313758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Postprandial hyperglycemia, typical manifestation of Type 2 Diabetes Mellitus (T2DM), is associated with notable global morbidity and mortality. Preventing the advancement of this condition by delaying the rate of glucose absorption through inhibition of α-amylase and α-glucosidase enzymatic activities is of utmost importance. Finding a safe antidiabetic drug is essential since those that are currently on the market have drawbacks like unpleasant side effects. The current study utilized computer-aided drug design (CADD), as a quick and affordable method to find a substitute drug template that can be used to control postprandial hyperglycemia by modulating the activity of α-amylase and α-glucosidase enzymes. The Natural Products Activity and Species database (NPASS) (30,926 compounds) was screened in silico, with a focus on evaluating drug-likeness, toxicity profiles and ability to bind on a target protein. Two molecules NPC204580 (Chrotacumine C) and NPC137813 (1-O-(2-Methoxy-4-Acetylphenyl)-6-O-(E-Cinnamoyl)-Beta-D-Glucopyranoside) were identified as potential dual inhibitors for α-amylase and α-glucosidase with free binding energies of -14.46 kcal/mol and -12.58 kcal/mol for α-amylase, and -8.42 kcal/mol and -8.76 kcal/mol for α-glucosidase, respectively. The molecules showed ionic, H-bonding and hydrophobic interactions with critical amino acid residues of both enzymes. Moreover, 100 ns molecular dynamic simulations showed that both molecules are stable on the receptors' active sites based on root mean square deviation (RMSD), root mean square fluctuation (RMSF), and the Generalized Born surface area (GBSA) energy calculated. The two compounds are thus promising therapeutic agents for T2DM that merit further investigation due to their excellent binding energies, encouraging pharmacokinetics, toxicity profiles, and stability as demonstrated in simulated studies.
Collapse
Affiliation(s)
| | | | - David Angwenyi
- Department of Mathematics, Masinde Muliro University of Science and Technology, Kakamega, Kenya
| | - Jorddy N. Cruz
- Department of Biological Sciences and Health, Laboratory of Modeling and Computational Chemistry, Federal University of Amapá, Macapá, Amapá, Brazil
| | - Cleydson B. R. Santos
- Department of Biological Sciences and Health, Laboratory of Modeling and Computational Chemistry, Federal University of Amapá, Macapá, Amapá, Brazil
- Graduate Program in Medicinal Chemistry and Molecular Modelling, Health Science Institute, Federal University of Pará, Belém, PA, Brazil
| | - Njogu M. Kimani
- Department of Physical Sciences, University of Embu, Embu, Kenya
- Natural Product Chemistry and Computational Drug Discovery Laboratory, Embu, Kenya
| |
Collapse
|
10
|
Antonova NP, Vasina DV, Grigoriev IV, Laishevtsev AI, Kapustin AV, Savinov VA, Vorobev AM, Aleshkin AV, Zackharova AA, Remizov TA, Makarov VV, Yudin SM, Gushchin VA. Pharmacokinetic and preclinical safety studies of endolysin-based therapeutic for intravenous administration. Int J Antimicrob Agents 2024; 64:107328. [PMID: 39244166 DOI: 10.1016/j.ijantimicag.2024.107328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
Pharmacokinetics and safety studies of innovative drugs is an essential part of drug development process. Previously we have developed a novel drug for intravenous administration (lyophilizate) containing modified endolysin LysECD7-SMAP that showed notable antibacterial effect in different animal models of systemic infections. Here we present data on pharmacokinetics of endolysin in mice after single and multiple injections. Time-concentration curves were obtained, and pharmacokinetic parameters for preparation (C0, kel t1/2, AUC0-∞, MRT, ClT, Vss) were calculated. It was shown that although endolysin is rather short-lived in blood serum (t1/2 = 12.5 min), the therapeutic concentrations of LysECD7-SMAP (in degraded and non-degraded form) were detected for 60 minutes after injection that is sufficient for antibacterial effect. Based on the obtained data, it was proposed that endolysin distributes presumably in murine blood, degrades in blood and liver, and is eliminated via glomerular filtration. Safety profile of the preparation relating to general toxicity, immunotoxicity and allergenicity was assessed in rodents. It was demonstrated that LysECD7-SMAP in potential therapeutic (12.5 mg/kg), 10-fold (125 mg/kg) and 40-fold (500 mg/kg) doses showed no signs of intoxication and significant abnormalities after single and repeated i.v. administrations, preparation was non-immunogenic and induced minor and reversible allergic reaction in animals.
Collapse
Affiliation(s)
- Nataliia P Antonova
- Laboratory of Pathogen Population Variability Mechanisms, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Daria V Vasina
- Laboratory of Pathogen Population Variability Mechanisms, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Igor V Grigoriev
- Translational Biomedicine Laboratory, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Aleksei I Laishevtsev
- Federal State Budget Scientific Institution "Federal Scientific Centre VIEV" (FSC VIEV), Moscow, Russia
| | - Andrey V Kapustin
- Federal State Budget Scientific Institution "Federal Scientific Centre VIEV" (FSC VIEV), Moscow, Russia
| | - Vasiliy A Savinov
- Federal State Budget Scientific Institution "Federal Scientific Centre VIEV" (FSC VIEV), Moscow, Russia
| | - Aleksei M Vorobev
- Laboratory of Clinical Microbiology and Biotechnology of Bacteriophages, G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, Moscow, Russia
| | - Andrei V Aleshkin
- Laboratory of Clinical Microbiology and Biotechnology of Bacteriophages, G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, Moscow, Russia
| | - Anastasia A Zackharova
- Translational Biomedicine Laboratory, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Timofey A Remizov
- Translational Biomedicine Laboratory, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Valentine V Makarov
- Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Moscow, Russia
| | - Sergey M Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Moscow, Russia
| | - Vladimir A Gushchin
- Laboratory of Pathogen Population Variability Mechanisms, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia.
| |
Collapse
|
11
|
Patidar K, Pillai N, Dhakal S, Avery LB, Mavroudis PD. A minimal physiologically based pharmacokinetic model to study the combined effect of antibody size, charge, and binding affinity to FcRn/antigen on antibody pharmacokinetics. J Pharmacokinet Pharmacodyn 2024; 51:477-492. [PMID: 38400996 DOI: 10.1007/s10928-023-09899-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 12/26/2023] [Indexed: 02/26/2024]
Abstract
Protein therapeutics have revolutionized the treatment of a wide range of diseases. While they have distinct physicochemical characteristics that influence their absorption, distribution, metabolism, and excretion (ADME) properties, the relationship between the physicochemical properties and PK is still largely unknown. In this work we present a minimal physiologically-based pharmacokinetic (mPBPK) model that incorporates a multivariate quantitative relation between a therapeutic's physicochemical parameters and its corresponding ADME properties. The model's compound-specific input includes molecular weight, molecular size (Stoke's radius), molecular charge, binding affinity to FcRn, and specific antigen affinity. Through derived and fitted empirical relationships, the model demonstrates the effect of these compound-specific properties on antibody disposition in both plasma and peripheral tissues using observed PK data in mice and humans. The mPBPK model applies the two-pore hypothesis to predict size-based clearance and exposure of full-length antibodies (150 kDa) and antibody fragments (50-100 kDa) within a onefold error. We quantitatively relate antibody charge and PK parameters like uptake rate, non-specific binding affinity, and volume of distribution to capture the relatively faster clearance of positively charged mAb as compared to negatively charged mAb. The model predicts the terminal plasma clearance of slightly positively and negatively charged antibody in humans within a onefold error. The mPBPK model presented in this work can be used to predict the target-mediated disposition of a drug when compound-specific and target-specific properties are known. To our knowledge, a combined effect of antibody weight, size, charge, FcRn, and antigen has not been incorporated and studied in a single mPBPK model previously. By conclusively incorporating and relating a multitude of protein's physicochemical properties to observed PK, our mPBPK model aims to contribute as a platform approach in the early stages of drug development where many of these properties can be optimized to improve a molecule's PK and ultimately its efficacy.
Collapse
Affiliation(s)
- Krutika Patidar
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Nikhil Pillai
- Global DMPK Modeling & Simulation, Sanofi, 350 Water St, Cambridge, MA, 02141, USA
| | - Saroj Dhakal
- Global DMPK Modeling & Simulation, Sanofi, 350 Water St, Cambridge, MA, 02141, USA
| | | | | |
Collapse
|
12
|
Torres-Herrero B, Armenia I, Ortiz C, de la Fuente JM, Betancor L, Grazú V. Opportunities for nanomaterials in enzyme therapy. J Control Release 2024; 372:619-647. [PMID: 38909702 DOI: 10.1016/j.jconrel.2024.06.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
In recent years, enzyme therapy strategies have rapidly evolved to catalyze essential biochemical reactions with therapeutic potential. These approaches hold particular promise in addressing rare genetic disorders, cancer treatment, neurodegenerative conditions, wound healing, inflammation management, and infectious disease control, among others. There are several primary reasons for the utilization of enzymes as therapeutics: their substrate specificity, their biological compatibility, and their ability to generate a high number of product molecules per enzyme unit. These features have encouraged their application in enzyme replacement therapy where the enzyme serves as the therapeutic agent to rectify abnormal metabolic and physiological processes, enzyme prodrug therapy where the enzyme initiates a clinical effect by activating prodrugs, and enzyme dynamic or starving therapy where the enzyme acts upon host substrate molecules. Currently, there are >20 commercialized products based on therapeutic enzymes, but approval rates are considerably lower than other biologicals. This has stimulated nanobiotechnology in the last years to develop nanoparticle-based solutions that integrate therapeutic enzymes. This approach aims to enhance stability, prevent rapid clearance, reduce immunogenicity, and even enable spatio-temporal activation of the therapeutic catalyst. This comprehensive review delves into emerging trends in the application of therapeutic enzymes, with a particular emphasis on the synergistic opportunities presented by incorporating enzymes into nanomaterials. Such integration holds the promise of enhancing existing therapies or even paving the way for innovative nanotherapeutic approaches.
Collapse
Affiliation(s)
- Beatriz Torres-Herrero
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Ilaria Armenia
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain
| | - Cecilia Ortiz
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Jesús Martinez de la Fuente
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain
| | - Lorena Betancor
- Laboratorio de Biotecnología, Facultad de Ingeniería, Universidad ORT Uruguay, Mercedes 1237, 11100 Montevideo, Uruguay
| | - Valeria Grazú
- Instituto de Nanociencia y Materiales de Aragón (INMA), CSIC/Universidad de Zaragoza, c/ Edificio I+D, Mariano Esquillor Gómez, 50018 Zaragoza, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Avenida Monforte de Lemos, 3-5, 28029 Madrid, Spain.
| |
Collapse
|
13
|
Yoshijima C, Suzuki Y, Tanaka R, Ono H, Oda A, Ozaki T, Shibata H, Itoh H, Ohno K. Evaluation of intra- and inter-individual variations in plasma belimumab concentrations in adult patients with systemic lupus erythematosus. Pharmacol Res Perspect 2024; 12:e1255. [PMID: 39096038 PMCID: PMC11297282 DOI: 10.1002/prp2.1255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/19/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
In this study, plasma belimumab concentrations were measured over the course of treatment in systemic lupus erythematosus (SLE) patients on belimumab therapy, and intra- and interindividual variations in plasma belimumab concentration were evaluated. A single-center prospective study was conducted at Oita University Hospital to evaluate trough plasma concentrations over the course of treatment in 13 SLE patients treated with intravenous belimumab. Plasma belimumab concentrations were measured by a validated ultra-high performance liquid chromatography with tandem mass spectrometry method. The median age of the patients was 40 (interquartile range: 35-51) years and the median weight was 51.8 (47.0-58.1) kg. A mean of 9.4 (range: 1-13) blood samples was collected per patient at routine visits. The mean (± SD) plasma belimumab concentration was 33.4 ± 11.9 μg/mL in the patient with the lowest concentration and 170.0 ± 16.6 μg/mL in the patient with the highest concentration, indicating a 5-fold difference between patients. On the other hand, the within-patient coefficient of variation ranged from 7.1% to 35.7%, showing no large variations. No significant correlation was observed between plasma belimumab concentration and belimumab dose (mg/kg) (Spearman's rank correlation coefficient = 0.22, p = .54). Examinations of trough plasma belimumab concentrations over the course of treatment in patients with SLE showed small intraindividual variation but large interindividual variation. Plasma belimumab trough concentration varied widely among patients administered the approved dose.
