1
|
Baral KC, Choi KY. Barriers and Strategies for Oral Peptide and Protein Therapeutics Delivery: Update on Clinical Advances. Pharmaceutics 2025; 17:397. [PMID: 40284395 PMCID: PMC12030352 DOI: 10.3390/pharmaceutics17040397] [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: 02/17/2025] [Revised: 03/17/2025] [Accepted: 03/17/2025] [Indexed: 04/29/2025] Open
Abstract
Peptide and protein (PP) therapeutics are highly specific and potent biomolecules that treat chronic and complex diseases. However, their oral delivery is significantly hindered by enzymatic degradation, instability, and poor permeability through the gastrointestinal (GI) epithelium, resulting in low bioavailability. Various strategies have emerged as transformative solutions to address existing challenges, offering enhanced protection, stabilization, and absorption of PPs. These strategies primarily focus on two major challenges: protecting the PP against harsh conditions and enhancing permeation across the intestinal membrane. Innovative approaches such as pH modulation and incorporation of enzyme inhibitors are usually used to mitigate proteolytic degradation of PP during transit across the GI tract. In a similar vein, absorption enhancers and prodrug strategies facilitate epithelial transport, while targeted delivery systems focus on specific areas of the GI tract to enhance absorption. Likewise, mucus-penetrating and mucoadhesive strategies have enhanced retention and interaction with epithelial cells, effectively overcoming barriers like the mucus layer and tight epithelial junctions. Furthermore, structural modifications such as lipidation, peptide cyclization, and polyethylene glycosylation are promising alternatives to render stability, prolong circulation time, and membrane permeability. In particular, functional biomaterials, active targeting, and lymphatic transport strategies have provided new platforms for oral PP delivery. Advancing in materials science, nanotechnology, and the disruption of medical devices holds new frontiers to overcome barriers. Despite substantial advancements, the limited success in clinical translation underscores the urgency of innovative strategies. This review presents oral PPs as a promising platform, highlighting the key barriers and strategies to transform their therapeutic landscapes.
Collapse
Affiliation(s)
- Kshitis Chandra Baral
- Department of Marine Bio-Food Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea;
| | - Ki Young Choi
- Department of Marine Bio-Food Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea;
- NVience Inc., Seoul 04323, Republic of Korea
| |
Collapse
|
2
|
López-Laguna H, Favaro MTP, Chellou-Bakkali S, Voltà-Durán E, Parladé E, Sánchez J, Corchero JL, Unzueta U, Villaverde A, Vázquez E. Citrate-Assisted Regulation of Protein Stability and Secretability from Synthetic Amyloids. ACS APPLIED MATERIALS & INTERFACES 2025; 17:14940-14951. [PMID: 40009529 DOI: 10.1021/acsami.4c20784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
The mammalian endocrine system uses functional amyloids as dynamic depots to store and release protein hormones into the bloodstream. Such depots, acting as secretory granules within the microscale, are formed in specialized cells by the coordination between the ionic, divalent form of zinc (Zn2+) and the imidazole ring from accessible His residues. The reversibility of such cross-linking events allows for the release of monomeric or oligomeric forms of the functional protein for biological activity. In vitro, and mimicking such a natural coordination process, synthetic amyloidal granules with secretory properties can be fabricated using selected therapeutic proteins as building blocks. Then, these microparticles act as delivery systems for endocrine-like, sustained protein release, with proven applicability in vaccinology, cancer therapy, regenerative medicine, and as antimicrobial agents. While the temporal profile in which the protein is leaked from the material might be highly relevant to clinically oriented applications, the fine control of such parameters remains unclear. We have explored here how the kinetics of protein release can be regulated by intervening in the storage formulation of the granules, through the concentration of citrate not only as a buffer component and protein stabilizer but also as a chelating agent. The citrate-assisted, time-prolonged regulatable release of proteins, in their functional form, opens a spectrum of possibilities to adjust the preparation of synthetic secretory granules to specific clinical needs.
Collapse
Affiliation(s)
- Hèctor López-Laguna
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Marianna T P Favaro
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Sara Chellou-Bakkali
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Eric Voltà-Durán
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Eloi Parladé
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Julieta Sánchez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departamento de Química, Cátedra de Química Biológica, Facultad de Ciencias Exactas, Físicas y Naturales, ICTA, Universidad Nacional de Córdoba, Av. Vélez Sársfield 1611, Córdoba 5016, Argentina
- Instituto de Investigaciones Biológicas y Tecnológicas (IIByT), CONICET-Universidad Nacional de Córdoba, Córdoba 5016, Argentina
| | - José Luis Corchero
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Ugutz Unzueta
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Institut de Recerca Sant Pau (IR SANT PAU), Barcelona 08041, Spain
| | - Antonio Villaverde
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| | - Esther Vázquez
- Institut de Biotecnologia i de Biomedicina, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN, ISCIII), Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
- Departament de Genètica i de Microbiologia, Universitat Autònoma de Barcelona, Bellaterra 08193, Spain
| |
Collapse
|
3
|
Jiao Q, Huang Y, He J, Xu Y. Advances in Oral Biomacromolecule Therapies for Metabolic Diseases. Pharmaceutics 2025; 17:238. [PMID: 40006605 PMCID: PMC11859201 DOI: 10.3390/pharmaceutics17020238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Metabolic diseases like obesity and diabetes are on the rise, and therapies with biomacromolecules (such as proteins, peptides, antibodies, and oligonucleotides) play a crucial role in their treatment. However, these drugs are traditionally injected. For patients with chronic diseases (e.g., metabolic diseases), long-term injections are accompanied by inconvenience and low compliance. Oral administration is preferred, but the delivery of biomacromolecules is challenging due to gastrointestinal barriers. In this article, we introduce the available biomacromolecule drugs for the treatment of metabolic diseases. The gastrointestinal barriers to oral drug delivery and strategies to overcome these barriers are also explored. We then discuss strategies for alleviating metabolic defects, including glucose metabolism, lipid metabolism, and energy metabolism, with oral biomacromolecules such as insulin, glucagon-like peptide-1 receptor agonists, proprotein convertase subtilisin/kexin type 9 inhibitors, fibroblast growth factor 21 analogues, and peptide YY analogues.
Collapse
Affiliation(s)
- Qiuxia Jiao
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuan Huang
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Jinhan He
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Yining Xu
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
4
|
Wang X, Yang Z, Zhang W, Xing L, Luo R, Cao S. Obstacles, research progress, and prospects of oral delivery of bioactive peptides: a comprehensive review. Front Nutr 2024; 11:1496706. [PMID: 39610876 PMCID: PMC11602335 DOI: 10.3389/fnut.2024.1496706] [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: 09/15/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
Bioactive peptides hold significant potential for enhancing human health, however, their limited oral bioavailability poses a substantial barrier to their widespread use in the food and pharmaceutical industries. This article reviews the key factors influencing the absorption efficiency of oral bioactive peptides, including issues related to bitter taste perception, challenges in gastrointestinal environmental stability, and limitations in transmembrane transport. Furthermore, it highlights the latest technologies, such as osmotic technology, chemical modification, and advanced delivery systems, and discusses their advantages in enhancing the stability of bioactive peptides and facilitating intestinal absorption. In addition, the application and challenges of common delivery systems such as liposomes, emulsions, polymer nanoparticles, and hydrogels in oral bioactive peptide delivery are also discussed. This paper aims to provide a theoretical foundation for scientific research and practical applications of oral delivery of bioactive peptides, thereby promoting the further development of bioactive peptides in the context of human health.
