1
|
Yang L, Haeckel A, Beindorff N, Poetzsch SML, Mi H, Ni F, Hojjat H, Brenner W, de Moraes PAD, Guo J, Savic LJ, Schellenberger E. Long circulating XTEN864-HGV-Apoptin fusion protein for selective cancer therapy. Int J Biol Macromol 2025:141679. [PMID: 40037452 DOI: 10.1016/j.ijbiomac.2025.141679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 02/26/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
The virus protein CAV-Apoptin and its homologue HGV-Apoptin selectively kill cancer cells but are not suitable for systemic treatment. The aim was to develop Apoptin-based fusion proteins for intravenous application in cancer therapy, which also contain the hydrophilic polypeptide XTEN, a cleavage site for MMP-2/9, and a TAT peptide for cell penetration. Expression of XTEN864-HGV-Apoptin in E. coli and purification using XTEN as a tag yielded 100 mg protein/L tissue culture. The expression of XTEN864-CAV-Apoptin did not generate a sufficient yield. Cytotoxic effects were assessed using MTT and Annexin A5 assays, whereas cellular uptake was visualized using Cy3.5-XTEN864-HGV-Apoptin. Blood half-life and biodistribution were evaluated with 99mTc-XTEN864-HGV-Apoptin using SPECT-CT and gamma counting. The fusion protein significantly reduced cancer cell growth and induced apoptosis with minimal effects on non-cancerous cells. It accumulates in the nucleus and associates with F-actin. In mice, the protein showed a blood half-life of 0.68 (fast phase) and 17 h (slow phase), with a tumor/muscle ratio of 9.36 ± 6.22 (SD). In a 4 T1 mouse tumor model, it effectively inhibited tumor growth. The cancer-specific cytotoxicity and prolonged circulation of XTEN864-HGV-Apoptin suggest its potential for systemically applicable, biodegradable, and E. coli-producible antitumor drugs.
Collapse
Affiliation(s)
- Liu Yang
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Akvile Haeckel
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Nicola Beindorff
- Charité - Universitätsmedizin Berlin, Berlin Experimental Radionuclide Imaging Center (BERIC), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Simon Marc Levin Poetzsch
- Charité - Universitätsmedizin Berlin, Berlin Experimental Radionuclide Imaging Center (BERIC), Augustenburger Platz 1, 13353 Berlin, Germany
| | - Honglan Mi
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Fei Ni
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Hamidreza Hojjat
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Winfried Brenner
- Charité - Universitätsmedizin Berlin, Berlin Experimental Radionuclide Imaging Center (BERIC), Augustenburger Platz 1, 13353 Berlin, Germany; Charité - Universitätsmedizin Berlin, Department of Nuclear Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Pedro Augusto Dantas de Moraes
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Jing Guo
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany
| | - Lynn Jeanette Savic
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eyk Schellenberger
- Charité - Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Charitéplatz 1, 10117 Berlin, Germany.
| |
Collapse
|
2
|
Gong B, Wang T, Sun L. Evolution and therapeutic potential of glucagon-like peptide 2 analogs. Biochem Pharmacol 2025; 233:116758. [PMID: 39842552 DOI: 10.1016/j.bcp.2025.116758] [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: 08/22/2024] [Revised: 12/18/2024] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Glucagon-like peptide 2 (GLP-2) is a proglucagon-derived peptide released by intestinal endocrine cells. However, its therapeutic potential is limited by rapid inactivation via dipeptidyl peptidase-IV. The elucidation of three-dimensional structures of G-protein-coupled receptors, including GLP-2 receptor, has facilitated the rational design of novel peptide therapeutics. Recent studies have explored various structural modifications based on the structure of GLP-2, such as amino acid substitution, lipidation, and fusion with proteins, to extend the half-life of GLP-2 and enhance its biological activity. One promising avenue involves the development of multifunctional molecules targeting multiple pharmacological systems to boost therapeutic efficacy. This paper reviews the recent advancements in understanding GLP-2, including its physiological roles and structure-activity relationships, and evaluates the development prospects of GLP-2 analogs.
Collapse
Affiliation(s)
- Binbin Gong
- College of Medicine, Jiaxing University, Jiaxing 314001, PR China; College of Pharmacy, Zhejiang University of Technology, Hangzhou 310000, PR China
| | - Ting Wang
- College of Medicine, Jiaxing University, Jiaxing 314001, PR China
| | - Lidan Sun
- College of Medicine, Jiaxing University, Jiaxing 314001, PR China; Taizhou Hospital, Zhejiang University, Taizhou 317000, PR China.
| |
Collapse
|
3
|
Zhou Y, Li J, Gao G, Li Y, Zhang C. Exploring a novel long-acting glucagon-like peptide-1 receptor agonist built on the albumin-binding domain and XTEN scaffolds. Heliyon 2024; 10:e24340. [PMID: 38293540 PMCID: PMC10826136 DOI: 10.1016/j.heliyon.2024.e24340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 02/01/2024] Open
Abstract
In recent years, glucagon-like peptide-1 (GLP-1) has demonstrated considerable potential in the treatment of type 2 diabetes (T2D) and obesity. However, the half-life of naturally occurring GLP-1 is quite short in vivo. Two common strategies employed for half-life extension are the use of the Albumin-binding domain (ABD) and XTEN polypeptide, which operate through different mechanisms. In this study, we designed an innovative GLP-1 receptor agonist with an extended duration of action. This new construct incorporated an albumin binding domain (ABD) and an XTEN sequence (either XTEN144 or XTEN288) as carriers. We referred to these fusion proteins as GLP-ABD-XTEN144 and GLP-ABD-XTEN288. In an E. coli system, the said constructs were efficaciously produced in substantial quantity. It was observed from in vitro studies that the fusion protein GLP-ABD-XTEN144 demonstrated a five times stronger affinity towards human serum albumin (HSA), boasting a binding affinity (Kd) of 5.50 nM. This was in contrast to GLP-ABD-XTEN288, whose Kd value was registered at 27.78 nM. Moreover, GLP-ABD-XTEN144 presented a half-life of 12.9 h in mice, thus exceeding the corresponding value for GLP-ABD-XTEN288, 7.32 h in mice. Both these fusion proteins significantly mitigated non-fasting blood sugar levels and overall food consumption for 48 h subsequent to a one-time injection in mice. Notably, GLP-ABD-XTEN144 exhibited more pronounced hypoglycemic activity and food inhibitory effects than GLP-ABD-XTEN288. The designed GLP-ABD-XTEN144 fusion protein shows promising prospects for clinical application in T2D treatment. Our findings also suggest that ABD and XTEN polypeptides synergistically contribute to half-life extension, further enhancing the pharmacokinetic characteristics of a payload.
Collapse
Affiliation(s)
- Yan Zhou
- Department of Pharmacy, Ningbo No. 2 Hospital, Ningbo, China
| | - Jianhui Li
- Department of Endocrinology, Ningbo No. 2 Hospital, Ningbo, China
| | - Guosheng Gao
- Department of Clinical Laboratory, Ningbo No. 2 Hospital, Ningbo, China
| | - Yafeng Li
- Department of Pharmacology, Duchuangsanzhong Biotech Co., Ltd., Jiaxing, China
| | - Changzhen Zhang
- Department of Pharmacy, Ningbo No. 2 Hospital, Ningbo, China
| |
Collapse
|
4
|
Chen H, Zhang Q. Polypeptides as alternatives to PEGylation of therapeutic agents. Expert Opin Drug Deliv 2024; 21:1-12. [PMID: 38116624 DOI: 10.1080/17425247.2023.2297937] [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: 10/05/2023] [Accepted: 12/18/2023] [Indexed: 12/21/2023]
Abstract
INTRODUCTION Due to the concerns raised by the extensive application of PEGylation, polypeptides have stood out as excellent candidates with adequate biocompatibility and biodegradability with tunable hydrophilicity. AREAS COVERED In this review, polypeptides with the potential to replace PEGylation have been summarized and their application has been reviewed, including XTEN, PASylation, polysarcosine, zwitterion polypeptides, ELPylation, etc. Besides their strengths, the remaining challenges have also been discussed and the future perspectives have been provided. EXPERT OPINION Polypeptides have been applied in the designing of peptide/protein drugs as well as nanomedicines, and some of the pharmaceutics have made it into the clinical trials and got approved. These polypeptides showed similar hydrophilic properties to PEGylation, which increased the hydrodynamic volumes of protein drugs, reduced kidney elimination, decreased protein-polymer interaction and potentially improved the drug delivery efficiency due to the extended circulation time in the system. Moreover, they demonstrated superior biodegradability and biocompatibility, compensating for the deficiencies for polymers such as PEG.