Collapse
MESH Headings
- Humans
- Lupus Erythematosus, Systemic/drug therapy
- Lupus Erythematosus, Systemic/blood
- Antibodies, Monoclonal, Humanized/pharmacokinetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/blood
- Adult
- Female
- Middle Aged
- Prospective Studies
- Male
- Immunosuppressive Agents/pharmacokinetics
- Immunosuppressive Agents/blood
- Immunosuppressive Agents/administration & dosage
- Immunosuppressive Agents/therapeutic use
- Tandem Mass Spectrometry
- Chromatography, High Pressure Liquid
- Drug Monitoring/methods
Collapse
Affiliation(s)
- Chisato Yoshijima
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| | - Yosuke Suzuki
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| | - Ryota Tanaka
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Hiroyuki Ono
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Ayako Oda
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| | - Takashi Ozaki
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of MedicineOita UniversityYufuOitaJapan
| | - Hirotaka Shibata
- Department of Endocrinology, Metabolism, Rheumatology and Nephrology, Faculty of MedicineOita UniversityYufuOitaJapan
| | - Hiroki Itoh
- Department of Clinical PharmacyOita University HospitalYufuOitaJapan
| | - Keiko Ohno
- Department of Medication Use Analysis and Clinical ResearchMeiji Pharmaceutical UniversityTokyoJapan
| |
Collapse
|
14
|
Antonova NP, Vasina DV, Grigoriev IV, Usachev EV, Aleshkin AV, Vorobev AM, Laishevtsev AI, Kapustin AV, Savinov VA, Anurova MN, Zackharova AA, Remizov TA, Makarov VV, Yudin SM, Gushchin VA. Pharmacokinetics and Preclinical Safety Studies of Modified Endolysin-based Gel for Topical Application. J Pharm Sci 2024; 113:2093-2100. [PMID: 38692487 DOI: 10.1016/j.xphs.2024.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/03/2024]
Abstract
Antibacterial therapy with phage-encoded endolysins or their modified derivatives with improved antibacterial, biochemical and pharmacokinetic properties is one of the most promising strategies that can supply existing antibacterial drugs array. Gram-negative bacteria-induced infections treatment is especially challenging because of rapidly spreading bacterial resistance. We have developed modified endolysin LysECD7-SMAP with a significant antibacterial activity and broad spectra of action against gram-negative bacteria. Endolysin was formulated in a bactericidal gel for topical application with pronounced effectivity in local animal infectious models. Here we present preclinical safety studies and pharmacokinetics of LysECD7-SMAP-based gel. We have detected LysECD7-SMAP in the skin and underlying muscle at therapeutic concentrations when the gel is applied topically to intact or injured skin. Moreover, the protein does not enter the bloodstream, and has no systemic bioavailability, assuming no systemic adverse effects. In studies of general toxicology, local tolerance, and immunotoxicology it was approved that LysECD7-SMAP gel local application results in the absence of toxic effects after single and multiple administration. Thus, LysECD7-SMAP-containing gel has appropriate pharmacokinetics and can be considered as safe that supports the initiation of the phase I clinical trials of novel antibacterial drug intending to treat acute wound infections caused by resistant gram-negative bacteria.
Collapse
Affiliation(s)
- Nataliia P Antonova
- Laboratory of Pathogen Population Variability Mechanisms, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia.
| | - Daria V Vasina
- Laboratory of Pathogen Population Variability Mechanisms, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Igor V Grigoriev
- Translational Biomedicine Laboratory, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Evgeny V Usachev
- Laboratory of Pathogen Population Variability Mechanisms, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Andrey V Aleshkin
- Laboratory of Clinical Microbiology and Biotechnology of Bacteriophages, G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, Moscow, Russia
| | - Aleksei M Vorobev
- Laboratory of Clinical Microbiology and Biotechnology of Bacteriophages, G.N. Gabrichevsky Moscow Research Institute for Epidemiology and Microbiology, Moscow, Russia
| | - Aleksei I Laishevtsev
- Federal State Budget Scientific Institution "Federal Scientific Centre VIEV" (FSC VIEV), Moscow, Russia
| | - Andrey V Kapustin
- Federal State Budget Scientific Institution "Federal Scientific Centre VIEV" (FSC VIEV), Moscow, Russia
| | - Vasiliy A Savinov
- Federal State Budget Scientific Institution "Federal Scientific Centre VIEV" (FSC VIEV), Moscow, Russia
| | - Mariia N Anurova
- Department of Pharmaceutical Technology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Anastasia A Zackharova
- Translational Biomedicine Laboratory, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Timofey A Remizov
- Translational Biomedicine Laboratory, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| | - Valentine V Makarov
- Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Moscow, Russia
| | - Sergey M Yudin
- Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, Moscow, Russia
| | - Vladimir A Gushchin
- Laboratory of Pathogen Population Variability Mechanisms, N.F. Gamaleya National Research Centre for Epidemiology and Microbiology, Ministry of Health of the Russian Federation, Moscow, Russia
| |
Collapse
|
15
|
Qahwaji R, Ashankyty I, Sannan NS, Hazzazi MS, Basabrain AA, Mobashir M. Pharmacogenomics: A Genetic Approach to Drug Development and Therapy. Pharmaceuticals (Basel) 2024; 17:940. [PMID: 39065790 PMCID: PMC11279827 DOI: 10.3390/ph17070940] [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: 05/06/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
The majority of the well-known pharmacogenomics research used in the medical sciences contributes to our understanding of medication interactions. It has a significant impact on treatment and drug development. The broad use of pharmacogenomics is required for the progress of therapy. The main focus is on how genes and an intricate gene system affect the body's reaction to medications. Novel biomarkers that help identify a patient group that is more or less likely to respond to a certain medication have been discovered as a result of recent developments in the field of clinical therapeutics. It aims to improve customized therapy by giving the appropriate drug at the right dose at the right time and making sure that the right prescriptions are issued. A combination of genetic, environmental, and patient variables that impact the pharmacokinetics and/or pharmacodynamics of medications results in interindividual variance in drug response. Drug development, illness susceptibility, and treatment efficacy are all impacted by pharmacogenomics. The purpose of this work is to give a review that might serve as a foundation for the creation of new pharmacogenomics applications, techniques, or strategies.
Collapse
Affiliation(s)
- Rowaid Qahwaji
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia; (R.Q.); (I.A.); (M.S.H.); (A.A.B.)
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ibraheem Ashankyty
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia; (R.Q.); (I.A.); (M.S.H.); (A.A.B.)
| | - Naif S. Sannan
- College of Applied Medical Sciences, King Saud bin Abdulaziz University for Health Sciences, Ar Rimayah, Riyadh 14611, Saudi Arabia;
- King Abdullah International Medical Research Center, Jeddah 22384, Saudi Arabia
| | - Mohannad S. Hazzazi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia; (R.Q.); (I.A.); (M.S.H.); (A.A.B.)
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ammar A. Basabrain
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia; (R.Q.); (I.A.); (M.S.H.); (A.A.B.)
- Hematology Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohammad Mobashir
- Department of Biomedical Laboratory Science, Faculty of Natural Sciences, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway
| |
Collapse
|
16
|
Alshammari FA. Exploring the Antibacterial Potential of Artemisia judaica Compounds Targeting the Hydrolase/Antibiotic Protein in Klebsiella pneumoniae: In Vitro and In Silico Investigations. Pharmaceuticals (Basel) 2024; 17:667. [PMID: 38931335 PMCID: PMC11207000 DOI: 10.3390/ph17060667] [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: 03/28/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 06/28/2024] Open
Abstract
Carbapenem antibiotic resistance is an emerging medical concern. Bacteria that possess the Klebsiella pneumoniae carbapenemase (KPC) protein, an enzyme that catalyzes the degradation of carbapenem antibiotics, have exhibited remarkable resistance to traditional and even modern therapeutic approaches. This study aimed to identify potential natural drug candidates sourced from the leaves of Artemisia judaica (A. judaica). The phytoconstituents present in A. judaica dried leaves were extracted using ethanol 80%. A reasonable amount of the extract was used to identify these phytochemicals via gas chromatography/mass spectrometry (GC/MS). One hundred twenty-two bioactive compounds from A. judaica were identified and subjected to docking analysis against the target bacterial protein. Four compounds (PubChem CID: 6917974, 159099, 628694, and 482788) were selected based on favorable docking scores (-9, -7.8, -7.7, and -7.5 kcal/mol). This computational investigation highlights the potential of these four compounds as promising antibacterial candidates against the specific KPC protein. Additionally, in vitro antibacterial assays using A. judaica extracts were conducted. The minimum inhibitory concentration (MIC) against the bacterium K. pneumonia was 125 μg/mL. Well-disk diffusion tests exhibited inhibition zones ranging from 10.3 ± 0.5 mm to 17 ± 0.5 mm at different concentrations, and time-kill kinetics at 12 h indicated effective inhibition of bacterial growth by A. judaica leaf extracts. Our findings have revealed the pharmaceutical potential of Artemisia judaica as a natural source for drug candidates against carbapenem-resistant pathogens.
Collapse
Affiliation(s)
- Fahdah Ayed Alshammari
- Department of Biology, College of Science, Northern Border University, Arar 76312, Saudi Arabia
| |
Collapse
|
17
|
Darbandi A, Abdi M, Dashtbin S, Yaghoubi S, Sholeh M, Kouhsari E, Darbandi T, Ghanavati R, Taheri B. Antibody-Antibiotic Conjugates: A Comprehensive Review on Their Therapeutic Potentials Against BacterialInfections. J Clin Lab Anal 2024; 38:e25071. [PMID: 38867639 PMCID: PMC11211676 DOI: 10.1002/jcla.25071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/09/2024] [Accepted: 05/09/2024] [Indexed: 06/14/2024] Open
Abstract
INTRODUCTION Antibodies are significant agents in the immune system and have proven to be effective in treating bacterial infections. With the advancement of antibody engineering in recent decades, antibody therapy has evolved widely. AIM This review aimed to investigate a new method as a therapeutic platform for the treatment of bacterial infections and explore the novel features of this method in conferring pathogen specificity to broad-spectrum antibiotics. MATERIAL AND METHODS A literature review was conducted addressing the following topics about antibody-antibiotic conjugates (AACs): (1) structure and mechanism of action; (2) clinical effectiveness; (3) advantages and disadvantages. RESULT Antibody conjugates are designed to build upon the progress made in the development of monoclonal antibodies for the treatment of diseases. Despite the growing emergence of antibiotic resistance among pathogenic bacteria worldwide, novel antimicrobials have not been sufficiently expanded to combat the global crisis of antibiotic resistance. A recently developed strategy for the treatment of infectious diseases is the use of AACs, which are specifically activated only in host cells. CONCLUSION A novel therapeutic AAC employs an antibody to deliver the antibiotic to the bacteria. The AACs can release potent antibacterial components that unconjugated forms may not exhibit with an appropriate therapeutic index. This review highlights how this science has guided the design principles of an impressive AAC and discusses how the AAC model promises to enhance the antibiotic effect against bacterial infections.
Collapse
Affiliation(s)
- Atieh Darbandi
- Molecular Microbiology Research CenterShahed UniversityTehranIran
| | - Milad Abdi
- Research Center of Tropical and Infectious DiseasesKerman University of Medical SciencesKermanIran
| | - Shirin Dashtbin
- Department of Microbiology, School of MedicineIran University of Medical SciencesTehranIran
| | - Sajad Yaghoubi
- Basic Sciences DepartmentNeyshabur University of Medical SciencesNeyshaburIran
| | - Mohammad Sholeh
- Department of BacteriologyPasteur Institute of IranTehranIran
| | - Ebrahim Kouhsari
- Laboratory Sciences Research CenterGolestan University of Medical SciencesGorganIran
| | - Talieh Darbandi
- Pharmaceutical Sciences BranchIslamic Azad University of Medical SciencesTehranIran
| | | | - Behrouz Taheri
- Department of Medical Biotechnology, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| |
Collapse
|
18
|
Pant T, Uche N, Juric M, Zielonka J, Bai X. Regulation of immunomodulatory networks by Nrf2-activation in immune cells: Redox control and therapeutic potential in inflammatory diseases. Redox Biol 2024; 70:103077. [PMID: 38359749 PMCID: PMC10877431 DOI: 10.1016/j.redox.2024.103077] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/26/2024] [Accepted: 02/05/2024] [Indexed: 02/17/2024] Open
Abstract
Inflammatory diseases present a serious health challenge due to their widespread prevalence and the severe impact on patients' lives. In the quest to alleviate the burden of these diseases, nuclear factor erythroid 2-related factor 2 (Nrf2) has emerged as a pivotal player. As a transcription factor intimately involved in cellular defense against metabolic and oxidative stress, Nrf2's role in modulating the inflammatory responses of immune cells has garnered significant attention. Recent findings suggest that Nrf2's ability to alter the redox status of cells underlies its regulatory effects on immune responses. Our review delves into preclinical and clinical evidence that underscores the complex influence of Nrf2 activators on immune cell phenotypes, particularly in the inflammatory milieu. By offering a detailed analysis of Nrf2's role in different immune cell populations, we cast light on the potential of Nrf2 activators in shaping the immune response towards a more regulated state, mitigating the adverse effects of inflammation through modeling redox status of immune cells. Furthermore, we explore the innovative use of nanoencapsulation techniques that enhance the delivery and efficacy of Nrf2 activators, potentially advancing the treatment strategies for inflammatory ailments. We hope this review will stimulate the development and expansion of Nrf2-targeted treatments that could substantially improve outcomes for patients suffering from a broad range of inflammatory diseases.