Collapse
Affiliation(s)
- Xinyu Wang
- School of Food Science and Engineering, Ningxia University, Yinchuan, China
| | - Zeyao Yang
- School of Food Science and Engineering, Ningxia University, Yinchuan, China
| | - Wangang Zhang
- Key Lab of Meat Processing and Quality Control, MOE, School of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Lujuan Xing
- Key Lab of Meat Processing and Quality Control, MOE, School of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Ruiming Luo
- School of Food Science and Engineering, Ningxia University, Yinchuan, China
| | - Songmin Cao
- School of Food Science and Engineering, Ningxia University, Yinchuan, China
| |
Collapse
|
5
|
Ke Z, Ma Q, Ye X, Wang Y, Jin Y, Zhao X, Su Z. Peptide GLP-1 receptor agonists: From injection to oral delivery strategies. Biochem Pharmacol 2024; 229:116471. [PMID: 39127152 DOI: 10.1016/j.bcp.2024.116471] [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: 04/19/2024] [Revised: 07/20/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Peptide glucagon-like peptide-1 receptor agonists (GLP-1RAs) are effective drugs for treating type 2 diabetes (T2DM) and have been proven to benefit the heart and kidney. Apart from oral semaglutide, which does not require injection, other peptide GLP-1RAs need to be subcutaneously administered. However, oral semaglutide also faces significant challenges, such as low bioavailability and frequent gastrointestinal discomfort. Thus, it is imperative that advanced oral strategies for peptide GLP-1RAs need to be explored. This review mainly compares the current advantages and disadvantages of various oral delivery strategies for peptide GLP-1RAs in the developmental stage and discusses the latest research progress of peptide GLP-1RAs, providing a useful guide for the development of new oral peptide GLP-1RA drugs.
Collapse
Affiliation(s)
- Zhiqiang Ke
- Protein Engineering and Biopharmaceuticals Science, Hubei University of Technology, Wuhan 430068, China; Hubei Key Laboratory of Diabetes and Angiopathy, National Demonstration Center for Experimental General Medicine Education, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, China
| | - Qianqian Ma
- Protein Engineering and Biopharmaceuticals Science, Hubei University of Technology, Wuhan 430068, China; School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China
| | - Xiaonan Ye
- Protein Engineering and Biopharmaceuticals Science, Hubei University of Technology, Wuhan 430068, China
| | - Yanlin Wang
- Protein Engineering and Biopharmaceuticals Science, Hubei University of Technology, Wuhan 430068, China
| | - Yan Jin
- Protein Engineering and Biopharmaceuticals Science, Hubei University of Technology, Wuhan 430068, China
| | - Xinyuan Zhao
- Hubei Key Laboratory of Diabetes and Angiopathy, National Demonstration Center for Experimental General Medicine Education, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei 437100, China.
| | - Zhengding Su
- Protein Engineering and Biopharmaceuticals Science, Hubei University of Technology, Wuhan 430068, China; School of Pharmaceutical Sciences and Institute of Materia Medica, Xinjiang University, Urumqi 830017, China.
| |
Collapse
|
6
|
Parida P, Prusty AK, Patro SK, Jena BR. Current Advancements on Oral Protein and Peptide Drug Delivery Approaches to Bioavailability: Extensive Review on Patents. RECENT ADVANCES IN DRUG DELIVERY AND FORMULATION 2024; 18:227-246. [PMID: 39356096 DOI: 10.2174/0126673878299775240719061653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 05/09/2024] [Accepted: 06/06/2024] [Indexed: 10/03/2024]
Abstract
Protein and peptide-based drugs have greater therapeutic efficacy and potential application and lower toxicity compared to chemical entities in long-term use within optimum concentration as they are easily biodegradable due to biological origin. While oral administration is preferable, most of these substances are currently administered intravenously or subcutaneously. This is primarily due to the breakdown and poor absorption in the GI tract. Hence, ongoing research is focused on investigating absorption enhancers, enzyme inhibitors, carrier systems, and stability enhancers as potential strategies to facilitate the oral administration of proteins and peptides. Investigations have been directed towards advancing novel technologies to address gastrointestinal (GI) barriers associated with protein and peptide medications. The current review intensifies formulation and stability approaches for oral protein & peptide drug delivery systems with all significant parameters intended for patient safety. Notably, certain innovative technologies have been patented and are currently undergoing clinical trials or have already been introduced into the market. All the approaches stated for the administration of protein and peptide drugs are critically discussed, having their current status, future directions, and recent patents published in the last decades.
Collapse
Affiliation(s)
- Prasanna Parida
- Department of Pharmacy, Biju Patnaik University of Technology, Rourkela, Odisha, India
- School of Pharmacy & Life Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| | - Amiya Kumar Prusty
- Faculty of Pharmacy, C.V Raman Global University, Bhubaneswar, Odisha, India
| | - Saroj Kumar Patro
- Institute of Pharmacy and Technology, Salipur, Cuttack (Affiliated to Biju Patnaik University of Technology, Rourkela), Odisha, India
| | - Bikash Ranjan Jena
- School of Pharmacy & Life Sciences, Centurion University of Technology and Management, Bhubaneswar, Odisha, India
| |
Collapse
|
7
|
Asano D, Takakusa H, Nakai D. Oral Absorption of Middle-to-Large Molecules and Its Improvement, with a Focus on New Modality Drugs. Pharmaceutics 2023; 16:47. [PMID: 38258058 PMCID: PMC10820198 DOI: 10.3390/pharmaceutics16010047] [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: 10/31/2023] [Revised: 12/11/2023] [Accepted: 12/26/2023] [Indexed: 01/24/2024] Open
Abstract
To meet unmet medical needs, middle-to-large molecules, including peptides and oligonucleotides, have emerged as new therapeutic modalities. Owing to their middle-to-large molecular sizes, middle-to-large molecules are not suitable for oral absorption, but there are high expectations around orally bioavailable macromolecular drugs, since oral administration is the most convenient dosing route. Therefore, extensive efforts have been made to create bioavailable middle-to-large molecules or develop absorption enhancement technology, from which some successes have recently been reported. For example, Rybelsus® tablets and Mycapssa® capsules, both of which contain absorption enhancers, were approved as oral medications for type 2 diabetes and acromegaly, respectively. The oral administration of Rybelsus and Mycapssa exposes their pharmacologically active peptides with molecular weights greater than 1000, namely, semaglutide and octreotide, respectively, into systemic circulation. Although these two medications represent major achievements in the development of orally absorbable peptide formulations, the oral bioavailability of peptides after taking Rybelsus and Mycapssa is still only around 1%. In this article, we review the approaches and recent advances of orally bioavailable middle-to-large molecules and discuss challenges for improving their oral absorption.