Collapse
Affiliation(s)
- Huali Chen
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| | - Qianyu Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, China
| |
Collapse
|
5
|
Ji Y, Liu D, Zhu H, Bao L, Chang R, Gao X, Yin J. Unstructured Polypeptides as a Versatile Drug Delivery Technology. Acta Biomater 2023; 164:74-93. [PMID: 37075961 DOI: 10.1016/j.actbio.2023.04.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/23/2023] [Accepted: 04/13/2023] [Indexed: 04/21/2023]
Abstract
Although polyethylene glycol (PEG), or "PEGylation" has become a widely applied approach for improving the efficiency of drug delivery, the immunogenicity and non-biodegradability of this synthetic polymer have prompted an evident need for alternatives. To overcome these caveats and to mimic PEG -or other natural or synthetic polymers- for the purpose of drug half-life extension, unstructured polypeptides are designed. Due to their tunable length, biodegradability, low immunogenicity and easy production, unstructured polypeptides have the potential to replace PEG as the preferred technology for therapeutic protein/peptide delivery. This review provides an overview of the evolution of unstructured polypeptides, starting from natural polypeptides to engineered polypeptides and discusses their characteristics. Then, it is described that unstructured polypeptides have been successfully applied to numerous drugs, including peptides, proteins, antibody fragments, and nanocarriers, for half-life extension. Innovative applications of unstructured peptides as releasable masks, multimolecular adaptors and intracellular delivery carriers are also discussed. Finally, challenges and future perspectives of this promising field are briefly presented. STATEMENT OF SIGNIFICANCE: : Polypeptide fusion technology simulating PEGylation has become an important topic for the development of long-circulating peptide or protein drugs without reduced activity, complex processes, and kidney injury caused by PEG modification. Here we provide a detailed and in-depth review of the recent advances in unstructured polypeptides. In addition to the application of enhanced pharmacokinetic performance, emphasis is placed on polypeptides as scaffolders for the delivery of multiple drugs, and on the preparation of reasonably designed polypeptides to manipulate the performance of proteins and peptides. This review will provide insight into future application of polypeptides in peptide or protein drug development and the design of novel functional polypeptides.
Collapse
Affiliation(s)
- Yue Ji
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Dingkang Liu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Haichao Zhu
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Lichen Bao
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing 210009, China
| | - Ruilong Chang
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangdong Gao
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| | - Jun Yin
- Jiangsu Key Laboratory of Druggability of Biopharmaceuticals and State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
6
|
Cattaruzza F, Nazeer A, To M, Hammond M, Koski C, Liu LY, Pete Yeung V, Rennerfeldt DA, Henkensiefken A, Fox M, Lam S, Morrissey KM, Lange Z, Podust VN, Derynck MK, Irving BA, Schellenberger V. Precision-activated T-cell engagers targeting HER2 or EGFR and CD3 mitigate on-target, off-tumor toxicity for immunotherapy in solid tumors. NATURE CANCER 2023; 4:485-501. [PMID: 36997747 PMCID: PMC10132983 DOI: 10.1038/s43018-023-00536-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 03/02/2023] [Indexed: 04/01/2023]
Abstract
To enhance the therapeutic index of T-cell engagers (TCEs), we engineered masked, precision-activated TCEs (XPAT proteins), targeting a tumor antigen (human epidermal growth factor receptor 2 (HER2) or epidermal growth factor receptor (EGFR)) and CD3. Unstructured XTEN polypeptide masks flank the N and C termini of the TCE and are designed to be released by proteases in the tumor microenvironment. In vitro, unmasked HER2-XPAT (uTCE) demonstrates potent cytotoxicity, with XTEN polypeptide masking providing up to 4-log-fold protection. In vivo, HER2-XPAT protein induces protease-dependent antitumor activity and is proteolytically stable in healthy tissues. In non-human primates, HER2-XPAT protein demonstrates a strong safety margin (>400-fold increase in tolerated maximum concentration versus uTCE). HER2-XPAT protein cleavage is low and similar in plasma samples from healthy and diseased humans and non-human primates, supporting translatability of stability to patients. EGFR-XPAT protein confirmed the utility of XPAT technology for tumor targets more widely expressed in healthy tissues.
Collapse
Affiliation(s)
- Fiore Cattaruzza
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Ayesha Nazeer
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Milton To
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Mikhail Hammond
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Caitlin Koski
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Lucas Y Liu
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - V Pete Yeung
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | | | | | - Michael Fox
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Sharon Lam
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Kari M Morrissey
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Zachary Lange
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Vladimir N Podust
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Mika K Derynck
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | - Bryan A Irving
- Amunix Pharmaceuticals, a Sanofi Company, South San Francisco, CA, USA
| | | |
Collapse
|
7
|
Xia J, Gao G, Zhang C, Ying J, Li J. Albumin-binding DARPins as scaffold improve the hypoglycemic and anti-obesity effects of exendin-4 in vivo. Eur J Pharm Sci 2023; 185:106422. [PMID: 36906110 DOI: 10.1016/j.ejps.2023.106422] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/17/2023] [Accepted: 03/07/2023] [Indexed: 03/11/2023]
Abstract
Type 2 diabetes mellitus (T2DM) and obesity have been considered epidemics and threats to public health worldwide. Exendin-4 (Ex), a GLP-1R agonist, has potential for treating T2DM and obesity. However, Ex has a half-life of only 2.4 h in humans and needs to be administered twice daily, which hampers its clinical application. In this study, we synthesized four new GLP-1R agonists by genetically fusing Ex to the N-terminus of HSA-binding ankyrin repeat proteins (DARPins) via linkers of different lengths, denoted as Ex-DARPin-GSx fusion proteins (x = 0, 1, 2, and 3). The Ex-DARPin fusion proteins were substantially stable, resulting in incomplete denaturation even at 80 °C. The in vitro bioactivity results demonstrated that Ex-DARPin fusion proteins could bind to HSA and activate GLP-1R. The Ex-DARPin fusion proteins had a comparable half-life (29-32 h), which is much longer than that of native Ex (0.5 h in rats). Subcutaneous injection of 25 nmol/kg Ex-DARPin fusion protein normalized blood glucose (BG) levels for at least 72 h in mice. The Ex-DARPin fusion proteins, injected at 25 nmol/kg every three days, significantly lowered BG, inhibited food consumption, and reduced body weight (BW) for 30 days in STZ-induced diabetic mice. Histological analysis of pancreatic tissues using H&E staining revealed that Ex-DARPin fusion proteins significantly improved the survival of pancreatic islets in diabetic mice. The differences in in vivo bioactivity of fusion proteins with different linker lengths were not significant. According to the findings in this study, long-acting Ex-DARPin fusion proteins designed by us hold promise for further development as antidiabetic and antiobesity therapeutic agents. Our findings also indicate that DARPins are a universal platform for generating long-acting therapeutic proteins via genetic fusion, thus broadening the application scope of DARPins.