Collapse
Affiliation(s)
- Tarun Pant
- Department of Medicine, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA; Department of Pediatrics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| | - Nnamdi Uche
- Department of Pharmacology and Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Matea Juric
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Jacek Zielonka
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Xiaowen Bai
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
19
|
Kim A, Oh MS, Lee GH, Song S, Byun MS, Choi D, Yu BY, Lee H. Understanding the pharmacokinetic journey of Fc-fusion protein, rhIL-7-hyFc using complementary approach of two analytical methods, accelerator mass spectrometry and ELISA. Antib Ther 2024; 7:105-113. [PMID: 38566969 PMCID: PMC10983079 DOI: 10.1093/abt/tbae004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/21/2024] [Accepted: 01/28/2024] [Indexed: 04/04/2024] Open
Abstract
Antibody-based therapeutics (ABTs), including monoclonal/polyclonal antibodies and fragment crystallizable region (Fc)-fusion proteins, are increasingly used in disease treatment, driving the global market growth. Understanding the pharmacokinetic (PK) properties of ABTs is crucial for their clinical effectiveness. This study investigated the PK profile and tissue distribution of efineptakin alfa, a long-acting recombinant human interleukin-7 (rhIL-7-hyFc), using enzyme-linked immunosorbent assay (ELISA) and accelerator mass spectrometry (AMS). Totally, four rats were injected intramuscularly with 1 mg/kg of rhIL-7-hyFc containing 14C-rhIL-7-hyFc, which was prepared via reductive methylation. Serum total radioactivity (TRA) and serum rhIL-7-hyFc concentrations were quantified using AMS and ELISA, respectively. The TRA concentrations in organs were determined by AMS. Serum TRA peaked at 10 hours with a terminal half-life of 40 hours. The rhIL-7-hyFc exhibited a mean peak concentration at around 17 hours and a rapid elimination with a half-life of 12.3 hours. Peak concentration and area under the curve of TRA were higher than those of rhIL-7-hyFc. Tissue distribution analysis showed an elevated TRA concentrations in lymph nodes, kidneys, and spleen, indicating rhIL-7-hyFc's affinity for these organs. The study also simulated the positions of 14C labeling in rhIL-7-hyFc, identifying specific residues in the fragment of rhIL-7 portion, and provided the explanation of distinct analytes targeted by each method. Combining ELISA and AMS provided advantages by offering sensitivity and specificity for quantification as well as enabling the identification of analyte forms. The integrated use of ELISA and AMS offers valuable insights for the development and optimization of ABT.
Collapse
Affiliation(s)
- Anhye Kim
- Department of Clinical Pharmacology and Therapeutics, CHA Bundang Medical Center, CHA University, Seongnam 13496, Republic of Korea
- Department of Biomedical Informatics, CHA University School of Medicine, CHA University, Seongnam 13488, Republic of Korea
- Institute for Biomedical Informatics, CHA University School of Medicine, CHA University, Seongnam 13488, Republic of Korea
| | - Min-Seok Oh
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of Stem Cell Biology, School of Medicine, Konkuk University, Seoul 05029, Republic of Korea
| | - Gwan-Ho Lee
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Seongeun Song
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
- Department of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon 21983, Republic of Korea
| | - Mi-sun Byun
- Clinical Development Division, Genexine, Inc., Seoul 07789, Republic of Korea
| | - Donghoon Choi
- Research Institute, NeoImmuneTech, co. Ltd., Pohang 37666, Republic of Korea
| | - Byung-Yong Yu
- Research Resources Division, Advanced Analysis and Data Center, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Howard Lee
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea
- Department of Clinical Pharmacology and Therapeutics, Seoul National University Hospital, Seoul 03080, Republic of Korea
- Advanced Institute of Convergence Technology, Suwon 16229, Republic of Korea
| |
Collapse
|
20
|
Alshehri MM, Kumar N, Kuthi NA, Olaide Z, Alshammari MK, Bello RO, Alghazwni MK, Alshehri AM, Alshlali OM, Ashimiyu-Abdusalam Z, Umar HI. Computer-aided drug discovery of c-Abl kinase inhibitors from plant compounds against chronic myeloid leukemia. J Biomol Struct Dyn 2024:1-21. [PMID: 38517058 DOI: 10.1080/07391102.2024.2329297] [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: 08/21/2023] [Accepted: 03/06/2024] [Indexed: 03/23/2024]
Abstract
Chronic myeloid leukemia (CML) is a hematological malignancy characterized by the neoplastic transformation of hematopoietic stem cells, driven by the Philadelphia (Ph) chromosome resulting from a translocation between chromosomes 9 and 22. This Ph chromosome harbors the breakpoint cluster region (BCR) and the Abelson (ABL) oncogene (BCR-ABL1) which have a constitutive tyrosine kinase activity. However, the tyrosine kinase activity of BCR-ABL1 have been identified as a key player in CML initiation and maintenance through c-Abl kinase. Despite advancements in tyrosine kinase inhibitors, challenges such as efficacy, safety concerns, and recurring drug resistance persist. This study aims to discover potential c-Abl kinase inhibitors from plant compounds with anti-leukemic properties, employing drug-likeness assessment, molecular docking, in silico pharmacokinetics (ADMET) screening, density function theory (DFT), and molecular dynamics simulations (MDS). Out of 58 screened compounds for drug-likeness, 44 were docked against c-Abl kinase. The top hit compound (isovitexin) and nilotinib (control drug) were subjected to rigorous analyses, including ADMET profiling, DFT evaluation, and MDS for 100 ns. Isovitexin demonstrated a notable binding affinity (-15.492 kcal/mol), closely comparable to nilotinib (-16.826 kcal/mol), showcasing a similar binding pose and superior structural stability and reactivity. While these findings suggest isovitexin as a potential c-Abl kinase inhibitor, further validation through urgent in vitro and in vivo experiments is imperative. This research holds promise for providing an alternative avenue to address existing CML treatment and management challenges.
Collapse
Affiliation(s)
- Mohammed M Alshehri
- Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Neeraj Kumar
- Department of Pharmaceutical Chemistry, Bhupal Nobles' College of Pharmacy, Udaipur, India
| | - Najwa Ahmad Kuthi
- Department of Chemistry, Faculty of Science, Universiti Teknologi Malaysia (UTM), Johor, Malaysia
| | - Zainab Olaide
- Department of Biochemistry, Ibrahim Badamasi Babangida University, Lapai, Nigeria
| | | | - Ridwan Opeyemi Bello
- Computer-Aided Therapeutic Discovery and Design Platform, Federal University of Technology, Akure, Nigeria
| | | | | | | | - Zainab Ashimiyu-Abdusalam
- Computer-Aided Therapeutic Discovery and Design Platform, Federal University of Technology, Akure, Nigeria
- Department of Biochemistry and Nutrition, Nigerian Institute of Medical Research, Yaba, Nigeria
| | - Haruna Isiyaku Umar
- Computer-Aided Therapeutic Discovery and Design Platform, Federal University of Technology, Akure, Nigeria
- Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| |
Collapse
|
21
|
Ait-Oudhia S, Wang YM, Dosne AG, Roy A, Jin JY, Shen J, Kagan L, Musuamba FT, Zhang L, Kijima S, Gastonguay MR, Ouellet D. Challenging the Norm: A Multidisciplinary Perspective on Intravenous to Subcutaneous Bridging Strategies for Biologics. Clin Pharmacol Ther 2024; 115:412-421. [PMID: 38069528 DOI: 10.1002/cpt.3133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023]
Abstract
The transition from intravenous (i.v.) to subcutaneous (s.c.) administration of biologics is a critical strategy in drug development aimed at improving patient convenience, compliance, and therapeutic outcomes. Focusing on the increasing role of model-informed drug development (MIDD) in the acceleration of this transition, an in-depth overview of the essential clinical pharmacology, and regulatory considerations for successful i.v. to s.c. bridging for biologics after the i.v. formulation has been approved are presented. Considerations encompass multiple aspects beginning with adequate pharmacokinetic (PK) and pharmacodynamic (i.e., exposure-response) evaluations which play a vital role in establishing comparability between the i.v. and s.c. routes of administrations. Selected key recommendations and points to consider include: (i) PK characterization of the s.c. formulation, supported by the increasing preclinical understanding of the s.c. absorption, and robust PK study design and analyses in humans; (ii) a thorough characterization of the exposure-response profiles including important metrics of exposure for both efficacy and safety; (iii) comparability studies designed to meet regulatory considerations and support approval of the s.c. formulation, including noninferiority studies with PK and/or efficacy and safety as primary end points; and (iv) comprehensive safety package addressing assessments of immunogenicity and patients' safety profile with the new route of administration. Recommendations for successful bridging strategies are evolving and MIDD approaches have been used successfully to accelerate the transition to s.c. dosing, ultimately leading to improved patient experiences, adherence, and clinical outcomes.
Collapse
Affiliation(s)
| | - Yow-Ming Wang
- US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Anne-Gaelle Dosne
- Janssen Research & Development, LLC, Beerse, Belgium
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Amit Roy
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Jin Y Jin
- Genentech Inc., South San Francisco, California, USA
| | - Jun Shen
- Bristol Myers Squibb, Princeton, New Jersey, USA
| | - Leonid Kagan
- Department of Pharmaceutics and Center of Excellence for Pharmaceutical Translational Research and Education, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Flora T Musuamba
- Belgian Federal Agency for Medicines and Health Products, Brussels, Belgium
- NAmur Research Institute for LIfe Sciences (NARILIS), University of Namur, Namur, Belgium
| | - Lucia Zhang
- Health Canada, Biologic and Radiopharmaceutical Drugs Directorate, Ottawa, Ontario, Canada
| | - Shinichi Kijima
- Pharmaceuticals and Medical Devices Agency (PMDA), Tokyo, Japan
| | | | - Daniele Ouellet
- Janssen Research & Development, LLC, Spring House, Pennsylvania, USA
| |
Collapse
|
22
|
Tyagi S, Mishra R, Mazumder A, Mazumder R, Singh G, Pandey P. Synthesis, in silico screening, and biological evaluation of novel pyridine congeners as anti-epileptic agents targeting AMPA (α-amino-3-hydroxy-5-methylisoxazole) receptors. Chem Biol Drug Des 2024; 103:e14498. [PMID: 38453241 DOI: 10.1111/cbdd.14498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 12/23/2023] [Accepted: 02/26/2024] [Indexed: 03/09/2024]
Abstract
The research involves the synthesis of a series of new pyridine analogs 5(i-x) and their evaluation for anti-epileptic potential using in silico and in vivo models. Synthesis of the compounds was accomplished by using the Vilsmeier-Haack reaction principle. AutoDock 4.2 was used for their in silico screening against AMPA (-amino-3-hydroxy-5-methylisoxazole) receptor (PDB ID:3m3f). For in vivo testing, the maximal electroshock seizure (MES) model was used. The physicochemical, pharmacokinetic, drug-like, and drug-score features of all synthesized compounds were assessed using the online Swiss ADME and Protein Plus software. The in silico results showed that all the synthesized compounds 5(i-x) had 1-3 interactions and affinities ranging from -6.5 to -8.0 kJ/mol with the targeted receptor compared to the binding affinities of the standard drug phenytoin and the original ligand of the target (P99), which were -7.6 and -6.8 kJ/mol, respectively. In vivo study results showed that the compound 5-Carbamoyl-2-formyl-1-[2-(4-nitrophenyl)-2-oxo-ethyl]-pyridinium gave 60% protection against epileptic seizures compared to 59% protection afforded by regular phenytoin. All of them met Lipinski's rule of five and had drug-likeness and drug score values of 0.55 and 0.8, respectively, making them chemically and functionally like phenytoin. According to the findings of the studies, the synthesized derivatives have the potential to be employed as a stepping stone in the development of novel anti-epileptic drugs.
Collapse
Affiliation(s)
- Shivani Tyagi
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Rakhi Mishra
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Rupa Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
| | - Gurvinder Singh
- School of Pharmaceutical Science, Lovely Professional University, Phagwara, Punjab, India
| | - Pratibha Pandey
- Noida Institute of Engineering and Technology (Pharmacy Institute), Greater Noida, Uttar Pradesh, India
- Noida Institute of Engineering and Technology, Biotechnology Department, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
23
|
Mourya A, Prajapati N. Precision Deuteration in Search of Anticancer Agents: Approaches to Cancer Drug Discovery. Cancer Biother Radiopharm 2024; 39:1-18. [PMID: 37585602 DOI: 10.1089/cbr.2023.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023] Open
Abstract
Cancer chemotherapy has been shifted from conventional cytotoxic drug therapy to selective and target-specific therapy after the findings about DNA changes and proteins that are responsible for cancer. A large number of newer drugs were discovered as targeted therapy for particular types of neoplastic disease. The initial discovery includes the development of the first in the category, imatinib, a Bcr-Abl tyrosine kinase inhibitor (TKI) for the treatment of chronic myelocytic leukemia in 2001. But the joy did not last for long as the drug developed a point mutation within the ABL1 kinase domain of BCR-ABL1, which subsequently led to the discovery of many other TKIs. Resistance was observed for newer TKIs a few years after their launching, but the use of TKIs in life-threatening cancer therapy is considered as far better compared with the risks of disease because of its target specificity and hence less toxicity. In search of a better anticancer agent, the physiochemical properties of the lead molecule have been modified for its efficacy toward disease and delay in the development of resistance. Deuteration in the drug molecule is one of such modifications that alter the pharmacokinetic properties, generally its metabolism, as compared with its pharmacodynamic effects. Precision deuteration in many anticancer drugs has been carried out to search for better drugs for cancer. In this review, the majority of anticancer drugs and molecules for which deuteration was applied to get better anticancer molecules were discussed. This review will provide a complete guide about the benefits of deuteration in cancer chemotherapy.