Collapse
Affiliation(s)
- Daigo Asano
- Drug Metabolism and Pharmacokinetics Research Laboratories, Daiichi Sankyo Co., Ltd., 1-2-58, Hiromachi, Shinagawa-ku, Tokyo 140-8710, Japan; (H.T.); (D.N.)
| | | | | |
Collapse
|
8
|
Pamshong SR, Bhatane D, Sarnaik S, Alexander A. Mesoporous silica nanoparticles: An emerging approach in overcoming the challenges with oral delivery of proteins and peptides. Colloids Surf B Biointerfaces 2023; 232:113613. [PMID: 37913702 DOI: 10.1016/j.colsurfb.2023.113613] [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: 06/15/2023] [Revised: 09/21/2023] [Accepted: 10/22/2023] [Indexed: 11/03/2023]
Abstract
Proteins and peptides (PPs), as therapeutics are widely explored in the past few decades, by virtue of their inherent advantages like high specificity and biocompatibility with minimal side effects. However, owing to their macromolecular size, poor membrane permeability, and high enzymatic susceptibility, the effective delivery of PPs is often challenging. Moreover, their subjection to varying environmental conditions, when administered orally, results in PPs denaturation and structural conformation, thereby lowering their bioavailability. Hence, for effective delivery with enhanced bioavailability, protection of PPs using nanoparticle-based delivery system has gained a growing interest. Mesoporous silica nanoparticles (MSNs), with their tailored morphology and pore size, high surface area, easy surface modification, versatile loading capacity, excellent thermal stability, and good biocompatibility, are eligible candidates for the effective delivery of macromolecules to the target site. This review highlights the different barriers hindering the oral absorption of PPs and the various strategies available to overcome them. In addition, the potential benefits of MSNs, along with their diversifying role in controlling the loading of PPs and their release under the influence of specific stimuli, are also discussed in length. Further, the tuning of MSNs for enhanced gene transfection efficacy is also highlighted. Since extensive research is ongoing in this area, this review is concluded with an emphasis on the potential risks of MSNs that need to be addressed prior to their clinical translation.
Collapse
Affiliation(s)
- Sharon Rose Pamshong
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781101, India
| | - Dhananjay Bhatane
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781101, India
| | - Santosh Sarnaik
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781101, India
| | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781101, India.
| |
Collapse
|
9
|
Salehi T, Raeisi Estabragh MA, Salarpour S, Ohadi M, Dehghannoudeh G. Absorption enhancer approach for protein delivery by various routes of administration: a rapid review. J Drug Target 2023; 31:950-961. [PMID: 37842966 DOI: 10.1080/1061186x.2023.2271680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
As bioactive molecules, peptides and proteins are essential in living organisms, including animals and humans. Defects in their function lead to various diseases in humans. Therefore, the use of proteins in treating multiple diseases, such as cancers and hepatitis, is increasing. There are different routes to administer proteins, which have limitations due to their large and hydrophilic structure. Another limitation is the presence of biological and lipophilic membranes that do not allow proteins to pass quickly. There are different strategies to increase the absorption of proteins from these biological membranes. One of these strategies is to use compounds as absorption enhancers. Absorption enhancers are compounds such as surfactants, phospholipids and cyclodextrins that increase protein passage through the biological membrane and their absorption by different mechanisms. This review focuses on using other absorption enhancers and their mechanism in protein administration routes.
Collapse
Affiliation(s)
- Toktam Salehi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Amin Raeisi Estabragh
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Soodeh Salarpour
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mandana Ohadi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Dehghannoudeh
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
10
|
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
|
11
|
Liu S, Wen X, Zhang X, Mao S. Oral delivery of biomacromolecules by overcoming biological barriers in the gastrointestinal tract: an update. Expert Opin Drug Deliv 2023; 20:1333-1347. [PMID: 37439101 DOI: 10.1080/17425247.2023.2231343] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 06/27/2023] [Indexed: 07/14/2023]
Abstract
INTRODUCTION Biomacromolecules have proven to be an attractive choice for treating diseases due to their properties of strong specificity, high efficiency, and low toxicity. Besides greatly improving the patient's complaint, oral delivery of macromolecules also complies with hormone physiological secretion, which has become one of the most innovative fields of research in recent years. AREAS COVERED Oral delivery biological barriers for biomacromolecule, transport mechanisms, and various administration strategies were discussed in this review, including absorption enhancers, targeting nanoparticles, mucoadhesion nanoparticles, mucus penetration nanoparticles, and intelligent bionic drug delivery systems. EXPERT OPINION The oral delivery of biomacromolecules has important clinical implications; however, these are still facing the challenges of low bioavailability due to certain barriers. Various promising technologies have been developed to overcome the barriers and improve the therapeutic effect of oral biomacromolecules. By considering safety and efficacy comprehensively, the development of intelligent nanoparticles based on the GIT environment has demonstrated some promise in overcoming these barriers; however, a more comprehensive understanding of the oral fate of oral biomacromolecules is still required.
Collapse
Affiliation(s)
- Shiyun Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiangce Wen
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xin Zhang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Shirui Mao
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
12
|
Spoorthi Shetty S, Halagali P, Johnson AP, Spandana KMA, Gangadharappa HV. Oral insulin delivery: Barriers, strategies, and formulation approaches: A comprehensive review. Int J Biol Macromol 2023:125114. [PMID: 37263330 DOI: 10.1016/j.ijbiomac.2023.125114] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/23/2023] [Accepted: 05/24/2023] [Indexed: 06/03/2023]
Abstract
Diabetes Mellitus is characterized by a hyperglycemic condition which can either be caused by the destruction of the beta cells or by the resistance developed against insulin in the cells. Insulin is a peptide hormone that regulates the metabolism of carbohydrates, proteins, and fats. Type 1 Diabetes Mellitus needs the use of Insulin for efficient management. However invasive methods of administration may lead to reduced adherence by the patients. Hence there is a need for a non-invasive method of administration. Oral Insulin has several merits over the conventional method including patient compliance, and reduced cost, and it also mimics endogenous insulin and hence reaches the liver by the portal vein at a higher concentration and thereby showing improved efficiency. However oral Insulin must pass through several barriers in the gastrointestinal tract. Some strategies that could be utilized to bypass these barriers include the use of permeation enhancers, absorption enhancers, use of suitable polymers, use of suitable carriers, and other agents. Several formulation types have been explored for the oral delivery of Insulin like hydrogels, capsules, tablets, and patches which have been described briefly by the article. A lot of attempts have been made for developing oral insulin delivery however none of them have been commercialized due to numerous shortcomings. Currently, there are several formulations from the companies that are still in the clinical phase, the success or failure of some is yet to be seen in the future.
Collapse
Affiliation(s)
- S Spoorthi Shetty
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Praveen Halagali
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Asha P Johnson
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - K M Asha Spandana
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - H V Gangadharappa
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India.
| |
Collapse
|
13
|
Masloh S, Culot M, Gosselet F, Chevrel A, Scapozza L, Zeisser Labouebe M. Challenges and Opportunities in the Oral Delivery of Recombinant Biologics. Pharmaceutics 2023; 15:pharmaceutics15051415. [PMID: 37242657 DOI: 10.3390/pharmaceutics15051415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 04/22/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Recombinant biological molecules are at the cutting-edge of biomedical research thanks to the significant progress made in biotechnology and a better understanding of subcellular processes implicated in several diseases. Given their ability to induce a potent response, these molecules are becoming the drugs of choice for multiple pathologies. However, unlike conventional drugs which are mostly ingested, the majority of biologics are currently administered parenterally. Therefore, to improve their limited bioavailability when delivered orally, the scientific community has devoted tremendous efforts to develop accurate cell- and tissue-based models that allow for the determination of their capacity to cross the intestinal mucosa. Furthermore, several promising approaches have been imagined to enhance the intestinal permeability and stability of recombinant biological molecules. This review summarizes the main physiological barriers to the oral delivery of biologics. Several preclinical in vitro and ex vivo models currently used to assess permeability are also presented. Finally, the multiple strategies explored to address the challenges of administering biotherapeutics orally are described.