Collapse
Affiliation(s)
- Jinying Xia
- Department of Endocrinology, Ningbo No. 2 Hospital, Ningbo, China; Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China
| | - Guosheng Gao
- Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China; Department of Clinical Laboratory, Ningbo No. 2 Hospital, Ningbo, China
| | - Changzhen Zhang
- Department of Pharmacy, Ningbo No. 2 Hospital, Ningbo, China
| | - Jingjing Ying
- Department of Pharmacy, Ningbo No. 2 Hospital, Ningbo, China
| | - Jianhui Li
- Department of Endocrinology, Ningbo No. 2 Hospital, Ningbo, China; Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, China.
| |
Collapse
|
8
|
Mahmood T, Shahbaz A, Hussain N, Ali R, Bashir H, Rizwan K. Recent advancements in fusion protein technologies in oncotherapy: A review. Int J Biol Macromol 2023; 230:123161. [PMID: 36610574 DOI: 10.1016/j.ijbiomac.2023.123161] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/01/2023] [Accepted: 01/02/2023] [Indexed: 01/05/2023]
Abstract
Cancer is a complicated, adaptable, and heterogeneous disease caused by a wide variety of genetic changes that might impair ability of cells to function normally. The majority of the tumors can only be shrunk using conventional oncology therapies like chemotherapy, radiation, and surgical resection, and the tumor often recurs. The inability of conventional cancer therapies to completely destroy the Cancer Stem Cells (CSCs) that otherwise lead to therapy resistance is thus addressed by therapeutic approaches that concentrate on targeting CSCs and their micro-environmental niche. In this review, we summarize approaches that are used for the development of fusion proteins and their therapeutic applications for treating cancer. The main purpose of making advancements towards the fusion technology instead of using conventional treatment methods is to achieve a prolonged half-life of the therapeutic drugs. The fusion of drugs to the immune response enhancing cytokines or the fusion of antibody and cytokines not only increases half-life but also increase the stability of the anti-tumor drug. Several molecules including different fragments of antibodies, cytokines, Human Serum Albumin, transferrin, XTEN polymers, Elastin-like polypeptides (ELPs) can be employed as a fusion partner and the resulting fusion proteins are reported to show enhanced anti-tumor response.
Collapse
Affiliation(s)
- Tehreem Mahmood
- Department of Biotechnology, Quaid-i-azam University, Islamabad, Pakistan
| | - Areej Shahbaz
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Nazim Hussain
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan.
| | - Rahat Ali
- Department of Chemistry, University of Agriculture Faisalabad, Pakistan
| | - Hamid Bashir
- Center for Applied Molecular Biology (CAMB), University of the Punjab, Lahore, Pakistan
| | - Komal Rizwan
- Department of Chemistry, University of Sahiwal, Sahiwal 57000, Pakistan.
| |
Collapse
|
9
|
Kurtzhals P, Østergaard S, Nishimura E, Kjeldsen T. Derivatization with fatty acids in peptide and protein drug discovery. Nat Rev Drug Discov 2023; 22:59-80. [PMID: 36002588 DOI: 10.1038/s41573-022-00529-w] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2022] [Indexed: 01/28/2023]
Abstract
Peptides and proteins are widely used to treat a range of medical conditions; however, they often have to be injected and their effects are short-lived. These shortcomings of the native structure can be addressed by molecular engineering, but this is a complex undertaking. A molecular engineering technology initially applied to insulin - and which has now been successfully applied to several biopharmaceuticals - entails the derivatization of peptides and proteins with fatty acids. Various protraction mechanisms are enabled by the specific characteristics and positions of the attached fatty acid. Furthermore, the technology can ensure a long half-life following oral administration of peptide drugs, can alter the distribution of peptides and may hold potential for tissue targeting. Due to the inherent safety and well-defined chemical nature of the fatty acids, this technology provides a versatile approach to peptide and protein drug discovery.
Collapse
|
10
|
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
|
11
|
Michot N, Guyochin A, Cinier M, Savignard C, Kitten O, Pascual MH, Pouzieux S, Ozoux ML, Verdier P, Vicat P, Dumas J. Albumin binding Nanofitins, a new scaffold to extend half-life of biologics - a case study with exenatide peptide. Peptides 2022; 152:170760. [PMID: 35150805 DOI: 10.1016/j.peptides.2022.170760] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 01/01/2023]
Abstract
A new strategy of peptide half-life extension has been evaluated. We investigated libraries of a small and very stable protein scaffold called Nanofitin, capable of high affinity for protein targets. We have identified Nanofitins targeting Human and mouse Serum Albumin, which could significantly improve the pharmacokinetics of an active associated peptide, mobilizing the patient's own albumin without external source. To demonstrate the impact of this approach on half-life extension, a genetic fusion of an Exenatide peptide with an Albumin Binding Nanofitin (ABNF) was performed. Specific activity of Exenatide-ABNF was measured and unaffected by the fusion. In vivo mice results provided convincing data (t½ of 8 min for Exenatide peptide compared to 20 h for Exenatide-ABNF) with sustained pharmacological activity over 3 days. This study constitutes a proof-of-concept of in vivo half-life extension of a biologic using an ABNF. Besides, the absence of cysteine in the Nanofitin scaffold, which is therefore devoid of structuring disulfide bonds, allows manufacturing in microbial cost effective systems.
Collapse
Affiliation(s)
- Nadine Michot
- Sanofi, Biologics Research, Vitry sur Seine, 94430, France
| | | | | | | | | | | | | | | | - Patrick Verdier
- Sanofi, Drug Safety & Animal Research, Alfortville 94430, France
| | - Pascale Vicat
- Sanofi, Drug Safety & Animal Research, Alfortville 94430, France
| | - Jacques Dumas
- Sanofi, Biologics Research, Vitry sur Seine, 94430, France
| |
Collapse
|
12
|
Zacharias N, Podust VN, Kajihara KK, Leipold D, Del Rosario G, Thayer D, Dong E, Paluch M, Fischer D, Zheng K, Lei C, He J, Ng C, Su D, Liu L, Masih S, Sawyer W, Tinianow J, Marik J, Yip V, Li G, Chuh J, Morisaki JH, Park S, Zheng B, Hernandez-Barry H, Loyet KM, Xu M, Kozak KR, Phillips GL, Shen BQ, Wu C, Xu K, Yu SF, Kamath A, Rowntree RK, Reilly D, Pillow T, Polson A, Schellenberger V, Hazenbos WLW, Sadowsky J. A homogeneous high-DAR antibody-drug conjugate platform combining THIOMAB antibodies and XTEN polypeptides. Chem Sci 2022; 13:3147-3160. [PMID: 35414872 PMCID: PMC8926172 DOI: 10.1039/d1sc05243h] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/27/2022] [Indexed: 11/21/2022] Open
Abstract
The antibody-drug conjugate (ADC) is a well-validated modality for the cell-specific delivery of small molecules with impact expanding rapidly beyond their originally-intended purpose of treating cancer. However, antibody-mediated delivery (AMD)...
Collapse
Affiliation(s)
| | - Vladimir N Podust
- Amunix Pharmaceuticals, Inc. 2 Tower Place South San Francisco CA 94080 USA
| | | | | | | | - Desiree Thayer
- Amunix Pharmaceuticals, Inc. 2 Tower Place South San Francisco CA 94080 USA
| | - Emily Dong
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Maciej Paluch
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - David Fischer
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Kai Zheng
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Corinna Lei
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Jintang He
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Carl Ng
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Dian Su
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Luna Liu
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | - William Sawyer
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Jeff Tinianow
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Jan Marik
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Victor Yip
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Guangmin Li
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Josefa Chuh
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | - Summer Park
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Bing Zheng
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | - Kelly M Loyet
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Min Xu
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | | | - Ben-Quan Shen
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Cong Wu
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Keyang Xu
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Shang-Fan Yu
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Amrita Kamath
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | | | - Thomas Pillow
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | - Andrew Polson
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| | | | | | - Jack Sadowsky
- Genentech, Inc. 1 DNA Way South San Francisco CA 94080 USA
| |
Collapse
|
13
|
Gadolinium-labeled affibody-XTEN recombinant vector for detection of HER2+ lesions of ovarian cancer lung metastasis using quantitative MRI. J Control Release 2021; 337:132-143. [PMID: 34284047 PMCID: PMC8440463 DOI: 10.1016/j.jconrel.2021.07.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 06/01/2021] [Accepted: 07/14/2021] [Indexed: 11/21/2022]
Abstract
Ovarian cancer has the highest mortality rate among all gynecologic malignancies. HER2+ ovarian cancer is a subtype that is aggressive and associated with metastasis to distant sites such as the lungs. Therefore, accurate biological characterization of metastatic lesions is vital as it helps physicians select the most effective treatment strategy. Functional imaging of ovarian cancer with PET/CT is routinely used in the clinic to detect metastatic disease and evaluate treatment response. However, this imaging method does not provide information regarding the presence or absence of cancer-specific cell surface biomarkers such as HER2. As a result, this method does not help physicians decide whether to choose immunotherapy to treat metastasis. To differentiate the HER2+ from HER2¯ lesions in ovarian cancer lung metastasis, AbX50C4:Gd vector composed of a HER2 targeting affibody and XTEN peptide was genetically engineered. It was then labeled with gadolinium (Gd) via a stable linker. The vector was characterized physicochemically and biologically to determine its purity, molecular weight, hydrodynamic size and surface charge, stability in serum, endotoxin levels, relaxivity and ability to target the HER2 antigen. Then, SCID mice were implanted with SKOV-3 (HER2+) and OVASC-1 (HER2¯) tumors in the lungs and injected with the Gd-labeled HER2 targeted AbX50C4:Gd vector. The mice were imaged using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), followed by R1-mapping and quantitative analysis of the images. Our data demonstrate that the developed HER2-targeted vector can differentiate HER2+ lung metastasis from HER2¯ lesions using DCE-MRI. The developed vector could potentially be used in conjunction with other imaging modalities to prescreen patients and identify candidates for immunotherapy while triaging those who may not be considered responsive.