Collapse
MESH Headings
- Humans
- Drug Resistance, Neoplasm/genetics
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Imatinib Mesylate/therapeutic use
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Drug Discovery
Collapse
Affiliation(s)
- Aman Mourya
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, India
| | - Navnit Prajapati
- Faculty of Pharmacy, The Maharaja Sayajirao University of Baroda, Vadodara, India
| |
Collapse
|
24
|
Singh S, Singh PK, Sachan K, Kumar M, Bhardwaj P. Automation of Drug Discovery through Cutting-edge In-silico Research in Pharmaceuticals: Challenges and Future Scope. Curr Comput Aided Drug Des 2024; 20:723-735. [PMID: 37807412 DOI: 10.2174/0115734099260187230921073932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/05/2023] [Accepted: 08/18/2023] [Indexed: 10/10/2023]
Abstract
The rapidity and high-throughput nature of in silico technologies make them advantageous for predicting the properties of a large array of substances. In silico approaches can be used for compounds intended for synthesis at the beginning of drug development when there is either no or very little compound available. In silico approaches can be used for impurities or degradation products. Quantifying drugs and related substances (RS) with pharmaceutical drug analysis (PDA) can also improve drug discovery (DD) by providing additional avenues to pursue. Potential future applications of PDA include combining it with other methods to make insilico predictions about drugs and RS. One possible outcome of this is a determination of the drug potential of nontoxic RS. ADME estimation, QSAR research, molecular docking, bioactivity prediction, and toxicity testing all involve impurity profiling. Before committing to DD, RS with minimal toxicity can be utilised in silico. The efficacy of molecular docking in getting a medication to market is still debated despite its refinement and improvement. Biomedical labs and pharmaceutical companies were hesitant to adopt molecular docking algorithms for drug screening despite their decades of development and improvement. Despite the widespread use of "force fields" to represent the energy exerted within and between molecules, it has been impossible to reliably predict or compute the binding affinities between proteins and potential binding medications.
Collapse
Affiliation(s)
- Smita Singh
- Department of Pharmaceutics, SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology, Delhi NCR Campus, Modinagar, Ghaziabad, India
| | - Pranjal Kumar Singh
- Department of Pharmaceutics, SRM Modinagar College of Pharmacy, SRM Institute of Science and Technology, Delhi NCR Campus, Modinagar, Ghaziabad, India
| | - Kapil Sachan
- KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, India
| | - Mukesh Kumar
- IIMT College of Medical Sciences, IIMT University, Ganga Nagar, Meerut, India
| | - Poonam Bhardwaj
- NKBR College of Pharmacy and Research Center, Phaphunda, Meerut, India
| |
Collapse
|
25
|
Bei R, Thomas J, Kapur S, Woldeyes M, Rauk A, Robarge J, Feng J, Abbou Oucherif K. Predicting the clinical subcutaneous absorption rate constant of monoclonal antibodies using only the primary sequence: a machine learning approach. MAbs 2024; 16:2352887. [PMID: 38745390 PMCID: PMC11110684 DOI: 10.1080/19420862.2024.2352887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/03/2024] [Indexed: 05/16/2024] Open
Abstract
Subcutaneous injections are an increasingly prevalent route of administration for delivering biological therapies including monoclonal antibodies (mAbs). Compared with intravenous delivery, subcutaneous injections reduce administration costs, shorten the administration time, and are strongly preferred from a patient experience point of view. An understanding of the absorption process of a mAb from the injection site to the systemic circulation is critical to the process of subcutaneous mAb formulation development. In this study, we built a model to predict the absorption rate constant (ka), which denotes how fast a mAb is absorbed from the site of administration. Once trained, our model (enabled by the XGBoost algorithm in machine learning) can predict the ka of a mAb following a subcutaneous injection using in silico molecular properties alone (generated from the primary sequence). Our model does not need clinically observed plasma concentration-time data; this is a novel capability not previously achieved in predictive pharmacokinetic models. The model also showed improved performance when benchmarked against a recently reported mechanistic model that relied on clinical data to predict subcutaneous absorption of mAbs. We further interpreted the model to understand which molecular properties affect the absorption rate and showed that our findings are consistent with previous studies evaluating subcutaneous absorption through direct experimentation. Taken altogether, this study reports the development, validation, benchmarking, and interpretation of a model that can predict the clinical ka of a mAb using its primary sequence as the only input.
Collapse
Affiliation(s)
- Ronghua Bei
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Justin Thomas
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Shiven Kapur
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Mahlet Woldeyes
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Adam Rauk
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jason Robarge
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Jiangyan Feng
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| | - Kaoutar Abbou Oucherif
- Lilly Research Laboratories, Lilly Corporate Center, Eli Lilly and Company, Indianapolis, IN, USA
| |
Collapse
|
26
|
Rahban M, Ahmad F, Piatyszek MA, Haertlé T, Saso L, Saboury AA. Stabilization challenges and aggregation in protein-based therapeutics in the pharmaceutical industry. RSC Adv 2023; 13:35947-35963. [PMID: 38090079 PMCID: PMC10711991 DOI: 10.1039/d3ra06476j] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/30/2023] [Indexed: 04/26/2024] Open
Abstract
Protein-based therapeutics have revolutionized the pharmaceutical industry and become vital components in the development of future therapeutics. They offer several advantages over traditional small molecule drugs, including high affinity, potency and specificity, while demonstrating low toxicity and minimal adverse effects. However, the development and manufacturing processes of protein-based therapeutics presents challenges related to protein folding, purification, stability and immunogenicity that should be addressed. These proteins, like other biological molecules, are prone to chemical and physical instabilities. The stability of protein-based drugs throughout the entire manufacturing, storage and delivery process is essential. The occurrence of structural instability resulting from misfolding, unfolding, and modifications, as well as aggregation, poses a significant risk to the efficacy of these drugs, overshadowing their promising attributes. Gaining insight into structural alterations caused by aggregation and their impact on immunogenicity is vital for the advancement and refinement of protein therapeutics. Hence, in this review, we have discussed some features of protein aggregation during production, formulation and storage as well as stabilization strategies in protein engineering and computational methods to prevent aggregation.
Collapse
Affiliation(s)
- Mahdie Rahban
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences Kerman Iran
| | - Faizan Ahmad
- Department of Biochemistry, School of Chemical & Life Sciences, Jamia Hamdard New Delhi-110062 India
| | | | | | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University Rome Italy
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran Tehran 1417614335 Iran +9821 66404680 +9821 66956984
| |
Collapse
|
27
|
Buchanan D, Pham AM, Singh SK, Panda SS. Molecular Hybridization of Alkaloids Using 1,2,3-Triazole-Based Click Chemistry. Molecules 2023; 28:7593. [PMID: 38005315 PMCID: PMC10674395 DOI: 10.3390/molecules28227593] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 10/30/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Alkaloids found in multiple species, known as 'driver species', are more likely to be included in early-stage drug development due to their high biodiversity compared to rare alkaloids. Many synthetic approaches have been employed to hybridize the natural alkaloids in drug development. Click chemistry is a highly efficient and versatile reaction targeting specific areas, making it a valuable tool for creating complex natural products and diverse molecular structures. It has been used to create hybrid alkaloids that address their limitations and serve as potential drugs that mimic natural products. In this review, we highlight the recent advancements made in modifying alkaloids using click chemistry and their potential medicinal applications. We discuss the significance, current trends, and prospects of click chemistry in natural product-based medicine. Furthermore, we have employed computational methods to evaluate the ADMET properties and drug-like qualities of hybrid molecules.
Collapse
Affiliation(s)
- Devan Buchanan
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA 30912, USA; (D.B.); (A.M.P.)
| | - Ashley M. Pham
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA 30912, USA; (D.B.); (A.M.P.)
| | - Sandeep K. Singh
- Jindal Global Business School, OP Jindal Global University, Sonipat 131001, India;
| | - Siva S. Panda
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA 30912, USA; (D.B.); (A.M.P.)
- Department Biochemistry and Molecular Biology, Augusta University Augusta, GA 30912, USA
| |
Collapse
|
28
|
Rajendran D, Chandrasekaran N. Molecular Interaction of Functionalized Nanoplastics with Human Hemoglobin. J Fluoresc 2023; 33:2257-2272. [PMID: 37014521 DOI: 10.1007/s10895-023-03221-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
Humans are exposed to excessive nanoplastics (NPs) which have ample affinity for globular proteins. We investigated the interaction of functionalized polystyrene nanoplastics (plain: PS, carboxy: PS-COOH, and amine: PS-NH2) with human hemoglobin (Hb) utilizing multi-spectroscopic and docking approaches to acquire insights into molecular aspects of binding mechanism, which will be helpful in assessing the toxicokinetics or toxicodynamics of nanoplastics NPs. Hypsochromicity and hypochromicity were observed invariably in all the spectra (steady-state fluorescence emission, synchronous and three-dimensional) for all complexes, among which PS-NH2 binds effectively and changes the Hb's conformation by enhancing hydrophobicity around aromatic residues, notably tryptophan. All the NPs bind with the hydrophobic pocket of B-chain in Hb, where PS and PS-NH2 bind via hydrophobic force while PS-COOH binds via hydrogen bonding (predominantly) and van der Waals force, consistent validated with docking results. The minimal shift in absorbance peak also indicates enhanced hydrophobicity by PS-NH2 with larger aggregation as demonstrated in resonance light scattering. The amide band's shift, secondary structural analysis, and presence of characteristic functional group peaks in complexes in Infra-Red spectra confirm the structural changes in the protein. As seen in field emission scanning microscopy images, NPs penetrate the surface of proteins. These findings conclude that polystyrene NPs interact with Hb, causing structural alterations that may affect functional characteristics as well, with the greatest effect being in the order: PS-NH2>PS-COOH>PS.
Collapse
Affiliation(s)
- Durgalakshmi Rajendran
- Centre for Nanobiotechnology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632014, India
| | - Natarajan Chandrasekaran
- Centre for Nanobiotechnology, Vellore Institute of Technology, Tamil Nadu, Vellore, 632014, India.
| |
Collapse
|
29
|
Bhuia MS, Rokonuzzman M, Hossain MI, Ansari SA, Ansari IA, Islam T, Al Hasan MS, Mubarak MS, Islam MT. Anxiolytic- like Effects by trans-Ferulic Acid Possibly Occur through GABAergic Interaction Pathways. Pharmaceuticals (Basel) 2023; 16:1271. [PMID: 37765079 PMCID: PMC10535412 DOI: 10.3390/ph16091271] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 09/02/2023] [Accepted: 09/05/2023] [Indexed: 09/29/2023] Open
Abstract
Numerous previous studies reported that ferulic acid exerts anxiolytic activity. However, the mechanisms have yet to be elucidated. The current study aimed to investigate the anxiolytic effect of trans-ferulic acid (TFA), a stereoisomer of ferulic acid, and evaluated its underlying mechanism using in vivo and computational studies. For this, different experimental doses of TFA (25, 50, and 75 mg/kg) were administered orally to Swiss albino mice, and various behavioral methods of open field, hole board, swing box, and light-dark tests were carried out. Diazepam (DZP), a positive allosteric modulator of the GABAA receptor, was employed as a positive control at a dose of 2 mg/kg, and distilled water served as a vehicle. Additionally, molecular docking was performed to estimate the binding affinities of the TFA and DZP toward the GABAA receptor subunits of α2 and α3, which are associated with the anxiolytic effect; visualizations of the ligand-receptor interaction were carried out using various computational tools. Our findings indicate that TFA dose-dependently reduces the locomotor activity of the animals in comparison with the controls, calming their behaviors. In addition, TFA exerted the highest binding affinity (-5.8 kcal/mol) to the α2 subunit of the GABAA receptor by forming several hydrogen and hydrophobic bonds. Taken together, our findings suggest that TFA exerts a similar effect to DZP, and the compound exerts moderate anxiolytic activity through the GABAergic interaction pathway. We suggest further clinical studies to develop TFA as a reliable anxiolytic agent.