Collapse
Affiliation(s)
- Solene Masloh
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Maxime Culot
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
| | - Fabien Gosselet
- Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, University of Artois, UR 2465, Rue Jean Souvraz, 62300 Lens, France
| | - Anne Chevrel
- Affilogic, 24 Rue de la Rainière, 44300 Nantes, France
| | - Leonardo Scapozza
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| | - Magali Zeisser Labouebe
- School of Pharmaceutical Sciences, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1 Rue Michel Servet, 1201 Geneva, Switzerland
| |
Collapse
|
14
|
Fein KC, Gleeson JP, Cochran K, Lamson NG, Doerfler R, Melamed JR, Whitehead KA. Long-term daily oral administration of intestinal permeation enhancers is safe and effective in mice. Bioeng Transl Med 2023; 8:e10342. [PMID: 36684095 PMCID: PMC9842030 DOI: 10.1002/btm2.10342] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/25/2022] [Accepted: 04/29/2022] [Indexed: 01/25/2023] Open
Abstract
Although protein drugs are powerful biologic therapeutics, they cannot be delivered orally because their large size and hydrophilicity limit their absorption across the intestinal epithelium. One potential solution is the incorporation of permeation enhancers into oral protein formulations; however, few have advanced clinically due to toxicity concerns surrounding chronic use. To better understand these concerns, we conducted a 30-day longitudinal study of daily oral permeation enhancer use in mice and resultant effects on intestinal health. Specifically, we investigated three permeation enhancers: sodium caprate (C10), an industry standard, as well as 1-phenylpiperazine (PPZ) and sodium deoxycholate (SDC). Over 30 days of treatment, all mice gained weight, and none required removal from the study due to poor health. Furthermore, intestinal permeability did not increase following chronic use. We also quantified the gene expression of four tight junction proteins (claudin 2, claudin 3, ZO-1, and JAM-A). Significant differences in gene expression between untreated and permeation enhancer-treated mice were found, but these varied between treatment groups, with most differences resolving after a 1-week washout period. Immunofluorescence microscopy revealed no observable differences in protein localization or villus architecture between treated and untreated mice. Overall, PPZ and SDC performed comparably to C10, one of the most clinically advanced enhancers, and results suggest that the chronic use of some permeation enhancers may be therapeutically viable from a safety standpoint.
Collapse
Affiliation(s)
- Katherine C. Fein
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - John P. Gleeson
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Kyle Cochran
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Nicholas G. Lamson
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Rose Doerfler
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Jilian R. Melamed
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Kathryn A. Whitehead
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
- Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| |
Collapse
|
15
|
Core-shell nanosystems designed for effective oral delivery of polypeptide drugs. J Control Release 2022; 352:540-555. [PMID: 36323363 DOI: 10.1016/j.jconrel.2022.10.031] [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: 07/27/2022] [Revised: 10/11/2022] [Accepted: 10/18/2022] [Indexed: 11/06/2022]
Abstract
The stomach acid degradation, mucus clearance and intestinal epithelial impermeability severely limit the oral delivery of polypeptide drugs. To simultaneously address the three major barriers, novel self-assembled core-shell nanosystems (CA-NPs) were designed. The fabricated shell of citric acid cross-linked carboxymethyl cellulose (CA-CMC) wrapped on core nanoparticles (HA-NPs) maintained the integrity of CA-NPs in the stomach. When CA-NPs passed through the stomach, the CA-CMC shell was gradually degraded to release the core HA-NPs in the intestine. HA-NPs with numerous hydrophilic groups and mannose side chains rapidly penetrated through the mucus layer and efficiently transcellular transported via the glucose transporter (GLUT)-mediated and paracellular transport through reversible opening of tight junctions (TJs) by CA-CMC. The oral bioavailability and therapeutic effects of CA-NPs-loaded polypeptide colistin against Escherichia coli (E. coli) bacteremia in mice were significantly increased compared with the native colistin, respectively. Good safety was observed following oral daily delivery for 14 consecutive days. Thus, CA-NPs may offer a promising strategy for the oral delivery of polypeptide drugs.
Collapse
|
16
|
Considerations in the developability of peptides for oral administration when formulated together with transient permeation enhancers. Int J Pharm 2022; 628:122238. [PMID: 36174850 DOI: 10.1016/j.ijpharm.2022.122238] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/10/2022] [Accepted: 09/22/2022] [Indexed: 11/22/2022]
Abstract
This paper reviews many of the properties of a peptide that need to be considered prior to development as an oral dosage form when co-formulated with a permeation enhancer to improve oral bioavailability, including the importance and implications of peptide half-life on variability in pharmacokinetic profiles. Clinical considerations in terms of food and drug-drug interactions are also discussed. The paper further gives a brief overview how permeation enhancers overcome barriers that limit oral absorption of peptides and thereby improve their oral bioavailability, albeit bioavailabilities are still low single digit and variability is high.
Collapse
|
17
|
Tran H, Patel PJ, Aburub A, Sperry A, Estwick S, ElSayed MEH, -Mannan AD. Identification of a Multi-Component Formulation for Intestinal Delivery of a GLP-1/Glucagon Co-agonist Peptide. Pharm Res 2022; 39:2555-2567. [PMID: 36050547 DOI: 10.1007/s11095-022-03372-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/14/2022] [Indexed: 10/14/2022]
Abstract
PURPOSE Oral delivery of therapeutic peptides has been challenging due to multiple physiological factors and physicochemical properties of peptides. We report a systematic approach to identify formulation compositions combining a permeation enhancer and a peptidase inhibitor that minimize proteolytic degradation and increase absorption of a peptide across the small intestine. METHODS An acylated glucagon-like peptide-1/glucagon co-agonist peptide (4.5 kDa) was selected as a model peptide. Proteolytic stability of the peptide was investigated in rat and pig SIF. Effective PEs and multiple component formulations were identified in rats. Relative bioavailability of the peptide was determined in minipigs via intraduodenal administration (ID) of enteric capsules. RESULTS The peptide degraded rapidly in the rat and pig SIF. Citric acid, SBTI, and SBTCI inhibited the enzymatic degradation. The peptide self-associated into trimers in solution, however, addition of PEs monomerized the peptide. C10 was the most effective PE among tested PEs (DPC, LC, rhamnolipid, C12-maltosides, and SNAC) to improve intestinal absorption of the peptide in the rat IJ-closed loop model. A combination of C10 and SBTI or SBTCI increased the peptide exposure 5-tenfold compared to the exposure with the PE alone in the rat IJ-cannulated model, and achieved 1.06 ± 0.76% bioavailability in minipigs relative to subcutaneous via ID administration using enteric capsules. CONCLUSION We identified SBTI and C10 as an effective peptidase inhibitor and PE for intestinal absorption of the peptide. The combination of SBTI and C10 addressed the peptide physiochemical properties and provides a formulation strategy to achieve intestinal delivery of this peptide.