Collapse
|
14
|
Crawford A, Chiu D. Targeting Solid Tumors Using CD3 Bispecific Antibodies. Mol Cancer Ther 2021; 20:1350-1358. [PMID: 34045228 DOI: 10.1158/1535-7163.mct-21-0073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/06/2021] [Accepted: 05/24/2021] [Indexed: 11/16/2022]
Abstract
Immunotherapies to treat cancer have made tremendous progress over the past decade. In particular, T cell-directed therapies have gained considerable attention with CD3 bispecific antibodies and CAR T cells showing potent responses against hematologic tumors. At present, the ability to adapt these therapeutics to treat solid tumors is less established. Herein, we discuss recent advances in T cell-engaging CD3 bispecific antibodies targeting solid tumors, potential mechanisms of resistance, and future prospects. A better understanding of the mechanisms of immune evasion in solid tumors will enable the development of strategies to overcome this resistance and inform choices of therapeutic combinations.
Collapse
Affiliation(s)
| | - Danica Chiu
- Regeneron Pharmaceuticals, Inc., Tarrytown, New York
| |
Collapse
|
15
|
Zinsli LV, Stierlin N, Loessner MJ, Schmelcher M. Deimmunization of protein therapeutics - Recent advances in experimental and computational epitope prediction and deletion. Comput Struct Biotechnol J 2020; 19:315-329. [PMID: 33425259 PMCID: PMC7779837 DOI: 10.1016/j.csbj.2020.12.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/11/2022] Open
Abstract
Biotherapeutics, and antimicrobial proteins in particular, are of increasing interest for human medicine. An important challenge in the development of such therapeutics is their potential immunogenicity, which can induce production of anti-drug-antibodies, resulting in altered pharmacokinetics, reduced efficacy, and potentially severe anaphylactic or hypersensitivity reactions. For this reason, the development and application of effective deimmunization methods for protein drugs is of utmost importance. Deimmunization may be achieved by unspecific shielding approaches, which include PEGylation, fusion to polypeptides (e.g., XTEN or PAS), reductive methylation, glycosylation, and polysialylation. Alternatively, the identification of epitopes for T cells or B cells and their subsequent deletion through site-directed mutagenesis represent promising deimmunization strategies and can be accomplished through either experimental or computational approaches. This review highlights the most recent advances and current challenges in the deimmunization of protein therapeutics, with a special focus on computational epitope prediction and deletion tools.
Collapse
Key Words
- ABR, Antigen-binding region
- ADA, Anti-drug antibody
- ANN, Artificial neural network
- APC, Antigen-presenting cell
- Anti-drug-antibody
- B cell epitope
- BCR, B cell receptor
- Bab, Binding antibody
- CDR, Complementarity determining region
- CRISPR, Clustered regularly interspaced short palindromic repeats
- DC, Dendritic cell
- ELP, Elastin-like polypeptide
- EPO, Erythropoietin
- ER, Endoplasmatic reticulum
- GLK, Gelatin-like protein
- HAP, Homo-amino-acid polymer
- HLA, Human leukocyte antigen
- HMM, Hidden Markov model
- IL, Interleukin
- Ig, Immunoglobulin
- Immunogenicity
- LPS, Lipopolysaccharide
- MHC, Major histocompatibility complex
- NMR, Nuclear magnetic resonance
- Nab, Neutralizing antibody
- PAMP, Pathogen-associated molecular pattern
- PAS, Polypeptide composed of proline, alanine, and/or serine
- PBMC, Peripheral blood mononuclear cell
- PD, Pharmacodynamics
- PEG, Polyethylene glycol
- PK, Pharmacokinetics
- PRR, Pattern recognition receptor
- PSA, Sialic acid polymers
- Protein therapeutic
- RNN, Recurrent artificial neural network
- SVM, Support vector machine
- T cell epitope
- TAP, Transporter associated with antigen processing
- TCR, T cell receptor
- TLR, Toll-like receptor
- XTEN, “Xtended” recombinant polypeptide
Collapse
Affiliation(s)
- Léa V. Zinsli
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Noël Stierlin
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Martin J. Loessner
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| | - Mathias Schmelcher
- Institute of Food, Nutrition and Health, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
16
|
Ibeanu N, Egbu R, Onyekuru L, Javaheri H, Tee Khaw P, R. Williams G, Brocchini S, Awwad S. Injectables and Depots to Prolong Drug Action of Proteins and Peptides. Pharmaceutics 2020; 12:E999. [PMID: 33096803 PMCID: PMC7589296 DOI: 10.3390/pharmaceutics12100999] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 12/30/2022] Open
Abstract
Proteins and peptides have emerged in recent years to treat a wide range of multifaceted diseases such as cancer, diabetes and inflammation. The emergence of polypeptides has yielded advancements in the fields of biopharmaceutical production and formulation. Polypeptides often display poor pharmacokinetics, limited permeability across biological barriers, suboptimal biodistribution, and some proclivity for immunogenicity. Frequent administration of polypeptides is generally required to maintain adequate therapeutic levels, which can limit efficacy and compliance while increasing adverse reactions. Many strategies to increase the duration of action of therapeutic polypeptides have been described with many clinical products having been developed. This review describes approaches to optimise polypeptide delivery organised by the commonly used routes of administration. Future innovations in formulation may hold the key to the continued successful development of proteins and peptides with optimal clinical properties.
Collapse
Affiliation(s)
- Nkiruka Ibeanu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Raphael Egbu
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Lesley Onyekuru
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Hoda Javaheri
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Peng Tee Khaw
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Gareth R. Williams
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
| | - Steve Brocchini
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| | - Sahar Awwad
- School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (N.I.); (R.E.); (L.O.); (H.J.); (G.R.W.); (S.B.)