Collapse
Affiliation(s)
- Md. Shimul Bhuia
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (M.R.); (M.I.H.); (T.I.); (M.S.A.H.)
| | - Md. Rokonuzzman
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (M.R.); (M.I.H.); (T.I.); (M.S.A.H.)
| | - Md. Imran Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (M.R.); (M.I.H.); (T.I.); (M.S.A.H.)
| | - Siddique Akber Ansari
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia;
| | - Irfan Aamer Ansari
- Department of Drug Science and Technology, University of Turin, 10124 Turin, Italy;
| | - Tawhida Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (M.R.); (M.I.H.); (T.I.); (M.S.A.H.)
| | - Md. Sakib Al Hasan
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (M.R.); (M.I.H.); (T.I.); (M.S.A.H.)
| | | | - Muhammad Torequl Islam
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh; (M.S.B.); (M.R.); (M.I.H.); (T.I.); (M.S.A.H.)
| |
Collapse
|
30
|
Tanaka Y, Kawanishi M, Nakanishi M, Yamasaki H, Takeuchi T. Efficacy and safety of anti-TNF multivalent NANOBODY® compound 'ozoralizumab' without methotrexate co-administration in patients with active rheumatoid arthritis: A 52-week result of phase III, randomised, open-label trial (NATSUZORA trial). Mod Rheumatol 2023; 33:875-882. [PMID: 36201360 DOI: 10.1093/mr/roac126] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 09/19/2022] [Accepted: 09/28/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVES The aim is to assess the efficacy and safety of a 52-week subcutaneous ozoralizumab treatment at 30 and 80 mg without methotrexate (MTX) in active rheumatoid arthritis. METHODS This randomised, open-label, multicentre phase III trial randomly allocated 140 patients in 2:1 ratio as subcutaneous ozoralizumab at 30 or 80 mg every 4 weeks for 52 weeks without MTX. RESULTS Both groups administered ozoralizumab at 30 and 80 mg showed good clinical improvement. The American College of Rheumatology response rates were high at Week 24 and maintained through 52 weeks. The ozoralizumab groups also showed good improvement in other end points, and improvements observed from Week 1 were maintained through 52 weeks. Improvements in many efficacy assessments were similar between doses. No deaths were reported, and serious adverse events occurred in a total of 20 patients in the ozoralizumab groups. Increased antidrug antibodies were observed in approximately 40% of patients in the ozoralizumab groups, and 27.7% of the patients in the 30 mg group were neutralising antibody-positive. CONCLUSIONS Ozoralizumab, at 30 and 80 mg, demonstrated significant therapeutic effects without MTX, and the efficacy was maintained for 52 weeks with active rheumatoid arthritis. Ozoralizumab showed an acceptable tolerability profile over 52 weeks.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Japan, Kitakyusyu, Japan
| | | | - Megumi Nakanishi
- Development Headquarters, Taisho Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Hironori Yamasaki
- Development Headquarters, Taisho Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Tsutomu Takeuchi
- Keio University School of Medicine, Tokyo, Japan
- Saitama Medical University, Saitama, Japan
| |
Collapse
|
31
|
Tanaka Y, Kawanishi M, Nakanishi M, Yamasaki H, Takeuchi T. Efficacy and safety of the anti-TNF multivalent NANOBODY® compound ozoralizumab in patients with rheumatoid arthritis and an inadequate response to methotrexate: A 52-week result of a Phase II/III study (OHZORA trial). Mod Rheumatol 2023; 33:883-890. [PMID: 36197757 DOI: 10.1093/mr/roac119] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/19/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To assess the efficacy and safety through a 52-week treatment with subcutaneous ozoralizumab at 30 or 80 mg in patients with active rheumatoid arthritis despite methotrexate therapy. METHODS This multicentre, randomized, placebo-controlled, double-blind, parallel-group confirmatory trial included a 24-week double-blind treatment period followed by a 28-week open-label treatment period. The double-blind treatment period randomized 381 (2:2:1) patients to placebo and ozoralizumab at 30 or 80 mg, and patients receiving placebo were re-randomized (1:1) to ozoralizumab at 30 or 80 mg in the open-label period. RESULTS The ozoralizumab groups showed good clinical improvement, with high American College of Rheumatology response rates at 52 weeks, as well as good improvements in other endpoints, which were observed from Day 3 and maintained through Week 52. Furthermore, the ozoralizumab groups showed a high remission rate in clinical and functional remission at Week 52. Serious adverse events occurred in a total of 23 patients in the ozoralizumab groups, without differences in incidence between doses. CONCLUSIONS Ozoralizumab demonstrated significant therapeutic effects and efficacy, which was maintained for 52 weeks. The safety profile was consistent with the evaluated results in interim analysis at Week 24, and ozoralizumab was well-tolerated up to Week 52.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- The First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | | | - Megumi Nakanishi
- Development Headquarters, Taisho Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Hironori Yamasaki
- Development Headquarters, Taisho Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Tsutomu Takeuchi
- Keio University School of Medicine, Tokyo, Japan
- Saitama Medical University, Saitama, Japan
| |
Collapse
|
32
|
Niu J, Wang W, Ouellet D. Mechanism-based pharmacokinetic and pharmacodynamic modeling for bispecific antibodies: challenges and opportunities. Expert Rev Clin Pharmacol 2023; 16:977-990. [PMID: 37743720 DOI: 10.1080/17512433.2023.2257136] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023]
Abstract
INTRODUCTION Unlike conventional antibodies, bispecific antibodies (bsAbs) are engineered antibody- or antibody fragment-based molecules that can simultaneously recognize two different epitopes or antigens. Over the past decade, there has been an explosion of bsAbs being developed across therapeutic areas. Development of bsAbs presents unique challenges and mechanism-based pharmacokinetic/pharmacodynamic (PK/PD) modeling has served as a powerful tool to optimize their development and realize their clinical utility. AREAS COVERED In this review, the guiding principles and case examples of how fit-for-purpose, mechanism-based PK/PD models have been applied to answer questions commonly encountered in bsAb development are presented. Such models characterize the key pharmacological elements of bsAbs, and they can be utilized for model-informed drug development. We also include the discussion of challenges, knowledge gaps and future direction for such models. EXPERT OPINION Mechanistic PK/PD modeling is a powerful tool to support the development of bsAbs. These models can be extrapolated to predict treatment outcomes based on mechanisms of action (MoA) and clinical observations to form positive learn-and-confirm cycles during drug development, due to their abilities to differentiate system- and drug-specific parameters. Meanwhile, the models should keep being adapted according to novel drug design and MoA, providing continuous opportunities for model-informed drug development.
Collapse
Affiliation(s)
- Jin Niu
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, Spring House, PA, USA
| | - Weirong Wang
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, Spring House, PA, USA
| | - Daniele Ouellet
- Clinical Pharmacology and Pharmacometrics, Janssen Research & Development, Spring House, PA, USA
| |
Collapse
|
33
|
Peng H, Wang J, Chen J, Peng Y, Wang X, Chen Y, Kaplan DL, Wang Q. Challenges and opportunities in delivering oral peptides and proteins. Expert Opin Drug Deliv 2023; 20:1349-1369. [PMID: 37450427 PMCID: PMC10990675 DOI: 10.1080/17425247.2023.2237408] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION Rapid advances in bioengineering enable the use of complex proteins as therapeutic agents to treat diseases. Compared with conventional small molecule drugs, proteins have multiple advantages, including high bioactivity and specificity with low toxicity. Developing oral dosage forms with active proteins is a route to improve patient compliance and significantly reduce production costs. However, the gastrointestinal environment remains a challenge to this delivery path due to enzymatic degradation, low permeability, and weak absorption, leading to reduced delivery efficiency and poor clinical outcomes. AREAS COVERED This review describes the barriers to oral delivery of peptides and complex proteins, current oral delivery strategies utilized and the opportunities and challenges ahead to try and circumvent these barriers. Oral protein drugs on the market and clinical trials provide insights and approaches for advancing delivery strategies. EXPERT OPINION Although most current studies on oral protein delivery rely on in vitro and in vivo animal data, the safety and limitations of the approach in humans remain uncertain. The shortage of clinical data limits the development of new or alternative strategies. Therefore, designing appropriate oral delivery strategies remains a significant challenge and requires new ideas, innovative design strategies and novel model systems.
Collapse
Affiliation(s)
- Haisheng Peng
- Department of Pharmacology, Medical College, University of Shaoxing, Shaoxing, China
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| | - Jiahe Wang
- Department of Humanities, Daqing Branch, Harbin Medical University, Daqing, China
| | - Jiayu Chen
- Department of Pharmacology, Medical College, University of Shaoxing, Shaoxing, China
| | - Yanbo Peng
- Department of Pharmaceutical Engineering, China Pharmaceutical University, 639 Longmian Rd, Nanjing 211198, China
| | - Xiaoxian Wang
- The Affiliated Hospital of Medical College, University of Shaoxing, Shaoxing, Zhejiang Province, China
| | - Ying Chen
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA, 02155, USA
| | - Qun Wang
- Department of Chemical and Biological Engineering, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
34
|
Datta-Mannan A, Molitoris BA, Feng Y, Martinez MM, Sandoval RM, Brown RM, Merkel D, Croy JE, Dunn KW. Intravital Microscopy Reveals Unforeseen Biodistribution Within the Liver and Kidney Mechanistically Connected to the Clearance of a Bifunctional Antibody. Drug Metab Dispos 2023; 51:403-412. [PMID: 36460476 PMCID: PMC11022859 DOI: 10.1124/dmd.122.001049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 10/16/2022] [Accepted: 11/07/2022] [Indexed: 12/03/2022] Open
Abstract
Bifunctional antibody (BfAb) therapeutics offer the potential for novel functionalities beyond those of the individual monospecific entities. However, combining these entities into a single molecule can have unpredictable effects, including changes in pharmacokinetics that limit the compound's therapeutic profile. A better understanding of how molecular modifications affect in vivo tissue interactions could help inform BfAb design. The present studies were predicated on the observation that a BfAb designed to have minimal off-target interactions cleared from the circulation twice as fast as the monoclonal antibody (mAb) from which it was derived. The present study leverages the spatial and temporal resolution of intravital microscopy (IVM) to identify cellular interactions that may explain the different pharmacokinetics of the two compounds. Disposition studies of mice demonstrated that radiolabeled compounds distributed similarly over the first 24 hours, except that BfAb accumulated approximately two- to -three times more than mAb in the liver. IVM studies of mice demonstrated that both distributed to endosomes of liver endothelia but with different kinetics. Whereas mAb accumulated rapidly within the first hour of administration, BfAb accumulated only modestly during the first hour but continued to accumulate over 24 hours, ultimately reaching levels similar to those of the mAb. Although neither compound was freely filtered by the mouse or rat kidney, BfAb, but not mAb, was found to accumulate over 24 hours in endosomes of proximal tubule cells. These studies demonstrate how IVM can be used as a tool in drug design, revealing unpredicted cellular interactions that are undetectable by conventional analyses. SIGNIFICANCE STATEMENT: Bifunctional antibodies offer novel therapeutic functionalities beyond those of the individual monospecific entities. However, combining these entities into a single molecule can have unpredictable effects, including undesirable changes in pharmacokinetics. Studies of the dynamic distribution of a bifunctional antibody and its parent monoclonal antibody presented here demonstrate how intravital microscopy can expand our understanding of the in vivo disposition of therapeutics, detecting off-target interactions that could not be detected by conventional pharmacokinetics approaches or predicted by conventional physicochemical analyses.
Collapse
Affiliation(s)
- Amita Datta-Mannan
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Bruce A Molitoris
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Yiqing Feng
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Michelle M Martinez
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Ruben M Sandoval
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Robin M Brown
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Daniel Merkel
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Johnny E Croy
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| | - Kenneth W Dunn
- Exploratory Medicine and Pharmacology (A.D-M.), Clinical Laboratory Services (R.M.B.), and Biotechnology Discovery Research (Y.F., D.M., J.E.C.), Lilly Research Laboratories, Indianapolis, Indiana and Department of Medicine, Division of Nephrology, Indiana University School of Medicine, Indianapolis, Indiana (K.W.D.)
| |
Collapse
|
35
|
Akbarzadeh I, Rezaei N, Bazzazan S, Mezajin MN, Mansouri A, Karbalaeiheidar H, Ashkezari S, Moghaddam ZS, Lalami ZA, Mostafavi E. In silico and in vitro studies of GENT-EDTA encapsulated niosomes: A novel approach to enhance the antibacterial activity and biofilm inhibition in drug-resistant Klebsiella pneumoniae. BIOMATERIALS ADVANCES 2023; 149:213384. [PMID: 37060635 DOI: 10.1016/j.bioadv.2023.213384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 12/06/2022] [Accepted: 03/10/2023] [Indexed: 03/17/2023]
Abstract
Klebsiella pneumoniae (Kp) is a common pathogen inducing catheter-related biofilm infections. Developing effective therapy to overcome antimicrobial resistance (AMR) in Kp is a severe therapeutic challenge that must be solved. This study aimed to prepare niosome-encapsulated GENT (Gentamicin) and EDTA (Ethylenediaminetetraacetic acid) (GENT-EDTA/Nio) to evaluate its efficacy toward Kp strains. The thin-film hydration method was used to prepare various formulations of GENT-EDTA/Nio. Formulations were characterized for their physicochemical characteristics. GENT-EDTA/Nio properties were used for optimization with Design-Expert Software. Molecular docking was utilized to determine the antibacterial activity of GENT. The niosomes displayed a controlled drug release and storage stability of at least 60 days at 4 and 25 °C. GENT-EDTA/Nio performance as antimicrobial agents has been evaluated by employing agar well diffusion method, minimum bactericidal concentration (MBC), and minimum inhibitory concentration (MIC) against the Kp bacteria strains. Biofilm formation was investigated after GENT-EDTA/Nio administration through different detection methods, which showed that this formulation reduces biofilm formation. The effect of GENT-EDTA/Nio on the expression of biofilm-related genes (mrkA, ompA, and vzm) was estimated using QRT-PCR. MTT assay was used to evaluate the toxicity effect of niosomal formulations on HFF cells. The present study results indicate that GENT-EDTA/Nio decreases Kp's resistance to antibiotics and increases its antibiotic and anti-biofilm activity and could be helpful as a new approach for drug delivery.