Collapse
Affiliation(s)
- Huyen Tran
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA.
| | - Phenil J Patel
- Synthetic Molecule Design and Development, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Aktham Aburub
- Synthetic Molecule Design and Development, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Andrea Sperry
- Department of Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Selina Estwick
- Department of Drug Disposition, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Mohamed E H ElSayed
- Biotechnology Discovery Research, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Amita Datta -Mannan
- Exploratory Medicine and Pharmacology, Lilly Research Laboratories, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA.
| |
Collapse
|
18
|
Lambros M, Tran T(H, Fei Q, Nicolaou M. Citric Acid: A Multifunctional Pharmaceutical Excipient. Pharmaceutics 2022; 14:972. [PMID: 35631557 PMCID: PMC9148065 DOI: 10.3390/pharmaceutics14050972] [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: 02/11/2022] [Revised: 04/15/2022] [Accepted: 04/23/2022] [Indexed: 02/04/2023] Open
Abstract
Citric acid, a tricarboxylic acid, has found wide application in the chemical and pharmaceutical industry due to its biocompatibility, versatility, and green, environmentally friendly chemistry. This review emphasizes the pharmaceutical uses of citric acid as a strategic ingredient in drug formulation while focusing on the impact of its physicochemical properties. The functionality of citric acid is due to its three carboxylic groups and one hydroxyl group. These allow it to be used in many ways, including its ability to be used as a crosslinker to form biodegradable polymers and as a co-former in co-amorphous and co-crystal applications. This paper also analyzes the effect of citric acid in physiological processes and how this effect can be used to enhance the attributes of pharmaceutical preparations, as well as providing a critical discussion on the issues that may arise out of the presence of citric acid in formulations.
Collapse
Affiliation(s)
- Maria Lambros
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, 309 E Second Street, Pomona, CA 91766, USA; (T.T.); (Q.F.)
| | - Thac (Henry) Tran
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, 309 E Second Street, Pomona, CA 91766, USA; (T.T.); (Q.F.)
| | - Qinqin Fei
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, 309 E Second Street, Pomona, CA 91766, USA; (T.T.); (Q.F.)
| | - Mike Nicolaou
- Doric Pharma LLC, 5270 California Ave, Suite 300, Irvine, CA 92617, USA;
| |
Collapse
|
19
|
Karole A, Parvez S, Thakur RS, Mudavath SL. Effervescent based nano-gas carrier enhanced the bioavailability of poorly aqueous soluble drug: A comprehensive mechanistic understanding. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
20
|
Awad A, Madla CM, McCoubrey LE, Ferraro F, Gavins FK, Buanz A, Gaisford S, Orlu M, Siepmann F, Siepmann J, Basit AW. Clinical translation of advanced colonic drug delivery technologies. Adv Drug Deliv Rev 2022; 181:114076. [PMID: 34890739 DOI: 10.1016/j.addr.2021.114076] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 10/26/2021] [Accepted: 12/02/2021] [Indexed: 12/12/2022]
Abstract
Targeted drug delivery to the colon offers a myriad of benefits, including treatment of local diseases, direct access to unique therapeutic targets and the potential for increasing systemic drug bioavailability and efficacy. Although a range of traditional colonic delivery technologies are available, these systems exhibit inconsistent drug release due to physiological variability between and within individuals, which may be further exacerbated by underlying disease states. In recent years, significant translational and commercial advances have been made with the introduction of new technologies that incorporate independent multi-stimuli release mechanisms (pH and/or microbiota-dependent release). Harnessing these advanced technologies offers new possibilities for drug delivery via the colon, including the delivery of biopharmaceuticals, vaccines, nutrients, and microbiome therapeutics for the treatment of both local and systemic diseases. This review details the latest advances in colonic drug delivery, with an emphasis on emerging therapeutic opportunities and clinical technology translation.
Collapse
|
21
|
Shin TH, Manavalan B, Lee DY, Basith S, Seo C, Paik MJ, Kim SW, Seo H, Lee JY, Kim JY, Kim AY, Chung JM, Baik EJ, Kang SH, Choi DK, Kang Y, Maral Mouradian M, Lee G. Silica-coated magnetic-nanoparticle-induced cytotoxicity is reduced in microglia by glutathione and citrate identified using integrated omics. Part Fibre Toxicol 2021; 18:42. [PMID: 34819099 PMCID: PMC8614058 DOI: 10.1186/s12989-021-00433-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 10/25/2021] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Nanoparticles have been utilized in brain research and therapeutics, including imaging, diagnosis, and drug delivery, owing to their versatile properties compared to bulk materials. However, exposure to nanoparticles leads to their accumulation in the brain, but drug development to counteract this nanotoxicity remains challenging. To date, concerns have risen about the potential toxicity to the brain associated with nanoparticles exposure via penetration of the brain blood barrier to address this issue. METHODS Here the effect of silica-coated-magnetic nanoparticles containing the rhodamine B isothiocyanate dye [MNPs@SiO2(RITC)] were assessed on microglia through toxicological investigation, including biological analysis and integration of transcriptomics, proteomics, and metabolomics. MNPs@SiO2(RITC)-induced biological changes, such as morphology, generation of reactive oxygen species, intracellular accumulation of MNPs@SiO2(RITC) using transmission electron microscopy, and glucose uptake efficiency, were analyzed in BV2 murine microglial cells. Each omics data was collected via RNA-sequencing-based transcriptome analysis, liquid chromatography-tandem mass spectrometry-based proteome analysis, and gas chromatography- tandem mass spectrometry-based metabolome analysis. The three omics datasets were integrated and generated as a single network using a machine learning algorithm. Nineteen compounds were screened and predicted their effects on nanotoxicity within the triple-omics network. RESULTS Intracellular reactive oxygen species production, an inflammatory response, and morphological activation of cells were greater, but glucose uptake was lower in MNPs@SiO2(RITC)-treated BV2 microglia and primary rat microglia in a dose-dependent manner. Expression of 121 genes (from 41,214 identified genes), and levels of 45 proteins (from 5918 identified proteins) and 17 metabolites (from 47 identified metabolites) related to the above phenomena changed in MNPs@SiO2(RITC)-treated microglia. A combination of glutathione and citrate attenuated nanotoxicity induced by MNPs@SiO2(RITC) and ten other nanoparticles in vitro and in the murine brain, protecting mostly the hippocampus and thalamus. CONCLUSIONS Combination of glutathione and citrate can be one of the candidates for nanotoxicity alleviating drug against MNPs@SiO2(RITC) induced detrimental effect, including elevation of intracellular reactive oxygen species level, activation of microglia, and reduction in glucose uptake efficiency. In addition, our findings indicate that an integrated triple omics approach provides useful and sensitive toxicological assessment for nanoparticles and screening of drug for nanotoxicity.