- National Institute for Health Research (NIHR) Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London EC1V 9EL, UK;
| |
Collapse
|
17
|
Kjeldsen T, Hogendorf WFJ, Tornøe CW, Anderson J, Hubalek F, Stidsen CE, Sorensen JL, Hoeg-Jensen T. Dually Reactive Long Recombinant Linkers for Bioconjugations as an Alternative to PEG. ACS OMEGA 2020; 5:19827-19833. [PMID: 32803078 PMCID: PMC7424725 DOI: 10.1021/acsomega.0c02712] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/22/2020] [Indexed: 06/11/2023]
Abstract
Covalent cross-linking of biomolecules can be useful in pursuit of tissue targeting or dual targeting of two receptors on cell surfaces for avidity effects. Long linkers (>10 kDa) can be advantageous for such purposes, and poly(ethylene glycol) (PEG) linkers are most commonly used due to the high aqueous solubility of PEG and its relative inertness toward biological targets. However, PEG is non-biodegradable, and available PEG linkers longer than 5 kDa are heterogeneous (polydisperse), which means that conjugates based on such materials will be mixtures. We describe here recombinant linkers of distinct lengths, which can be expressed in yeast, which are polar, and which carry orthogonal reactivity at each end of the linker, thus allowing chemoselective cross-linking of proteins. A conjugate between insulin and either of the two trypsin inhibitor peptides/proteins exemplifies the technology, using a GQAP-based linker of molecular weight of 17 848, having one amine at the N-terminal, and one Cys, at the C-terminal. Notably, yeast-based expression systems typically give products with mixed disulfides when expressing proteins that are equipped with one unpaired Cys, namely, mixed disulfides with glutathione, free Cys amino acid, and/or a protein homodimer. To obtain a homogeneous linker, we worked out conditions for transforming the linker with mixed disulfides into a linker with a homogeneous disulfide, using excess 4-mercaptophenylacetic acid. Subsequently, the N-terminal amine of the linker was transformed into an azide, and the C-terminal Cys disulfide was reduced to a free thiol and reacted with halo-acetyl insulin. The N-terminal azide was finally conjugated to either of the two types of alkyne-containing trypsin inhibitor peptides/proteins. This reaction sequence allowed the cross-linked proteins to carry internal disulfides, as no reduction step was needed after protein conjugations. The insulin-trypsin inhibitor conjugates were shown to be stabilized toward enzymatic digestions and to have partially retained binding to the insulin receptor.
Collapse
|
18
|
Iyengar ARS, Gupta S, Jawalekar S, Pande AH. Protein Chimerization: A New Frontier for Engineering Protein Therapeutics with Improved Pharmacokinetics. J Pharmacol Exp Ther 2019; 370:703-714. [PMID: 31010843 DOI: 10.1124/jpet.119.257063] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 04/16/2019] [Indexed: 03/08/2025] Open
Abstract
With the advancement of medicine, the utility of protein therapeutics is increasing exponentially. However, a significant number of protein therapeutics suffer from grave limitations, which include their subpar pharmacokinetics. In this study, we have reviewed the emerging field of protein chimerization for improving the short circulatory half-life of protein therapeutics. We have discussed various aspects of protein therapeutics aiming at their mechanism of clearance and various approaches used to increase their short circulatory half-life with principal focus on the concept of chimerization. Furthermore, we have comprehensively reviewed various components of chimera, such as half-life extension partners and linkers, their shortcomings, and prospective work to be undertaken for developing effective chimeric protein therapeutics.
Collapse
Affiliation(s)
- A R Satvik Iyengar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
| | - Shreya Gupta
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
| | - Snehal Jawalekar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
| | - Abhay H Pande
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
| |
Collapse
|
19
|
Novel therapeutic interventions in cancer treatment using protein and peptide-based targeted smart systems. Semin Cancer Biol 2019; 69:249-267. [PMID: 31442570 DOI: 10.1016/j.semcancer.2019.08.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 01/12/2023]
Abstract
Cancer, being the most prevalent and resistant disease afflicting any gender, age or social status, is the ultimate challenge for the scientific community. The new generation therapeutics for cancer management has shifted the approach to personalized/precision medicine, making use of patient- and tumor-specific markers for specifying the targeted therapies for each patient. Peptides targeting these cancer-specific signatures hold enormous potential for cancer therapy and diagnosis. The rapid advancements in the combinatorial peptide libraries served as an impetus to the development of multifunctional peptide-based materials for targeted cancer therapy. The present review outlines benefits and shortcomings of peptides as cancer therapeutics and the potential of peptide modified nanomedicines for targeted delivery of anticancer agents.
Collapse
|
20
|
Brandl F, Merten H, Zimmermann M, Béhé M, Zangemeister-Wittke U, Plückthun A. Influence of size and charge of unstructured polypeptides on pharmacokinetics and biodistribution of targeted fusion proteins. J Control Release 2019; 307:379-392. [PMID: 31252038 DOI: 10.1016/j.jconrel.2019.06.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 01/19/2023]
Abstract
Alternative non-IgG binding proteins developed for therapy are small in size and, thus, are rapidly cleared from the circulation by renal filtration. To avoid repeated injection or continuous infusion for the maintenance of therapeutic serum concentrations, extensions of unfolded polypeptides have been developed to prolong serum half-life, but systematic, comparative studies investigating the influence of their size and charge on serum half-life, extravasation, tumor localization and excretion mechanisms have so far been lacking. Here we used a high-affinity Designed Ankyrin Repeat Protein (DARPin) targeting the tumor marker epithelial cell adhesion molecule (EpCAM) in a preclinical tumor xenograft model in mice, and fused it with a series of defined unstructured polypeptides. We used three different sizes of two previously described polypeptides, an uncharged one consisting of only Pro, Ala and Ser (termed PAS) and a charged one consisting of Pro, Ala, Ser, Thr, Gly, Glu (termed XTEN) and performed for the first time a precise comparative localization, distribution and extravasation study. Pharmacokinetic analysis showed a clear linear relationship between hydrodynamic radius and serum half-life across both polypeptides, reaching a half-life of up to 21 h in mice. Tumor uptake was EpCAM-dependent and directly proportional to half-life and size, showing an even tumor penetration for all fusion proteins without unspecific accumulation in non-target tissue. Unexpectedly, charge had no influence on any parameter, neither tumor nor tissue accumulation nor kidney elimination kinetics. Thus, both polypeptide types have a very similar potential for precise half-life modification and tumor targeting.
Collapse
Affiliation(s)
- Fabian Brandl
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Institute of Pharmacology, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland
| | - Hannes Merten
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Martina Zimmermann
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland
| | - Martin Béhé
- Center of Radiopharmaceutical Sciences, Paul Scherrer Institute, CH-5232 Villigen PSI, Switzerland
| | - Uwe Zangemeister-Wittke
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland; Institute of Pharmacology, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland.
| | - Andreas Plückthun
- Department of Biochemistry, University of Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| |
Collapse
|
21
|
Enhancing bioactivity, physicochemical, and pharmacokinetic properties of a nano-sized, anti-VEGFR2 Adnectin, through PASylation technology. Sci Rep 2019; 9:2978. [PMID: 30814652 PMCID: PMC6393559 DOI: 10.1038/s41598-019-39776-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 02/01/2019] [Indexed: 02/07/2023] Open
Abstract
The crucial role of VEGF receptor 2 (VEGFR2) signaling in the angiogenesis and metastasis of solid tumors has prompted the development of inhibitors with minimal bystander effects. Recently, Adnectin C has attracted attention for cancer treatment. To overcome the problematic properties of Adnectin, a novel form of Adnectin C has been designed by its fusion to a biodegradable polymeric peptide containing Pro/Ala/Ser (PAS) repetitive residues. E. coli-expressed recombinant fused and unfused proteins were compared in terms of bioactivity, physicochemical, and pharmacokinetic properties using standard methods. Dynamic light scattering (DLS) analysis of PASylated adnectin C revealed an approximate 2-fold increase in particle size with a slight change in the net charge. Additionally, fusion of the PAS sequence improved its stability against the growth of thermo-induced aggregated forms. The high receptor-binding and improved binding kinetic parameters of PASylated Adnectin C was confirmed by ELISA and surface plasmon resonance assays, respectively. Pharmacokinetic studies showed a noticeable increase in the terminal half-life of Adnectin C-PAS#1(200) by a factor of 4.57 after single dose by intravenous injection into female BALB/c mice. The results suggest that PASylation could offer a superior delivery strategy for developing Adnectin-derived drugs with improved patient compliance.