Collapse
|
36
|
Hakim MH, Jun BH, Ahmadzadegan A, Babiak PM, Xu Q, Buno KP, Liu JC, Ardekani AM, Vlachos PP, Solorio L. Investigation of macromolecular transport through tunable collagen hyaluronic acid matrices. Colloids Surf B Biointerfaces 2023; 222:113123. [PMID: 36640539 DOI: 10.1016/j.colsurfb.2023.113123] [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: 09/12/2022] [Revised: 12/29/2022] [Accepted: 01/01/2023] [Indexed: 01/05/2023]
Abstract
Therapeutic macromolecules possess properties such as size and electrostatic charge that will dictate their transport through subcutaneous (SC) tissue and ultimate bioavailability and efficacy. To improve therapeutic design, platforms that systematically measure the transport of macromolecules as a function of both drug and tissue properties are needed. We utilize a Transwell chamber with tunable collagen-hyaluronic acid (ColHA) hydrogels as an in vitro model to determine mass transport of macromolecules using non-invasive UV spectroscopy. Increasing hyaluronic acid (HA) concentration from 0 to 2 mg/mL within collagen gels decreases the mass transport of five macromolecules independent of size and charge and results in a maximum decrease in recovery of 23.3% in the case of bovine immunoglobulin G (IgG). However, in a pure 10 mg/mL HA solution, negatively-charged macromolecules bovine serum albumin (BSA), β-lactoglobulin (BLg), dextran (Dex), and IgG had drastically increased recovery by 20-40% compared to their performance in ColHA matrices. This result was different from the positively-charged macromolecule Lysozyme (Lys), which, despite its small size, showed reduced recovery by 3% in pure HA. These results demonstrate two distinct regimes of mass transport within our tissue model. In the presence of both collagen and HA, increasing HA concentrations decrease mass transport; however, in the absence of collagen, the high negative charge of HA sequesters and increases residence time of positively-charged macromolecules and decreases residence time of negatively-charged macromolecules. Through our approach, ColHA hydrogels serve as a platform for the systematic evaluation of therapeutic macromolecule transport as a function of molecular characteristics.
Collapse
Affiliation(s)
- Mazin H Hakim
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Brian H Jun
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Adib Ahmadzadegan
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Paulina M Babiak
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, USA
| | - Qinghua Xu
- Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, USA
| | - Kevin P Buno
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Julie C Liu
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Davidson School of Chemical Engineering, Purdue University, West Lafayette, IN, USA
| | - Arezoo M Ardekani
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA
| | - Pavlos P Vlachos
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA.
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA; Purdue Center for Cancer Research, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|
37
|
Estrogen receptor targeting with genistein radiolabeled Technetium-99 m as radiotracer of breast cancer: Its optimization, characterization, and predicting stability constants by DFT calculation. Heliyon 2023; 9:e13169. [PMID: 36747562 PMCID: PMC9898673 DOI: 10.1016/j.heliyon.2023.e13169] [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: 11/20/2022] [Revised: 01/19/2023] [Accepted: 01/20/2023] [Indexed: 01/23/2023] Open
Abstract
Objective Genistein is an isoflavone molecule with a high affinity for estrogen receptors (ER), which could lead to the mechanism of selective estrogen receptor modulators (SERMs) in breast cancer. Genistein labeling with technetium-99m can be a new promising strategy for diagnostic breast cancer. In this research, we evaluate the physicochemical characteristics of the [99mTc]Tc-genistein complex and describe the optimal labeling method parameters. We also calculated density functional theory to study the stability constants to support complex formation analysis (DFT). Methods The genistein was directly labeled with 99mTc, and its stability as well as its potential for usage as a radiotracer were all investigated. DFT calculations with thermodynamic cycles to determine chemical coordination models and calculate thermodynamic constants of complex more accurately. Results The radiochemical purity of [99mTc]Tc-genistein showed a high yield of 93.25% ± 0.30% and had good physicochemical properties. The stability of the Tc(IV)-genistein complex was confirmed by DFT calculations at a value of 99.0822. Conclusions As a result, [99mTc]Tc-genistein could be a potential radiotracer kit for SPECT imaging of breast cancer.
Collapse
|
38
|
Khamis Mahal R, Mohammrd Naser S, Abdulwahid Abdulhussain M, Taha A, Hachim SK, Abdullaha SA, Kadhim MM, Mahdi Rheima A, Zedan Taban T. First-principles studies on two-dimensional aluminum carbide as potential nanocarriers for drug delivery systems. INORG CHEM COMMUN 2022. [DOI: 10.1016/j.inoche.2022.110010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
39
|
Takeuchi T, Kawanishi M, Nakanishi M, Yamasaki H, Tanaka Y. Phase II/III Results of a Trial of Anti-Tumor Necrosis Factor Multivalent NANOBODY Compound Ozoralizumab in Patients With Rheumatoid Arthritis. Arthritis Rheumatol 2022; 74:1776-1785. [PMID: 35729713 PMCID: PMC9828347 DOI: 10.1002/art.42273] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/27/2022] [Accepted: 06/10/2022] [Indexed: 01/12/2023]
Abstract
OBJECTIVE To assess the efficacy and safety of subcutaneous administration of 30 mg or 80 mg of ozoralizumab plus methotrexate (MTX) in patients with rheumatoid arthritis (RA) whose disease remained active despite MTX therapy. METHODS In this multicenter, double-blind, parallel-group, placebo-controlled phase II/III trial, 381 patients were randomized to receive placebo, ozoralizumab 30 mg, or ozoralizumab 80 mg, plus MTX subcutaneously injected every 4 weeks for 24 weeks. The primary end points were the response rates based on the American College of Rheumatology 20% improvement criteria (ACR20) at week 16 and change in the Sharp/van der Heijde score (ΔSHS) from baseline to week 24. RESULTS The proportion of patients with an ACR20 response at week 16 was significantly higher (P < 0.001) in both ozoralizumab groups (79.6% for 30 mg, 75.3% for 80 mg), compared with placebo (37.3%); these improvements were observed from the first week of treatment. The proportion of the patients with structural nonprogression (ΔSHS ≤0) was significantly higher in both ozoralizumab groups than in the placebo group. For some secondary end points, significantly greater improvements were observed starting from as early as day 3. Serious adverse events occurred in 4 patients in the ozoralizumab 30-mg group and 5 patients in the ozoralizumab 80-mg group. CONCLUSION In patients with active RA who received ozoralizumab in combination with MTX, the signs and symptoms of RA were significantly reduced as compared with the outcomes in those receiving placebo. Ozoralizumab demonstrated acceptable tolerability with no new safety signals when compared with other antibodies against tumor necrosis factor.
Collapse
Affiliation(s)
- Tsutomu Takeuchi
- Keio University School of Medicine, Tokyo, and Saitama Medical UniversitySaitamaJapan
| | | | | | | | - Yoshiya Tanaka
- University of Occupational and Environmental Health JapanKitakyushuJapan
| |
Collapse
|
40
|
A novel anti-TNF-α drug ozoralizumab rapidly distributes to inflamed joint tissues in a mouse model of collagen induced arthritis. Sci Rep 2022; 12:18102. [PMID: 36302840 PMCID: PMC9613905 DOI: 10.1038/s41598-022-23152-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/25/2022] [Indexed: 12/30/2022] Open
Abstract
In clinical studies, the next-generation anti-tumor necrosis factor-alpha (TNF-α) single domain antibody ozoralizumab showed high clinical efficacy shortly after the subcutaneous injection. To elucidate the mechanism underlying the rapid onset of the effects of ozoralizumab, we compared the biodistribution kinetics of ozoralizumab and adalimumab after subcutaneous injection in an animal model of arthritis. Alexa Fluor 680-labeled ozoralizumab and adalimumab were administered by subcutaneous injection once (2 mg/kg) at five weeks after induction of collagen-induced arthritis (CIA) in an animal arthritis model. The time-course of changes in the fluorescence intensities of the two compounds in the paws and serum were evaluated. The paws of the CIA mice were harvested at four and eight hours after the injection for fluorescence microscopy. Biofluorescence imaging revealed better distribution of ozoralizumab to the joint tissues than of adalimumab, as early as at four hours after the injection. Fluorescence microscopy revealed a greater fluorescence intensity of ozoralizumab in the joint tissues than that of adalimumab at eight hours after the injection. Ozoralizumab showed a significantly higher absorption rate constant as compared with adalimumab. These results indicate that ozoralizumab enters the systemic circulation more rapidly and is distributed to the target tissues earlier and at higher levels than conventional IgG antibodies. Our investigation provides new insight into the mechanism underlying the rapid onset of the effects of ozoralizumab in clinical practice.
Collapse
|
41
|
Okafor SN, Angsantikul P, Ahmed H. Discovery of Novel HIV Protease Inhibitors Using Modern Computational Techniques. Int J Mol Sci 2022; 23:12149. [PMID: 36293006 PMCID: PMC9603388 DOI: 10.3390/ijms232012149] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/13/2022] [Accepted: 10/01/2022] [Indexed: 09/10/2023] Open
Abstract
The human immunodeficiency virus type 1 (HIV-1) has continued to be a global concern. With the new HIV incidence, the emergence of multi-drug resistance and the untoward side effects of currently used anti-HIV drugs, there is an urgent need to discover more efficient anti-HIV drugs. Modern computational tools have played vital roles in facilitating the drug discovery process. This research focuses on a pharmacophore-based similarity search to screen 111,566,735 unique compounds in the PubChem database to discover novel HIV-1 protease inhibitors (PIs). We used an in silico approach involving a 3D-similarity search, physicochemical and ADMET evaluations, HIV protease-inhibitor prediction (IC50/percent inhibition), rigid receptor-molecular docking studies, binding free energy calculations and molecular dynamics (MD) simulations. The 10 FDA-approved HIV PIs (saquinavir, lopinavir, ritonavir, amprenavir, fosamprenavir, atazanavir, nelfinavir, darunavir, tipranavir and indinavir) were used as reference. The in silico analysis revealed that fourteen out of the twenty-eight selected optimized hit molecules were within the acceptable range of all the parameters investigated. The hit molecules demonstrated significant binding affinity to the HIV protease (PR) when compared to the reference drugs. The important amino acid residues involved in hydrogen bonding and п-п stacked interactions include ASP25, GLY27, ASP29, ASP30 and ILE50. These interactions help to stabilize the optimized hit molecules in the active binding site of the HIV-1 PR (PDB ID: 2Q5K). HPS/002 and HPS/004 have been found to be most promising in terms of IC50/percent inhibition (90.15%) of HIV-1 PR, in addition to their drug metabolism and safety profile. These hit candidates should be investigated further as possible HIV-1 PIs with improved efficacy and low toxicity through in vitro experiments and clinical trial investigations.
Collapse
Affiliation(s)
- Sunday N. Okafor
- Center for Biomedical Research, Population Council, New York, NY 10065, USA
- Department of Pharmaceutical and Medicinal Chemistry, University of Nigeria, Nsukka 41001, Nigeria
| | | | - Hashim Ahmed
- Center for Biomedical Research, Population Council, New York, NY 10065, USA
| |
Collapse
|
42
|
Banik A, Ahmed SR, Sajib EH, Deb A, Sinha S, Azim KF. Identification of potential inhibitory analogs of metastasis tumor antigens (MTAs) using bioactive compounds: revealing therapeutic option to prevent malignancy. Mol Divers 2022; 26:2473-2502. [PMID: 34743299 DOI: 10.1007/s11030-021-10345-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 10/24/2021] [Indexed: 12/31/2022]
Abstract
The deeper understanding of metastasis phenomenon and detection of drug targets could be a potential approach to minimize cancer mortality. In this study, attempts were taken to unmask novel therapeutics to prevent metastasis and cancer progression. Initially, we explored the physiochemical, structural and functional insights of three metastasis tumor antigens (MTAs) and evaluated some plant-based bioactive compounds as potent MTA inhibitors. From 50 plant metabolites screened, isoflavone, gingerol, citronellal and asiatic acid showed maximum binding affinity with all three MTA proteins. The ADME analysis detected no undesirable toxicity that could reduce the drug likeness properties of top plant metabolites. Moreover, molecular dynamics studies revealed that the complexes were stable and showed minimum fluctuation at molecular level. We further performed ligand-based virtual screening to identify similar drug molecules using a large collection of 376,342 compounds from DrugBank. The results suggested that several structural analogs (e.g., tramadol, nabumetone, DGLA and hydrocortisone) may act as agonist to block the MTA proteins and inhibit cancer progression at early stage. The study could be useful to develop effective medications against cancer metastasis in future. Due to encouraging results, we highly recommend further in vitro and in vivo trials for the experimental validation of the findings.
Collapse
Affiliation(s)
- Anik Banik
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
- Department of Plant and Environmental Biotechnology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Sheikh Rashel Ahmed
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
- Department of Plant and Environmental Biotechnology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Emran Hossain Sajib
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Anamika Deb
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Shiuly Sinha
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Kazi Faizul Azim
- Department of Microbial Biotechnology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh.