Collapse
Affiliation(s)
- Tae Hwan Shin
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Balachandran Manavalan
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Da Yeon Lee
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Shaherin Basith
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Chan Seo
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon, 57922 Republic of Korea
| | - Man Jeong Paik
- College of Pharmacy, Sunchon National University, 255 Jungang-ro, Suncheon, 57922 Republic of Korea
| | - Sang-Wook Kim
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Haewoon Seo
- Department of Molecular Science and Technology, Ajou University, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Ju Yeon Lee
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanji-ro, Cheongju, 28119 Republic of Korea
| | - Jin Young Kim
- Research Center of Bioconvergence Analysis, Korea Basic Science Institute, 162 Yeongudanji-ro, Cheongju, 28119 Republic of Korea
| | - A Young Kim
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Jee Min Chung
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Eun Joo Baik
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - Seong Ho Kang
- Department of Chemistry, Graduate School, Kyung Hee University, Yongin-si, Gyeonggi-do 17104 Republic of Korea
- Department of Applied Chemistry and Institute of Natural Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104 Republic of Korea
| | - Dong-Kug Choi
- Department of Biotechnology, College of Biomedical and Health Science, Konkuk University, 268 Chungwondaero, Chungju, 27478 Republic of Korea
| | - Yup Kang
- Department of Physiology, Ajou University School of Medicine, 206 World cup-ro, Suwon, 16499 Republic of Korea
| | - M. Maral Mouradian
- RWJMS Institute for Neurological Therapeutics, Rutgers Biomedical and Health Sciences, and Department of Neurology, Robert Wood Johnson Medical School, Rutgers University, Piscataway, NJ 08854 USA
| | - Gwang Lee
- Department of Molecular Science and Technology, Ajou University, Suwon-si, Gyeonggi-do 16499 Republic of Korea
- Department of Physiology, Ajou University School of Medicine, Suwon-si, Gyeonggi-do 16499 Republic of Korea
| |
Collapse
|
22
|
Burshtein G, Itin C, Tang JCY, Galitzer H, Fraser WD, Schwartz P. The combined effect of permeation enhancement and proteolysis inhibition on the systemic exposure of orally administrated peptides: Salcaprozate sodium, soybean trypsin inhibitor, and teriparatide study in pigs. INTERNATIONAL JOURNAL OF PHARMACEUTICS-X 2021; 3:100097. [PMID: 34704013 PMCID: PMC8524144 DOI: 10.1016/j.ijpx.2021.100097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/30/2021] [Accepted: 10/01/2021] [Indexed: 11/21/2022]
Abstract
Oral delivery of peptides and proteins is hindered by their rapid proteolysis in the gastrointestinal tract and their inability to permeate biological membranes. Various drug delivery approaches are being investigated and implemented to overcome these obstacles. In the discussed study conducted in pigs, an investigation was undertaken to assess the effect of combination of a permeation enhancer – salcaprozate sodium, and a proteolysis inhibitor – soybean trypsin inhibitor, on the systemic exposure of the peptide teriparatide, following intraduodenal administration. Results demonstrate that this combination achieves significantly higher Cmax and AUC (~10- and ~20-fold respectively) compared to each of these methodologies on their own. It was thus concluded that an appropriate combination of different technological approaches may considerably contribute to an efficient oral delivery of biological macromolecules. Soybean trypsin inhibitor (SBTI) protects hPTH(1–34) from proteolysis in the intestine. SNAC/SBTI combination significantly raises plasma exposure of oral hPTH(1–34). Oral formulation hPTH(1–34)/SNAC/SBTI befits the PK profile for osteoporosis treatment. Endoscopic intraduodenal delivery in pigs enables investigation of absorption mechanisms.
Collapse
Affiliation(s)
| | - Constantin Itin
- Entera Bio Ltd., Jerusalem BioPark, Jerusalem 9112002, Israel
| | - Jonathan C Y Tang
- Bioanalytical Facility, Biomedical Research Centre, Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Hillel Galitzer
- Entera Bio Ltd., Jerusalem BioPark, Jerusalem 9112002, Israel
| | - William D Fraser
- Bioanalytical Facility, Biomedical Research Centre, Norwich Medical School, Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK.,Departments of Endocrinology and Clinical Biochemistry, Norfolk and Norwich University Hospital, Norwich, UK
| | | |
Collapse
|
23
|
Mattar G, Haddarah A, Haddad J, Pujola M, Sepulcre F. New approaches, bioavailability and the use of chelates as a promising method for food fortification. Food Chem 2021; 373:131394. [PMID: 34710689 DOI: 10.1016/j.foodchem.2021.131394] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 01/24/2023]
Abstract
Food fortification has been used for many years to combat micronutrient deficiencies; the main challenge with food fortification is the combination of a bioavailable, affordable fortificant with the best (food) vehicle as a carrier to reach at-risk populations. This paper considers mineral deficiencies, especially iron, food fortification, target populations, and the use of chelates in food fortification, as well as different types of mineral-chelate complexes, advantages and limitations of previous trials, methods used for analysis of these complexes, bioavailability of minerals, factors influencing it, and methods particularly those in vitro for predicting outcomes. Three innovative methods (encapsulation, nanoparticulation, and chelation) were explored, which aim to overcome problems associated with conventional fortification, especially those affecting organoleptic properties and bioavailability; but often lead to the emergence of new limitations (for example instability, impracticality and high costs) requiring further research.
Collapse
Affiliation(s)
- Ghadeer Mattar
- Departament d'Enginyeria Agroalimentària i Biotecnologia, Universitat Politècnica de Catalunya, Campus del Baix Llobregat, Carrer Esteve Terradas 8, 08860, Castelldefels, Barcelona, Spain; octoral School of Sciences and Technology, Lebanese University, Rafic Hariri Campus, Hadath, Lebanon
| | - Amira Haddarah
- octoral School of Sciences and Technology, Lebanese University, Rafic Hariri Campus, Hadath, Lebanon
| | - Joseph Haddad
- octoral School of Sciences and Technology, Lebanese University, Rafic Hariri Campus, Hadath, Lebanon
| | - Montserrat Pujola
- Departament d'Enginyeria Agroalimentària i Biotecnologia, Universitat Politècnica de Catalunya, Campus del Baix Llobregat, Carrer Esteve Terradas 8, 08860, Castelldefels, Barcelona, Spain
| | - Franscesc Sepulcre
- Departament d'Enginyeria Agroalimentària i Biotecnologia, Universitat Politècnica de Catalunya, Campus del Baix Llobregat, Carrer Esteve Terradas 8, 08860, Castelldefels, Barcelona, Spain.
| |
Collapse
|
24
|
Formulation strategies to improve the efficacy of intestinal permeation enhancers . Adv Drug Deliv Rev 2021; 177:113925. [PMID: 34418495 DOI: 10.1016/j.addr.2021.113925] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/28/2021] [Accepted: 08/09/2021] [Indexed: 02/06/2023]
Abstract
The use of chemical permeation enhancers (PEs) is the most widely tested approach to improve oral absorption of low permeability active agents, as represented by peptides. Several hundred PEs increase intestinal permeability in preclinical bioassays, yet few have progressed to clinical testing and, of those, only incremental increases in oral bioavailability (BA) have been observed. Still, average BA values of ~1% were sufficient for two recent FDA approvals of semaglutide and octreotide oral formulations. PEs are typically screened in static in vitro and ex-vivo models where co-presentation of active agent and PE in high concentrations allows the PE to alter barrier integrity with sufficient contact time to promote flux across the intestinal epithelium. The capacity to maintain high concentrations of co-presented agents at the epithelium is not reached by standard oral dosage forms in the upper GI tract in vivo due to dilution, interference from luminal components, fast intestinal transit, and possible absorption of the PE per se. The PE-based formulations that have been assessed in clinical trials in either immediate-release or enteric-coated solid dosage forms produce low and variable oral BA due to these uncontrollable physiological factors. For PEs to appreciably increase intestinal permeability from oral dosage forms in vivo, strategies must facilitate co-presentation of PE and active agent at the epithelium for a sustained period at the required concentrations. Focusing on peptides as examples of a macromolecule class, we review physiological impediments to optimal luminal presentation, discuss the efficacy of current PE-based oral dosage forms, and suggest strategies that might be used to improve them.