Collapse
|
22
|
Liu X, Sun J, Gao W. Site-selective protein modification with polymers for advanced biomedical applications. Biomaterials 2018; 178:413-434. [DOI: 10.1016/j.biomaterials.2018.04.050] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 04/21/2018] [Accepted: 04/24/2018] [Indexed: 12/12/2022]
|
23
|
Zelikin AN, Ehrhardt C, Healy AM. Materials and methods for delivery of biological drugs. Nat Chem 2018; 8:997-1007. [PMID: 27768097 DOI: 10.1038/nchem.2629] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 08/26/2016] [Indexed: 12/23/2022]
Abstract
Biological drugs generated via recombinant techniques are uniquely positioned due to their high potency and high selectivity of action. The major drawback of this class of therapeutics, however, is their poor stability upon oral administration and during subsequent circulation. As a result, biological drugs have very low bioavailability and short therapeutic half-lives. Fortunately, tools of chemistry and biotechnology have been developed into an elaborate arsenal, which can be applied to improve the pharmacokinetics of biological drugs. Depot-type release systems are available to achieve sustained release of drugs over time. Conjugation to synthetic or biological polymers affords long circulating formulations. Administration of biological drugs through non-parenteral routes shows excellent performance and the first products have reached the market. This Review presents the main accomplishments in this field and illustrates the materials and methods behind existing and upcoming successful formulations and delivery strategies for biological drugs.
Collapse
Affiliation(s)
- Alexander N Zelikin
- Department of Chemistry, Aarhus University, Aarhus C 8000, Denmark.,iNano Interdisciplinary Nanoscience Centre, Aarhus University, Aarhus C 8000, Denmark
| | - Carsten Ehrhardt
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland.,Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| | - Anne Marie Healy
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland.,Synthesis and Solid State Pharmaceutical Centre, School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research (AMBER) Centre, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
24
|
Shoombuatong W, Schaduangrat N, Nantasenamat C. Unraveling the bioactivity of anticancer peptides as deduced from machine learning. EXCLI JOURNAL 2018; 17:734-752. [PMID: 30190664 PMCID: PMC6123611 DOI: 10.17179/excli2018-1447] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/10/2018] [Indexed: 12/13/2022]
Abstract
Cancer imposes a global health burden as it represents one of the leading causes of morbidity and mortality while also giving rise to significant economic burden owing to the associated expenditures for its monitoring and treatment. In spite of advancements in cancer therapy, the low success rate and recurrence of tumor has necessitated the ongoing search for new therapeutic agents. Aside from drugs based on small molecules and protein-based biopharmaceuticals, there has been an intense effort geared towards the development of peptide-based therapeutics owing to its favorable and intrinsic properties of being relatively small, highly selective, potent, safe and low in production costs. In spite of these advantages, there are several inherent weaknesses that are in need of attention in the design and development of therapeutic peptides. An abundance of data on bioactive and therapeutic peptides have been accumulated over the years and the burgeoning area of artificial intelligence has set the stage for the lucrative utilization of machine learning to make sense of these large and high-dimensional data. This review summarizes the current state-of-the-art on the application of machine learning for studying the bioactivity of anticancer peptides along with future outlook of the field. Data and R codes used in the analysis herein are available on GitHub at https://github.com/Shoombuatong2527/anticancer-peptides-review.
Collapse
Affiliation(s)
- Watshara Shoombuatong
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Nalini Schaduangrat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| | - Chanin Nantasenamat
- Center of Data Mining and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
25
|
Yu M, Benjamin MM, Srinivasan S, Morin EE, Shishatskaya EI, Schwendeman SP, Schwendeman A. Battle of GLP-1 delivery technologies. Adv Drug Deliv Rev 2018; 130:113-130. [PMID: 30009885 PMCID: PMC6843995 DOI: 10.1016/j.addr.2018.07.009] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/26/2018] [Accepted: 07/09/2018] [Indexed: 12/25/2022]
Abstract
Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) belong to an important therapeutic class for treatment of type 2 diabetes. Six GLP-1 RAs, each utilizing a unique drug delivery strategy, are now approved by the Food and Drug Administration (FDA) and additional, novel GLP-1 RAs are still under development, making for a crowded marketplace and fierce competition among the manufacturers of these products. As rapid elimination is a major challenge for clinical application of GLP-1 RAs, various half-life extension strategies have been successfully employed including sequential modification, attachment of fatty-acid to peptide, fusion with human serum albumin, fusion with the fragment crystallizable (Fc) region of a monoclonal antibody, sustained drug delivery systems, and PEGylation. In this review, we discuss the scientific rationale of the various half-life extension strategies used for GLP-1 RA development. By analyzing and comparing different approved GLP-1 RAs and those in development, we focus on assessing how half-life extending strategies impact the pharmacokinetics, pharmacodynamics, safety, patient usability and ultimately, the commercial success of GLP-1 RA products. We also anticipate future GLP-1 RA development trends. Since similar drug delivery strategies are also applied for developing other therapeutic peptides, we expect this case study of GLP-1 RAs will provide generalizable concepts for the rational design of therapeutic peptides products with extended duration of action.
Collapse
Affiliation(s)
- Minzhi Yu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States of America
| | - Mason M Benjamin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States of America
| | | | - Emily E Morin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States of America
| | - Ekaterina I Shishatskaya
- Siberian Federal University, 79 Svobodnuy Ave, Krasnoyarsk 660041, Russian Federation; Institute of Biophysics SBRAS, 50 Akademgorodok, 660036, Russian Federation
| | - Steven P Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States of America; Biointerfaces Institute, NCRC, 2800 Plymouth Rd, Ann Arbor, MI 48109, United States of America; Department of Biomedical Engineering, 2200 Bonisteel Blvd, Ann Arbor, MI 48109, United States of America.
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St, Ann Arbor, MI 48109, United States of America; Biointerfaces Institute, NCRC, 2800 Plymouth Rd, Ann Arbor, MI 48109, United States of America.
| |
Collapse
|
26
|
Balkaransingh P, Young G. Novel therapies and current clinical progress in hemophilia A. Ther Adv Hematol 2018; 9:49-61. [PMID: 29387330 PMCID: PMC5768270 DOI: 10.1177/2040620717746312] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 10/30/2017] [Indexed: 12/19/2022] Open
Abstract
The evolution of hemophilia treatment and care is a fascinating one but has been fraught with many challenges at every turn. Over the last 50 years or so patients with hemophilia and providers have witnessed great advances in the treatment of this disease. With these advances, there has been a dramatic decrease in the mortality and morbidity associated with hemophilia. Even with the remarkable advancements in treatment, however, new and old challenges continue to plague the hemophilia community. The cost of factor replacement and the frequency of infusions, especially in patients with severe hemophilia on prophylaxis, remains a significant challenge for this population. Other challenges include obtaining reliable venous access, especially in younger patients, and the development of neutralizing alloantibodies (inhibitors). The development of extended half-life products, a bispecific antibody which mimics the coagulation function of factor VIII (FVIII) and inhibition of anticoagulation proteins such as antithrombin with antibodies, aptamers or RNA interference technology have offered novel therapeutic approaches to overcome some of these existing challenges. Additionally, ongoing gene therapy research offers a way to possibly cure hemophilia. These novel treatment tools in conjunction with the establishment of an increasing number of comprehensive hemophilia centers and worldwide advocacy efforts have continued to push the progress of hemophilia care to new frontiers. This review highlights and summarizes these novel therapeutic approaches and the current clinical progress of hemophilia A.
Collapse
Affiliation(s)
| | - Guy Young
- Children’s Hospital Los Angeles, 455 Sunset Boulevard, Mail Stop 54, Los Angeles, CA 90027, USA
| |
Collapse
|
27
|
Kim D, Jeon H, Ahn S, Choi WI, Kim S, Jon S. An approach for half-life extension and activity preservation of an anti-diabetic peptide drug based on genetic fusion with an albumin-binding aptide. J Control Release 2017; 256:114-120. [DOI: 10.1016/j.jconrel.2017.04.036] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 04/19/2017] [Accepted: 04/25/2017] [Indexed: 10/19/2022]
|
28
|
Marqus S, Pirogova E, Piva TJ. Evaluation of the use of therapeutic peptides for cancer treatment. J Biomed Sci 2017; 24:21. [PMID: 28320393 PMCID: PMC5359827 DOI: 10.1186/s12929-017-0328-x] [Citation(s) in RCA: 338] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/14/2017] [Indexed: 12/25/2022] Open
Abstract
Cancer along with cardiovascular disease are the main causes of death in the industrialised countries around the World. Conventional cancer treatments are losing their therapeutic uses due to drug resistance, lack of tumour selectivity and solubility and as such there is a need to develop new therapeutic agents. Therapeutic peptides are a promising and a novel approach to treat many diseases including cancer. They have several advantages over proteins or antibodies: as they are (a) easy to synthesise, (b) have a high target specificity and selectivity and (c) have low toxicity. Therapeutic peptides do have some significant drawbacks related to their stability and short half-life. In this review, strategies used to overcome peptide limitations and to enhance their therapeutic effect will be compared. The use of short cell permeable peptides that interfere and inhibit protein-protein interactions will also be evaluated.