- Faculté de Pharmacie, Université de Tours, 37200, Tours, France.
| |
Collapse
|
43
|
Esposito S, Orsatti L, Pucci V. Subcutaneous Catabolism of Peptide Therapeutics: Bioanalytical Approaches and ADME Considerations. Xenobiotica 2022; 52:828-839. [PMID: 36039395 DOI: 10.1080/00498254.2022.2119180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Many peptide drugs such as insulin and glucagon-like peptide (GLP-1) analogues are successfully administered subcutaneously (SC). Following SC injection, peptides may undergo catabolism in the SC compartment before entering systemic circulation, which could compromise their bioavailability and in turn affect their efficacy.This review will discuss how both technology and strategy have evolved over the past years to further elucidate peptide SC catabolism.Modern bioanalytical technologies (particularly liquid chromatography-high-resolution mass spectrometry) and bioinformatics platforms for data mining has prompted the development of in silico, in vitro and in vivo tools for characterizing peptide SC catabolism to rapidly address proteolytic liabilities and, ultimately, guide the design of peptides with improved SC bioavailability.More predictive models able to recapitulate the interplay between SC catabolism and other factors driving SC absorption are highly desirable to improve in vitro/in vivo correlations.We envision the routine incorporation of in vitro and in vivo SC catabolism studies in ADME screening funnels to develop more effective peptide drugs for SC delivery.
Collapse
|
44
|
New Advances in Biomedical Application of Polymeric Micelles. Pharmaceutics 2022; 14:pharmaceutics14081700. [PMID: 36015325 PMCID: PMC9416043 DOI: 10.3390/pharmaceutics14081700] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/29/2022] [Accepted: 08/07/2022] [Indexed: 12/20/2022] Open
Abstract
In the last decade, nanomedicine has arisen as an emergent area of medicine, which studies nanometric systems, namely polymeric micelles (PMs), that increase the solubility and the stability of the encapsulated drugs. Furthermore, their application in dermal drug delivery is also relevant. PMs present unique characteristics because of their unique core-shell architecture. They are colloidal dispersions of amphiphilic compounds, which self-assemble in an aqueous medium, giving a structure-type core-shell, with a hydrophobic core (that can encapsulate hydrophobic drugs), and a hydrophilic shell, which works as a stabilizing agent. These features offer PMs adequate steric protection and determine their hydrophilicity, charge, length, and surface density properties. Furthermore, due to their small size, PMs can be absorbed by the intestinal mucosa with the drug, and they transport the drug in the bloodstream until the therapeutic target. Moreover, PMs improve the pharmacokinetic profile of the encapsulated drug, present high load capacity, and are synthesized by a reproducible, easy, and low-cost method. In silico approaches have been explored to improve the physicochemical properties of PMs. Based on this, a computer-aided strategy was developed and validated to enable the delivery of poorly soluble drugs and established critical physicochemical parameters to maximize drug loading, formulation stability, and tumor exposure. Poly(2-oxazoline) (POx)-based PMs display unprecedented high loading concerning water-insoluble drugs and over 60 drugs have been incorporated in POx PMs. Among various stimuli, pH and temperature are the most widely studied for enhanced drug release at the site of action. Researchers are focusing on dual (pH and temperature) responsive PMs for controlled and improved drug release at the site of action. These dual responsive systems are mainly evaluated for cancer therapy as certain malignancies can cause a slight increase in temperature and a decrease in the extracellular pH around the tumor site. This review is a compilation of updated therapeutic applications of PMs, such as PMs that are based on Pluronics®, micelleplexes and Pox-based PMs in several biomedical applications.
Collapse
|
45
|
Ball K, Bruin G, Escandón E, Funk C, Pereira JNS, 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] [MESH Headings] [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 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 members on this theme. A wide variety of in vitro, in vivo and in silico techniques are currently used to assess pharmacokinetics and biodistribution of TPs, and we discuss the relevance of these from an industry perspective, focusing on pharmacokinetic/pharmacodynamic 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 on 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 and Quality Consortium Translational and ADME Sciences Leadership Group working group for the absorption, distribution, metabolism, and excretion of therapeutic proteins evaluates the current practices and challenges in characterizing the pharmacokinetics 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 pharmacokinetic/pharmacodynamic relationships, and aid translational modeling for first-in-human dose predictions.
Collapse
Affiliation(s)
- Kathryn Ball
- Clinical Pharmacology & Quantitative Pharmacology, CPSS, R&D, AstraZeneca, Cambridge, United Kingdom (K.B.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B.); PPDM ADME Biologics, Merck & Co., Inc., South San Francisco, California (E.E.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (C.F.); Discovery & Development Technologies, Drug Disposition & Design, Merck Healthcare KGaA, Darmstadt, Germany (J.N.S.P.); Janssen BioTherapeutics, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Gerard Bruin
- Clinical Pharmacology & Quantitative Pharmacology, CPSS, R&D, AstraZeneca, Cambridge, United Kingdom (K.B.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B.); PPDM ADME Biologics, Merck & Co., Inc., South San Francisco, California (E.E.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (C.F.); Discovery & Development Technologies, Drug Disposition & Design, Merck Healthcare KGaA, Darmstadt, Germany (J.N.S.P.); Janssen BioTherapeutics, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Enrique Escandón
- Clinical Pharmacology & Quantitative Pharmacology, CPSS, R&D, AstraZeneca, Cambridge, United Kingdom (K.B.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B.); PPDM ADME Biologics, Merck & Co., Inc., South San Francisco, California (E.E.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (C.F.); Discovery & Development Technologies, Drug Disposition & Design, Merck Healthcare KGaA, Darmstadt, Germany (J.N.S.P.); Janssen BioTherapeutics, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Christoph Funk
- Clinical Pharmacology & Quantitative Pharmacology, CPSS, R&D, AstraZeneca, Cambridge, United Kingdom (K.B.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B.); PPDM ADME Biologics, Merck & Co., Inc., South San Francisco, California (E.E.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (C.F.); Discovery & Development Technologies, Drug Disposition & Design, Merck Healthcare KGaA, Darmstadt, Germany (J.N.S.P.); Janssen BioTherapeutics, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Joao N S Pereira
- Clinical Pharmacology & Quantitative Pharmacology, CPSS, R&D, AstraZeneca, Cambridge, United Kingdom (K.B.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B.); PPDM ADME Biologics, Merck & Co., Inc., South San Francisco, California (E.E.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (C.F.); Discovery & Development Technologies, Drug Disposition & Design, Merck Healthcare KGaA, Darmstadt, Germany (J.N.S.P.); Janssen BioTherapeutics, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Tong-Yuan Yang
- Clinical Pharmacology & Quantitative Pharmacology, CPSS, R&D, AstraZeneca, Cambridge, United Kingdom (K.B.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B.); PPDM ADME Biologics, Merck & Co., Inc., South San Francisco, California (E.E.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (C.F.); Discovery & Development Technologies, Drug Disposition & Design, Merck Healthcare KGaA, Darmstadt, Germany (J.N.S.P.); Janssen BioTherapeutics, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Hongbin Yu
- Clinical Pharmacology & Quantitative Pharmacology, CPSS, R&D, AstraZeneca, Cambridge, United Kingdom (K.B.); Pharmacokinetic Sciences, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B.); PPDM ADME Biologics, Merck & Co., Inc., South San Francisco, California (E.E.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (C.F.); Discovery & Development Technologies, Drug Disposition & Design, Merck Healthcare KGaA, Darmstadt, Germany (J.N.S.P.); Janssen BioTherapeutics, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| |
Collapse
|
46
|
Fayyaz F, Yar M, Gulzar A, Ayub K. First principles calculations of the adsorption of fluorouracil and nitrosourea on CTF-0; organic frameworks as drug delivery systems for cancer treatment. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.118941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
47
|
Bolleddula J, Brady K, Bruin G, Lee A, Martin JA, Walles M, Xu K, Yang TY, Zhu X, Yu H. Absorption, Distribution, Metabolism, and Excretion of Therapeutic Proteins: Current Industry Practices and Future Perspectives. Drug Metab Dispos 2022; 50:837-845. [PMID: 35149541 DOI: 10.1124/dmd.121.000461] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 01/05/2022] [Indexed: 11/22/2022] Open
Abstract
Therapeutic proteins (TPs) comprise a variety of modalities, including antibody-based drugs, coagulation factors, recombinant cytokines, enzymes, growth factors, and hormones. TPs usually cannot traverse cellular barriers and exert their pharmacological activity by interacting with targets on the exterior membrane of cells or with soluble ligands in the tissue interstitial fluid/blood. Due to their large size, lack of cellular permeability, variation in metabolic fate, and distinct physicochemical characteristics, TPs are subject to different absorption, distribution, metabolism, and excretion (ADME) processes as compared with small molecules. Limited regulatory guidance makes it challenging to determine the most relevant ADME data required for regulatory submissions. The TP ADME working group was sponsored by the Translational and ADME Sciences Leadership Group within the Innovation and Quality (IQ) consortium with objectives to: (1) better understand the current practices of ADME data generated for TPs across IQ member companies, (2) learn about their regulatory strategies and interaction experiences, and (3) provide recommendations on best practices for conducting ADME studies for TPs. To understand current ADME practices and regulatory strategies, an industry-wide survey was conducted within IQ member companies. In addition, ADME data submitted to the U.S. Food and Drug Administration was also collated by reviewing regulatory submission packages of TPs approved between 2011 and 2020. This article summarizes the key learnings from the survey and an overview of ADME data presented in biologics license applications along with future perspectives and recommendations for conducting ADME studies for internal decision-making as well as regulatory submissions for TPs. SIGNIFICANCE STATEMENT: This article provides comprehensive assessment of the current practices of absorption, distribution, metabolism, and excretion (ADME) data generated for therapeutic proteins (TPs) across the Innovation and Quality participating companies and the utility of the data in discovery, development, and regulatory submissions. The TP ADME working group also recommends the best practices for condu-cting ADME studies for internal decision-making and regulatory submissions.
Collapse
Affiliation(s)
- Jayaprakasam Bolleddula
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (J.B.); Quantitative Pharmacology & DMPK, UCB Pharma S.A., Slough, United Kingdom (K.B.); Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B., M.W.); Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington (A.L.); Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.A.M); BioAnalytical Sciences, Genentech, South San Francisco, California (K.X.); Preclinical Sciences and Translational Safety, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Kevin Brady
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (J.B.); Quantitative Pharmacology & DMPK, UCB Pharma S.A., Slough, United Kingdom (K.B.); Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B., M.W.); Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington (A.L.); Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.A.M); BioAnalytical Sciences, Genentech, South San Francisco, California (K.X.); Preclinical Sciences and Translational Safety, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Gerard Bruin
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (J.B.); Quantitative Pharmacology & DMPK, UCB Pharma S.A., Slough, United Kingdom (K.B.); Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B., M.W.); Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington (A.L.); Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.A.M); BioAnalytical Sciences, Genentech, South San Francisco, California (K.X.); Preclinical Sciences and Translational Safety, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Anthony Lee
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (J.B.); Quantitative Pharmacology & DMPK, UCB Pharma S.A., Slough, United Kingdom (K.B.); Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B., M.W.); Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington (A.L.); Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.A.M); BioAnalytical Sciences, Genentech, South San Francisco, California (K.X.); Preclinical Sciences and Translational Safety, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Jennifer A Martin
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (J.B.); Quantitative Pharmacology & DMPK, UCB Pharma S.A., Slough, United Kingdom (K.B.); Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B., M.W.); Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington (A.L.); Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.A.M); BioAnalytical Sciences, Genentech, South San Francisco, California (K.X.); Preclinical Sciences and Translational Safety, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Markus Walles
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (J.B.); Quantitative Pharmacology & DMPK, UCB Pharma S.A., Slough, United Kingdom (K.B.); Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B., M.W.); Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington (A.L.); Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.A.M); BioAnalytical Sciences, Genentech, South San Francisco, California (K.X.); Preclinical Sciences and Translational Safety, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Keyang Xu
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (J.B.); Quantitative Pharmacology & DMPK, UCB Pharma S.A., Slough, United Kingdom (K.B.); Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B., M.W.); Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington (A.L.); Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.A.M); BioAnalytical Sciences, Genentech, South San Francisco, California (K.X.); Preclinical Sciences and Translational Safety, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Tong-Yuan Yang
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (J.B.); Quantitative Pharmacology & DMPK, UCB Pharma S.A., Slough, United Kingdom (K.B.); Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B., M.W.); Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington (A.L.); Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.A.M); BioAnalytical Sciences, Genentech, South San Francisco, California (K.X.); Preclinical Sciences and Translational Safety, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Xiaochun Zhu
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (J.B.); Quantitative Pharmacology & DMPK, UCB Pharma S.A., Slough, United Kingdom (K.B.); Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B., M.W.); Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington (A.L.); Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.A.M); BioAnalytical Sciences, Genentech, South San Francisco, California (K.X.); Preclinical Sciences and Translational Safety, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| | - Hongbin Yu
- Quantitative Pharmacology, EMD Serono Research & Development Institute, Inc., Billerica, Massachusetts (J.B.); Quantitative Pharmacology & DMPK, UCB Pharma S.A., Slough, United Kingdom (K.B.); Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (G.B., M.W.); Quantitative Pharmacology and Disposition, Seagen Inc., Bothell, Washington (A.L.); Drug Disposition, Eli Lilly and Company, Indianapolis, Indiana (J.A.M); BioAnalytical Sciences, Genentech, South San Francisco, California (K.X.); Preclinical Sciences and Translational Safety, Janssen R&D, LLC, Spring House, Pennsylvania (T.-Y.Y.); Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.); and Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut (H.Y.)
| |
Collapse
|
48
|
Lai Y, Chu X, Di L, Gao W, Guo Y, Liu X, Lu C, Mao J, Shen H, Tang H, Xia CQ, Zhang L, Ding X. Recent advances in the translation of drug metabolism and pharmacokinetics science for drug discovery and development. Acta Pharm Sin B 2022; 12:2751-2777. [PMID: 35755285 PMCID: PMC9214059 DOI: 10.1016/j.apsb.2022.03.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023] Open
Abstract
Drug metabolism and pharmacokinetics (DMPK) is an important branch of pharmaceutical sciences. The nature of ADME (absorption, distribution, metabolism, excretion) and PK (pharmacokinetics) inquiries during drug discovery and development has evolved in recent years from being largely descriptive to seeking a more quantitative and mechanistic understanding of the fate of drug candidates in biological systems. Tremendous progress has been made in the past decade, not only in the characterization of physiochemical properties of drugs that influence their ADME, target organ exposure, and toxicity, but also in the identification of design principles that can minimize drug-drug interaction (DDI) potentials and reduce the attritions. The importance of membrane transporters in drug disposition, efficacy, and safety, as well as the interplay with metabolic processes, has been increasingly recognized. Dramatic increases in investments on new modalities beyond traditional small and large molecule drugs, such as peptides, oligonucleotides, and antibody-drug conjugates, necessitated further innovations in bioanalytical and experimental tools for the characterization of their ADME properties. In this review, we highlight some of the most notable advances in the last decade, and provide future perspectives on potential major breakthroughs and innovations in the translation of DMPK science in various stages of drug discovery and development.