Collapse
|
25
|
The evolution of commercial drug delivery technologies. Nat Biomed Eng 2021; 5:951-967. [PMID: 33795852 DOI: 10.1038/s41551-021-00698-w] [Citation(s) in RCA: 644] [Impact Index Per Article: 161.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023]
Abstract
Drug delivery technologies have enabled the development of many pharmaceutical products that improve patient health by enhancing the delivery of a therapeutic to its target site, minimizing off-target accumulation and facilitating patient compliance. As therapeutic modalities expanded beyond small molecules to include nucleic acids, peptides, proteins and antibodies, drug delivery technologies were adapted to address the challenges that emerged. In this Review Article, we discuss seminal approaches that led to the development of successful therapeutic products involving small molecules and macromolecules, identify three drug delivery paradigms that form the basis of contemporary drug delivery and discuss how they have aided the initial clinical successes of each class of therapeutic. We also outline how the paradigms will contribute to the delivery of live-cell therapies.
Collapse
|
26
|
Zhu Q, Chen Z, Paul PK, Lu Y, Wu W, Qi J. Oral delivery of proteins and peptides: Challenges, status quo and future perspectives. Acta Pharm Sin B 2021; 11:2416-2448. [PMID: 34522593 PMCID: PMC8424290 DOI: 10.1016/j.apsb.2021.04.001] [Citation(s) in RCA: 163] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 01/29/2021] [Accepted: 02/12/2021] [Indexed: 12/24/2022] Open
Abstract
Proteins and peptides (PPs) have gradually become more attractive therapeutic molecules than small molecular drugs due to their high selectivity and efficacy, but fewer side effects. Owing to the poor stability and limited permeability through gastrointestinal (GI) tract and epithelia, the therapeutic PPs are usually administered by parenteral route. Given the big demand for oral administration in clinical use, a variety of researches focused on developing new technologies to overcome GI barriers of PPs, such as enteric coating, enzyme inhibitors, permeation enhancers, nanoparticles, as well as intestinal microdevices. Some new technologies have been developed under clinical trials and even on the market. This review summarizes the history, the physiological barriers and the overcoming approaches, current clinical and preclinical technologies, and future prospects of oral delivery of PPs.
Collapse
Key Words
- ASBT, apical sodium-dependent bile acid transporter
- BSA, bovine serum albumin
- CAGR, compound annual growth
- CD, Crohn's disease
- COPD, chronic obstructive pulmonary disease
- CPP, cell penetrating peptide
- CaP, calcium phosphate
- Clinical
- DCs, dendritic cells
- DDVAP, desmopressin acetate
- DTPA, diethylene triamine pentaacetic acid
- EDTA, ethylene diamine tetraacetic acid
- EPD, empirical phase diagrams
- EPR, electron paramagnetic resonance
- Enzyme inhibitor
- FA, folic acid
- FDA, U.S. Food and Drug Administration
- FcRn, Fc receptor
- GALT, gut-associated lymphoid tissue
- GI, gastrointestinal
- GIPET, gastrointestinal permeation enhancement technology
- GLP-1, glucagon-like peptide 1
- GRAS, generally recognized as safe
- HBsAg, hepatitis B surface antigen
- HPMCP, hydroxypropyl methylcellulose phthalate
- IBD, inflammatory bowel disease
- ILs, ionic liquids
- LBNs, lipid-based nanoparticles
- LMWP, low molecular weight protamine
- MCT-1, monocarborxylate transporter 1
- MSNs, mesoporous silica nanoparticles
- NAC, N-acetyl-l-cysteine
- NLCs, nanostructured lipid carriers
- Oral delivery
- PAA, polyacrylic acid
- PBPK, physiologically based pharmacokinetics
- PCA, principal component analysis
- PCL, polycarprolacton
- PGA, poly-γ-glutamic acid
- PLA, poly(latic acid)
- PLGA, poly(lactic-co-glycolic acid)
- PPs, proteins and peptides
- PVA, poly vinyl alcohol
- Peptides
- Permeation enhancer
- Proteins
- RGD, Arg-Gly-Asp
- RTILs, room temperature ionic liquids
- SAR, structure–activity relationship
- SDC, sodium deoxycholate
- SGC, sodium glycocholate
- SGF, simulated gastric fluids
- SIF, simulated intestinal fluids
- SLNs, solid lipid nanoparticles
- SNAC, sodium N-[8-(2-hydroxybenzoyl)amino]caprylate
- SNEDDS, self-nanoemulsifying drug delivery systems
- STC, sodium taurocholate
- Stability
- TAT, trans-activating transcriptional peptide
- TMC, N-trimethyl chitosan
- Tf, transferrin
- TfR, transferrin receptors
- UC, ulcerative colitis
- UEA1, ulex europaeus agglutinin 1
- VB12, vitamin B12
- WGA, wheat germ agglutinin
- pHPMA, N-(2-hydroxypropyl)methacrylamide
- pI, isoelectric point
- sCT, salmon calcitonin
- sc, subcutaneous
Collapse
Affiliation(s)
- Quangang Zhu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Zhongjian Chen
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
| | - Pijush Kumar Paul
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Department of Pharmacy, Gono Bishwabidyalay (University), Mirzanagar Savar, Dhaka 1344, Bangladesh
| | - Yi Lu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wei Wu
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Jianping Qi
- Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
27
|
Brayden DJ, Maher S. Transient Permeation Enhancer® (TPE®) technology for oral delivery of octreotide: a technological evaluation. Expert Opin Drug Deliv 2021; 18:1501-1512. [PMID: 34128734 DOI: 10.1080/17425247.2021.1942838] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The FDA approval of oral semaglutide for type 2 diabetes (2019) and oral octreotide for acromegaly (2020) is evidence that selected niche peptides can be administered orally if formulated with selected intestinal permeation enhancers. AREAS COVERED We evaluated the oral octreotide formulation, MYCAPSSA® (Chiasma Pharmaceuticals, Needham, MA, USA). An outline of the current standard of care in acromegaly and the benefits of oral octreotide versus depot injections is provided. We discuss the Transient Permeation Enhancer (TPE®) technology used and detail the safety and efficacy data from animal models and clinical trials. EXPERT OPINION TPE® is an oily suspension of octreotide that includes a number of excipients that can transiently alter epithelial barrier integrity by opening of intestinal epithelial tight junctions arising from transcellular perturbation. Phase I studies using 20 mg octreotide capsules yielded a relative oral bioavailability of ~0.7% and primary endpoints were achieved in two Phase III studies. The oral octreotide dose required to achieve these endpoints was over 200 times that of the 0.1 mg immediate-release subcutaneous injection, a reminder of the difficulty in achieving oral absorption of macromolecules. Many acromegaly patients will prefer a convenient twice-daily oral formulation of octreotide compared to monthly depot injections.