Collapse
Affiliation(s)
- Susan Marqus
- School of Engineering, RMIT University, Bundoora, VIC 3083 Australia
| | - Elena Pirogova
- School of Engineering, RMIT University, Bundoora, VIC 3083 Australia
| | - Terrence J. Piva
- School of Health and Biomedical Sciences, RMIT University, Bundoora, VIC 3083 Australia
| |
Collapse
|
29
|
van Witteloostuijn SB, Pedersen SL, Jensen KJ. Half-Life Extension of Biopharmaceuticals using Chemical Methods: Alternatives to PEGylation. ChemMedChem 2016; 11:2474-2495. [DOI: 10.1002/cmdc.201600374] [Citation(s) in RCA: 126] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/24/2016] [Indexed: 12/21/2022]
Affiliation(s)
- Søren B. van Witteloostuijn
- Department of Chemistry; University of Copenhagen; Thorvaldsensvej 40 1871 Frederiksberg C Denmark
- Gubra ApS; Hørsholm Kongevej 11B 2970 Hørsholm Denmark
| | | | - Knud J. Jensen
- Department of Chemistry; University of Copenhagen; Thorvaldsensvej 40 1871 Frederiksberg C Denmark
| |
Collapse
|
30
|
Long-Acting Injections and Implants. Drug Deliv 2016. [DOI: 10.1201/9781315382579-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
|
31
|
Connor EE, Evock-Clover CM, Wall EH, Baldwin RL, Santin-Duran M, Elsasser TH, Bravo DM. Glucagon-like peptide 2 and its beneficial effects on gut function and health in production animals. Domest Anim Endocrinol 2016; 56 Suppl:S56-65. [PMID: 27345324 DOI: 10.1016/j.domaniend.2015.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Revised: 11/17/2015] [Accepted: 11/26/2015] [Indexed: 12/12/2022]
Abstract
Numerous endocrine cell subtypes exist within the intestinal mucosa and produce peptides contributing to the regulation of critical physiological processes including appetite, energy metabolism, gut function, and gut health. The mechanisms of action and the extent of the physiological effects of these enteric peptides are only beginning to be uncovered. One peptide in particular, glucagon-like peptide 2 (GLP-2) produced by enteroendocrine L cells, has been fairly well characterized in rodent and swine models in terms of its ability to improve nutrient absorption and healing of the gut after injury. In fact, a long-acting form of GLP-2 recently has been approved for the management and treatment of human conditions like inflammatory bowel disease and short bowel syndrome. However, novel functions of GLP-2 within the gut continue to be demonstrated, including its beneficial effects on intestinal barrier function and reducing intestinal inflammation. As knowledge continues to grow about GLP-2's effects on the gut and its mechanisms of release, the potential to use GLP-2 to improve gut function and health of food animals becomes increasingly more apparent. Thus, the purpose of this review is to summarize: (1) the current understanding of GLP-2's functions and mechanisms of action within the gut; (2) novel applications of GLP-2 (or stimulators of its release) to improve general health and production performance of food animals; and (3) recent findings, using dairy calves as a model, that suggest the therapeutic potential of GLP-2 to reduce the pathogenesis of intestinal protozoan infections.
Collapse
Affiliation(s)
- E E Connor
- US Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705 USA.
| | - C M Evock-Clover
- US Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705 USA
| | - E H Wall
- Pancosma S.A., CH-1218 Geneva, Switzerland
| | - R L Baldwin
- US Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705 USA
| | - M Santin-Duran
- US Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705 USA
| | - T H Elsasser
- US Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Beltsville, MD 20705 USA
| | - D M Bravo
- Pancosma S.A., CH-1218 Geneva, Switzerland
| |
Collapse
|
32
|
Griswold KE, Bailey-Kellogg C. Design and engineering of deimmunized biotherapeutics. Curr Opin Struct Biol 2016; 39:79-88. [PMID: 27322891 DOI: 10.1016/j.sbi.2016.06.003] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 06/03/2016] [Accepted: 06/06/2016] [Indexed: 12/26/2022]
Abstract
Therapeutic proteins are powerful next-generation drugs able to effectively treat diverse and devastating diseases, but the development and use of biotherapeutics entails unique challenges and risks. In particular, protein drugs are subject to immune surveillance in the human body, and ensuing antidrug immune responses can cause a wide range of problems including altered pharmacokinetics, loss of efficacy, and even life-threating complications. Here we review recent progress in technologies for engineering deimmunized biotherapeutics, placing particular emphasis on deletion of immunogenic antibody and T cell epitopes via experimentally or computationally guided mutagenesis.
Collapse
Affiliation(s)
- Karl E Griswold
- Thayer School of Engineering, Dartmouth, Hanover, NH, United States; Stealth Biologics LLC, Lyme, NH, United States.
| | - Chris Bailey-Kellogg
- Stealth Biologics LLC, Lyme, NH, United States; Department of Computer Science, Dartmouth, Hanover, NH, United States.
| |
Collapse
|
33
|
Connor EE, Evock-Clover CM, Walker MP, Elsasser TH, Kahl S. COMPARATIVE GUT PHYSIOLOGY SYMPOSIUM: Comparative physiology of glucagon-like peptide-2: Implications and applications for production and health of ruminants. J Anim Sci 2016; 93:492-501. [PMID: 26020740 DOI: 10.2527/jas.2014-8577] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Glucagon-like peptide-2 (GLP-2) is a 33-amino acid peptide derived from proteolytic cleavage of proglucagon by prohormone convertase 1/3 in enteroendocrine L cells. Studies conducted in humans, in rodent models, and in vitro indicate that GLP-2 is secreted in response to the presence of molecules in the intestinal lumen, including fatty acids, carbohydrates, amino acids, and bile acids, which are detected by luminal chemosensors. The physiological actions of GLP-2 are mediated by its G protein-coupled receptor expressed primarily in the intestinal tract on enteric neurons, enteroendocrine cells, and myofibroblasts. The biological activity of GLP-2 is further regulated by dipeptidyl peptidase IV, which rapidly cleaves the N-terminus of GLP-2 that is responsible for GLP-2 receptor activation. Within the gut, GLP-2 increases nutrient absorption, crypt cell proliferation, and mesenteric blood flow and decreases gut permeability and motility, epithelial cell apoptosis, and inflammation. Outside the gut, GLP-2 reduces bone resorption, can suppress appetite, and is cytoprotective in the lung. Thus, GLP-2 has been studied intensively as a therapeutic to improve intestinal function of humans during parenteral nutrition and following small bowel resection and, more recently, as a treatment for osteoporosis and obesity-related disorders and to reduce cellular damage associated with inflammation of the gut and lungs. Recent studies demonstrate that many biological actions and properties of GLP-2 in ruminants are similar to those in nonruminants, including the potential to reduce intestinal nitro-oxidative stress in calves caused by parasitic diseases such as coccidiosis. Because of its beneficial impacts on nutrient absorption, gut healing, and normal gut development, GLP-2 therapy offers significant opportunities to improve calf health and production efficiency. However, GLP-2 therapies require an extended time course to achieve desired physiological responses, as well as daily administration because of the hormone's short half-life. Thus, practical means of administration and alternative strategies to enhance basal GLP-2 secretion (e.g., through specific feed additives), which are more likely to achieve consumer acceptance, are needed. Opportunities to address these challenges are discussed.