Collapse
Affiliation(s)
- Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA 94404, USA
| | - Xiaoyan Chu
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Li Di
- Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Groton, CT 06340, USA
| | - Wei Gao
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Inc., Kenilworth, NJ 07033, USA
| | - Yingying Guo
- Eli Lilly and Company, Indianapolis, IN 46221, USA
| | - Xingrong Liu
- Drug Metabolism and Pharmacokinetics, Biogen, Cambridge, MA 02142, USA
| | - Chuang Lu
- Drug Metabolism and Pharmacokinetics, Accent Therapeutics, Inc. Lexington, MA 02421, USA
| | - Jialin Mao
- Department of Drug Metabolism and Pharmacokinetics, Genentech, A Member of the Roche Group, South San Francisco, CA 94080, USA
| | - Hong Shen
- Drug Metabolism and Pharmacokinetics Department, Bristol-Myers Squibb Company, Princeton, NJ 08540, USA
| | - Huaping Tang
- Bioanalysis and Biomarkers, Glaxo Smith Kline, King of the Prussia, PA 19406, USA
| | - Cindy Q. Xia
- Department of Drug Metabolism and Pharmacokinetics, Takeda Pharmaceuticals International Co., Cambridge, MA 02139, USA
| | - Lei Zhang
- Office of Research and Standards, Office of Generic Drugs, CDER, FDA, Silver Spring, MD 20993, USA
| | - Xinxin Ding
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
49
|
Walles M, Berna M, Jian W, Hauri S, Hengel S, King L, Tran JC, Wei C, Xu K, Zhu X. A Cross Company Perspective on the Assessment of Therapeutic Protein Biotransformation. Drug Metab Dispos 2022; 50:846-857. [PMID: 35306476 DOI: 10.1124/dmd.121.000462] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 03/02/2022] [Indexed: 02/13/2025] Open
Abstract
Unlike with new chemical entities, the biotransformation of therapeutic proteins (TPs) has not been routinely investigated or included in regulatory filings. Nevertheless, there is an expanding pool of evidence suggesting that a more in-depth understanding of biotransformation could better aid the discovery and development of increasingly diverse modalities. For instance, such biotransformation analysis of TPs affords important information on molecular stability, which in turn may shed light on any potential impact on binding affinity, potency, pharmacokinetics, efficacy, safety, or bioanalysis. This perspective summarizes the current practices in studying biotransformation of TPs and related findings in the biopharmaceutical industry. Various TP case studies are discussed, and a fit-for-purpose approach is recommended when investigating their biotransformation. In addition, we provide a decision tree to guide the biotransformation characterization for selected modalities. By raising the awareness of this important topic, which remains relatively underexplored in the development of TPs (Bolleddula et al., 2022), we hope that current and developing practices can pave the way for establishing a consensus on the biotransformation assessment of TPs. SIGNIFICANCE STATEMENT: This article provides a comprehensive perspective of the current practices for exploring the biotransformation of therapeutic proteins across the drug development industry. We, the participants of the Innovation and Quality therapeutic protein absorption distribution metabolism excretion working group, recommend and summarize appropriate approaches for conducting biotransformation studies to support internal decision making based on the data generated in discovery and development.
Collapse
Affiliation(s)
- Markus Walles
- Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.W.); Biotechnology Discovery Research-ADME, Eli Lilly and Company, Indianapolis, Indiana (M.B.); Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, Pennsylvania (W.J.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (S.Ha.); Quantitative Pharmacology and Disposition, Seagen, Inc., Bothell, Washington (S.He.); Drug Metabolism and Pharmacokinetics, UCB Biopharma, Slough, UK (L.K.); Bioanalytical Sciences (K.X.) and Biochemical and Cellular Pharmacology (J.C.T.), Genentech, South San Francisco, California; Drug Metabolism and Pharmacokinetics, Biogen, Inc., Cambridge, Massachusetts (C.W.); and Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.)
| | - Michael Berna
- Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.W.); Biotechnology Discovery Research-ADME, Eli Lilly and Company, Indianapolis, Indiana (M.B.); Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, Pennsylvania (W.J.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (S.Ha.); Quantitative Pharmacology and Disposition, Seagen, Inc., Bothell, Washington (S.He.); Drug Metabolism and Pharmacokinetics, UCB Biopharma, Slough, UK (L.K.); Bioanalytical Sciences (K.X.) and Biochemical and Cellular Pharmacology (J.C.T.), Genentech, South San Francisco, California; Drug Metabolism and Pharmacokinetics, Biogen, Inc., Cambridge, Massachusetts (C.W.); and Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.)
| | - Wenying Jian
- Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.W.); Biotechnology Discovery Research-ADME, Eli Lilly and Company, Indianapolis, Indiana (M.B.); Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, Pennsylvania (W.J.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (S.Ha.); Quantitative Pharmacology and Disposition, Seagen, Inc., Bothell, Washington (S.He.); Drug Metabolism and Pharmacokinetics, UCB Biopharma, Slough, UK (L.K.); Bioanalytical Sciences (K.X.) and Biochemical and Cellular Pharmacology (J.C.T.), Genentech, South San Francisco, California; Drug Metabolism and Pharmacokinetics, Biogen, Inc., Cambridge, Massachusetts (C.W.); and Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.)
| | - Simon Hauri
- Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.W.); Biotechnology Discovery Research-ADME, Eli Lilly and Company, Indianapolis, Indiana (M.B.); Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, Pennsylvania (W.J.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (S.Ha.); Quantitative Pharmacology and Disposition, Seagen, Inc., Bothell, Washington (S.He.); Drug Metabolism and Pharmacokinetics, UCB Biopharma, Slough, UK (L.K.); Bioanalytical Sciences (K.X.) and Biochemical and Cellular Pharmacology (J.C.T.), Genentech, South San Francisco, California; Drug Metabolism and Pharmacokinetics, Biogen, Inc., Cambridge, Massachusetts (C.W.); and Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.)
| | - Shawna Hengel
- Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.W.); Biotechnology Discovery Research-ADME, Eli Lilly and Company, Indianapolis, Indiana (M.B.); Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, Pennsylvania (W.J.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (S.Ha.); Quantitative Pharmacology and Disposition, Seagen, Inc., Bothell, Washington (S.He.); Drug Metabolism and Pharmacokinetics, UCB Biopharma, Slough, UK (L.K.); Bioanalytical Sciences (K.X.) and Biochemical and Cellular Pharmacology (J.C.T.), Genentech, South San Francisco, California; Drug Metabolism and Pharmacokinetics, Biogen, Inc., Cambridge, Massachusetts (C.W.); and Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.)
| | - Lloyd King
- Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.W.); Biotechnology Discovery Research-ADME, Eli Lilly and Company, Indianapolis, Indiana (M.B.); Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, Pennsylvania (W.J.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (S.Ha.); Quantitative Pharmacology and Disposition, Seagen, Inc., Bothell, Washington (S.He.); Drug Metabolism and Pharmacokinetics, UCB Biopharma, Slough, UK (L.K.); Bioanalytical Sciences (K.X.) and Biochemical and Cellular Pharmacology (J.C.T.), Genentech, South San Francisco, California; Drug Metabolism and Pharmacokinetics, Biogen, Inc., Cambridge, Massachusetts (C.W.); and Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.)
| | - John C Tran
- Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.W.); Biotechnology Discovery Research-ADME, Eli Lilly and Company, Indianapolis, Indiana (M.B.); Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, Pennsylvania (W.J.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (S.Ha.); Quantitative Pharmacology and Disposition, Seagen, Inc., Bothell, Washington (S.He.); Drug Metabolism and Pharmacokinetics, UCB Biopharma, Slough, UK (L.K.); Bioanalytical Sciences (K.X.) and Biochemical and Cellular Pharmacology (J.C.T.), Genentech, South San Francisco, California; Drug Metabolism and Pharmacokinetics, Biogen, Inc., Cambridge, Massachusetts (C.W.); and Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.)
| | - Cong Wei
- Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.W.); Biotechnology Discovery Research-ADME, Eli Lilly and Company, Indianapolis, Indiana (M.B.); Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, Pennsylvania (W.J.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (S.Ha.); Quantitative Pharmacology and Disposition, Seagen, Inc., Bothell, Washington (S.He.); Drug Metabolism and Pharmacokinetics, UCB Biopharma, Slough, UK (L.K.); Bioanalytical Sciences (K.X.) and Biochemical and Cellular Pharmacology (J.C.T.), Genentech, South San Francisco, California; Drug Metabolism and Pharmacokinetics, Biogen, Inc., Cambridge, Massachusetts (C.W.); and Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.)
| | - Keyang Xu
- Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.W.); Biotechnology Discovery Research-ADME, Eli Lilly and Company, Indianapolis, Indiana (M.B.); Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, Pennsylvania (W.J.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (S.Ha.); Quantitative Pharmacology and Disposition, Seagen, Inc., Bothell, Washington (S.He.); Drug Metabolism and Pharmacokinetics, UCB Biopharma, Slough, UK (L.K.); Bioanalytical Sciences (K.X.) and Biochemical and Cellular Pharmacology (J.C.T.), Genentech, South San Francisco, California; Drug Metabolism and Pharmacokinetics, Biogen, Inc., Cambridge, Massachusetts (C.W.); and Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.)
| | - Xiaochun Zhu
- Pharmacokinetic Science, Novartis Institutes for Biomedical Research, Basel, Switzerland (M.W.); Biotechnology Discovery Research-ADME, Eli Lilly and Company, Indianapolis, Indiana (M.B.); Drug Metabolism and Pharmacokinetics, Janssen Research & Development, Spring House, Pennsylvania (W.J.); Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland (S.Ha.); Quantitative Pharmacology and Disposition, Seagen, Inc., Bothell, Washington (S.He.); Drug Metabolism and Pharmacokinetics, UCB Biopharma, Slough, UK (L.K.); Bioanalytical Sciences (K.X.) and Biochemical and Cellular Pharmacology (J.C.T.), Genentech, South San Francisco, California; Drug Metabolism and Pharmacokinetics, Biogen, Inc., Cambridge, Massachusetts (C.W.); and Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, Cambridge, Massachusetts (X.Z.)
| |
Collapse
|
50
|
Dammen-Brower K, Epler P, Zhu S, Bernstein ZJ, Stabach PR, Braddock DT, Spangler JB, Yarema KJ. Strategies for Glycoengineering Therapeutic Proteins. Front Chem 2022; 10:863118. [PMID: 35494652 PMCID: PMC9043614 DOI: 10.3389/fchem.2022.863118] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 03/25/2022] [Indexed: 12/14/2022] Open
Abstract
Almost all therapeutic proteins are glycosylated, with the carbohydrate component playing a long-established, substantial role in the safety and pharmacokinetic properties of this dominant category of drugs. In the past few years and moving forward, glycosylation is increasingly being implicated in the pharmacodynamics and therapeutic efficacy of therapeutic proteins. This article provides illustrative examples of drugs that have already been improved through glycoengineering including cytokines exemplified by erythropoietin (EPO), enzymes (ectonucleotide pyrophosphatase 1, ENPP1), and IgG antibodies (e.g., afucosylated Gazyva®, Poteligeo®, Fasenra™, and Uplizna®). In the future, the deliberate modification of therapeutic protein glycosylation will become more prevalent as glycoengineering strategies, including sophisticated computer-aided tools for "building in" glycans sites, acceptance of a broad range of production systems with various glycosylation capabilities, and supplementation methods for introducing non-natural metabolites into glycosylation pathways further develop and become more accessible.
Collapse
Affiliation(s)
- Kris Dammen-Brower
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paige Epler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Stanley Zhu
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Zachary J. Bernstein
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| | - Paul R. Stabach
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Demetrios T. Braddock
- Department of Pathology, Yale University School of Medicine, New Haven, CT, United States
| | - Jamie B. Spangler
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Kevin J. Yarema
- Translational Tissue Engineering Center, Johns Hopkins School of Medicine, Baltimore, MD, United States
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|