Collapse
|
34
|
Podust VN, Balan S, Sim BC, Coyle MP, Ernst U, Peters RT, Schellenberger V. Extension of in vivo half-life of biologically active molecules by XTEN protein polymers. J Control Release 2015; 240:52-66. [PMID: 26497931 DOI: 10.1016/j.jconrel.2015.10.038] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 10/16/2015] [Accepted: 10/20/2015] [Indexed: 02/03/2023]
Abstract
XTEN™ is a class of unstructured hydrophilic, biodegradable protein polymers designed to increase the half-lives of therapeutic peptides and proteins. XTEN polymers and XTEN fusion proteins are typically expressed in Escherichia coli and purified by conventional protein chromatography as monodisperse polypeptides of exact length and sequence. Unstructured XTEN polypeptides have hydrodynamic volumes significantly larger than typical globular proteins of similar mass, thus imparting a bulking effect to the therapeutic payloads attached to them. Since their invention, XTEN polypeptides have been utilized to extend the half-lives of a variety of peptide- and protein-based therapeutics. Multiple clinical and preclinical studies and related drug discovery and development efforts are in progress. This review details the most current understanding of physicochemical properties and biological behavior of XTEN and XTENylated molecules. Additionally, the development path and status of several advanced drug discovery and development efforts are highlighted.
Collapse
Affiliation(s)
| | - Sibu Balan
- Amunix, 500 Ellis Street, Mountain View, CA 94043, USA
| | - Bee-Cheng Sim
- Amunix, 500 Ellis Street, Mountain View, CA 94043, USA
| | | | - Ulrich Ernst
- Amunix, 500 Ellis Street, Mountain View, CA 94043, USA
| | | | | |
Collapse
|
35
|
Kim SJ, Kwak HH, Cho SY, Sohn YB, Park SW, Huh R, Kim J, Ko AR, Jin DK. Pharmacokinetics, Pharmacodynamics, and Efficacy of a Novel Long-Acting Human Growth Hormone: Fc Fusion Protein. Mol Pharm 2015; 12:3759-65. [DOI: 10.1021/acs.molpharmaceut.5b00550] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Su Jin Kim
- Department
of Pediatrics, Myongji Hospital, Seonam University College of Medicine, Goyang, Republic of Korea
| | - Hyun-Hee Kwak
- Biopharmaceutical
Research Laboratoy, Dong-A Socio Holdings Research Institute, Gyeonggi, Republic of Korea
| | - Sung Yoon Cho
- Department
of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Young Bae Sohn
- Department
of Medical Genetics, Ajou University Hospital, Suwon, Republic of Korea
| | - Sung Won Park
- Department
of Pediatrics, Jeil Hospital, Dankook University College of Medicine, Seoul, Republic of Korea
| | - Rimm Huh
- Department
of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jinsup Kim
- Department
of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Ah-ra Ko
- Clinical
Research Center, Samsung Biomedical Research Institute, Seoul, Republic of Korea
| | - Dong-Kyu Jin
- Department
of Pediatrics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| |
Collapse
|
36
|
Hossain MA, Haugaard-Kedström LM, Rosengren KJ, Bathgate RAD, Wade JD. Chemically synthesized dicarba H2 relaxin analogues retain strong RXFP1 receptor activity but show an unexpected loss of in vitro serum stability. Org Biomol Chem 2015; 13:10895-903. [PMID: 26368576 DOI: 10.1039/c5ob01539a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Peptides and proteins are now acknowledged as viable alternatives to small molecules as potential therapeutic agents. A primary limitation to their more widespread acceptance is their generally short in vivo half-lives due to serum enzyme susceptibility and rapid renal clearance. Numerous chemical approaches to address this concern have been undertaken in recent years. The replacement of disulfide bonds with non-reducible elements has been demonstrated to be one effective means by eliminating the deleterious effect of serum reductases. In particular, substitution with dicarba bonds via ring closure metathesis has been increasingly applied to many bioactive cystine-rich peptides. We used this approach for the replacement of the A-chain intramolecular disulfide bond of human relaxin 2 (H2 relaxin), an insulin-like peptide that has important regulatory roles in cardiovascular and connective tissue homeostasis that has led to successful Phase IIIa clinical trials for the treatment of acute heart failure. Use of efficient solid phase synthesis of the two peptide chains was followed by on-resin ring closure metathesis and formation of the dicarba bond within the A-chain and then by off-resin combination with the B-chain via sequential directed inter-chain disulfide bond formation. After purification and comprehensive chemical characterization, the two isomeric synthetic H2 relaxin analogues were shown to retain near-equipotent RXFP1 receptor binding and activation propensity. Unexpectedly, the in vitro serum stability of the analogues was greatly reduced compared with the native peptide. Circular dichroism spectroscopy studies showed subtle differences in the secondary structures between dicarba analogues and H2 relaxin suggesting that, although the overall fold is retained, it may be destabilized which could account for rapid degradation of dicarba analogues in serum. Caution is therefore recommended when using ring closure metathesis as a general approach to enhance peptide stability.
Collapse
Affiliation(s)
- Mohammed Akhter Hossain
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia.
| | | | | | | | | |
Collapse
|
37
|
|
38
|
Human Therapies as a Successful Liaison between Chemistry and Biology. ACTA ACUST UNITED AC 2014; 21:1046-54. [DOI: 10.1016/j.chembiol.2014.08.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 08/25/2014] [Accepted: 08/27/2014] [Indexed: 12/24/2022]
|
39
|
Xu J, Jung K, Corrigan NA, Boyer C. Aqueous photoinduced living/controlled polymerization: tailoring for bioconjugation. Chem Sci 2014. [DOI: 10.1039/c4sc01309c] [Citation(s) in RCA: 168] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
40
|
Ding S, Song M, Sim BC, Gu C, Podust VN, Wang CW, McLaughlin B, Shah TP, Lax R, Gast R, Sharan R, Vasek A, Hartman MA, Deniston C, Srinivas P, Schellenberger V. Multivalent antiviral XTEN-peptide conjugates with long in vivo half-life and enhanced solubility. Bioconjug Chem 2014; 25:1351-9. [PMID: 24932887 PMCID: PMC4157762 DOI: 10.1021/bc500215m] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
XTENs are unstructured, nonrepetitive
protein polymers designed
to prolong the in vivo half-life of pharmaceuticals by introducing
a bulking effect similar to that of poly(ethylene glycol). While XTEN
can be expressed as a recombinant fusion protein with bioactive proteins
and peptides, therapeutic molecules of interest can also be chemically
conjugated to XTEN. Such an approach permits precise control over
the positioning, spacing, and valency of bioactive moieties along
the length of XTEN. We have demonstrated the attachment of T-20, an
anti-retroviral peptide indicated for the treatment of HIV-1 patients
with multidrug resistance, to XTEN. By reacting maleimide-functionalized
T-20 with cysteine-containing XTENs and varying the number and positioning
of cysteines in the XTENs, a library of different peptide–polymer
combinations were produced. The T-20-XTEN conjugates were tested using
an in vitro antiviral assay and were found to be effective in inhibiting
HIV-1 entry and preventing cell death, with the copy number and spacing
of the T-20 peptides influencing antiviral activity. The peptide–XTEN
conjugates were also discovered to have enhanced solubilities in comparison
with the native T-20 peptide. The pharmacokinetic profile of the most
active T-20-XTEN conjugate was measured in rats, and it was found
to exhibit an elimination half-life of 55.7 ± 17.7 h, almost
20 times longer than the reported half-life for T-20 dosed in rats.
As the conjugation of T-20 to XTEN greatly improved the in vivo half-life
and solubility of the peptide, the XTEN platform has been demonstrated
to be a versatile tool for improving the properties of drugs and enabling
the development of a class of next-generation therapeutics.
Collapse
Affiliation(s)
- Sheng Ding
- Amunix Operating Inc. , 500 Ellis Street, Mountain View, California 94043 United States
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|