Collapse
Affiliation(s)
- David J Brayden
- University College Dublin (UCD) School of Veterinary Medicine, UCD, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biotechnology, UCD, Belfield, Dublin 4, Ireland.,CÚRAM, the SFI Research Centre for Medical Devices, UCD, Belfield, Dublin 4, Ireland
| | - Sam Maher
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, St. Stephen's Green, Dublin 2, Ireland
| |
Collapse
|
28
|
Patient-centric design for peptide delivery: Trends in routes of administration and advancement in drug delivery technologies. MEDICINE IN DRUG DISCOVERY 2021. [DOI: 10.1016/j.medidd.2020.100079] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
29
|
Metry M, Shu Y, Abrahamsson B, Cristofoletti R, Dressman JB, Groot DW, Parr A, Langguth P, Shah VP, Tajiri T, Mehta MU, Polli JE. Biowaiver Monographs for Immediate Release Solid Oral Dosage Forms: Metformin Hydrochloride. J Pharm Sci 2021; 110:1513-1526. [PMID: 33450218 DOI: 10.1016/j.xphs.2021.01.011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 01/11/2023]
Abstract
Data are examined regarding possible waiver of in vivo bioequivalence testing (i.e. biowaiver) for approval of metformin hydrochloride (metformin) immediate-release solid oral dosage forms. Data include metformin's Biopharmaceutics Classification System (BCS) properties, including potential excipient interactions. Metformin is a prototypical transporter-mediated drug and is highly soluble, but only 50% of an orally administered dose is absorbed from the gut. Therefore, metformin is a BCS Class III substance. A BCS-based approval approach for major changes to marketed products and new generics is admissible if test and reference dosage forms have the identical active pharmaceutical ingredient and if in vitro dissolution from both are very rapid (i.e. at least 85% within 15 min at pH 1.2, 4.5, and 6.8). Recent International Council for Harmonisation BCS guidance indicates all excipients for Class III biowaivers are recommended to be qualitatively the same and quantitatively similar (except for preservatives, flavor agents, colorant, or capsule shell or film coating excipients). However, despite metformin being a prototypical transporter-mediated drug, there is no evidence that commonly used excipients impact metformin absorption, such that this restriction on excipients for BCS III drugs merits regulatory relief. Commonly used excipients in usual amounts are not likely to impact metformin absorption.
Collapse
Affiliation(s)
- Melissa Metry
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA
| | - Bertil Abrahamsson
- Oral Product Development, Pharmaceutical Technology & Development, Operations AstraZeneca, Gothenburg, Sweden
| | - Rodrigo Cristofoletti
- Brazilian Health Surveillance Agency (Anvisa), Division of Bioequivalence, Brasilia, Brazil
| | - Jennifer B Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - D W Groot
- RIVM-National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Alan Parr
- Bioceutics LCC, Raleigh-Durham, North Carolina, USA
| | - Peter Langguth
- Department of Pharmaceutical Technology and Biopharmaceutics, Johannes Gutenberg University, Mainz, Germany
| | - Vinod P Shah
- International Pharmaceutical Federation (FIP), The Hague, the Netherlands
| | - Tomokazu Tajiri
- Astellas Pharma Inc, Analytical Research Laboratories, Yaizu, Japan
| | - Mehul U Mehta
- United States Food and Drug Administration, Center for Drug Evaluation and Research, Silver Spring, MD, USA
| | - James E Polli
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
30
|
Song M, Dang L, Wei H. Evaluation of Calcium Binding Capacity of Chelating Agents in Calcium Carbonate Suspension and Effects on Calcium Distribution of Calcium Chelating Agents. Aust J Chem 2021. [DOI: 10.1071/ch20376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In this study, the binding capacity of calcium ions of sodium tripolyphosphate (STPP), tetrasodium pyrophosphate (TSPP), trisodium citrate (TSC), and potassium oxalate (PO) were evaluated, and the calcium distribution in the presence of STPP and TSPP in CaCl2 solutions (50mmolL−1) were investigated. Under conditions simulating industrial toothpaste, the concentration of fluoride in calcium carbonate suspensions (30 g/50 g) was measured by ion chromatography to investigate the effects of chelating agents on calcium ions. Among all the chelating agents, STPP and TSPP have the highest retention rate of fluoride, indicating better calcium binding capacity. Preliminary studies were carried out in CaCl2 solutions to investigate the influence of concentration and pH on the chelating performance of STPP and TSPP. The distribution of free calcium, chelated calcium, and precipitated calcium in CaCl2 solution in the presence of STPP and TSPP were investigated to reveal two different calcium-chelation mechanisms and laws for STPP and TSPP. This work has a positive guiding significance for the stabilisation of calcium and fluoride in toothpaste formula.
Collapse
|
31
|
Drescher S, van Hoogevest P. The Phospholipid Research Center: Current Research in Phospholipids and Their Use in Drug Delivery. Pharmaceutics 2020; 12:pharmaceutics12121235. [PMID: 33353254 PMCID: PMC7766331 DOI: 10.3390/pharmaceutics12121235] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022] Open
Abstract
This review summarizes the research on phospholipids and their use for drug delivery related to the Phospholipid Research Center Heidelberg (PRC). The focus is on projects that have been approved by the PRC since 2017 and are currently still ongoing or have recently been completed. The different projects cover all facets of phospholipid research, from basic to applied research, including the use of phospholipids in different administration forms such as liposomes, mixed micelles, emulsions, and extrudates, up to industrial application-oriented research. These projects also include all routes of administration, namely parenteral, oral, and topical. With this review we would like to highlight possible future research directions, including a short introduction into the world of phospholipids.
Collapse
|
32
|
Maher S, Geoghegan C, Brayden DJ. Intestinal permeation enhancers to improve oral bioavailability of macromolecules: reasons for low efficacy in humans. Expert Opin Drug Deliv 2020; 18:273-300. [PMID: 32937089 DOI: 10.1080/17425247.2021.1825375] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Intestinal permeation enhancers (PEs) are substances that transiently alter the intestinal epithelial barrier to facilitate permeation of macromolecules with low oral bioavailability (BA). While a number of PEs have progressed to clinical testing in conventional formulations with macromolecules, there has been only low single digit increases in oral BA, irrespective of whether the drug met primary or secondary clinical endpoints. AREAS COVERED This article considers the causes of sub-optimal BA of macromolecules from PE dosage forms and suggests approaches that may improve performance in humans. EXPERT OPINION Permeation enhancement is most effective when the PE is co-localized with the macromolecule at the epithelial surface. Conditions in the GI tract impede optimal co-localization. Novel delivery systems that limit dilution and spreading of the PE and macromolecule in the small intestine have attempted to replicate promising enhancement efficacy observed in static drug delivery models.
Collapse
Affiliation(s)
- Sam Maher
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Caroline Geoghegan
- School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - David J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Dublin, Ireland
| |
Collapse
|
33
|
Hubálek F, Refsgaard HHF, Gram-Nielsen S, Madsen P, Nishimura E, Münzel M, Brand CL, Stidsen CE, Claussen CH, Wulff EM, Pridal L, Ribel U, Kildegaard J, Porsgaard T, Johansson E, Steensgaard DB, Hovgaard L, Glendorf T, Hansen BF, Jensen MK, Nielsen PK, Ludvigsen S, Rugh S, Garibay PW, Moore MC, Cherrington AD, Kjeldsen T. Molecular engineering of safe and efficacious oral basal insulin. Nat Commun 2020; 11:3746. [PMID: 32719315 PMCID: PMC7385171 DOI: 10.1038/s41467-020-17487-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 07/01/2020] [Indexed: 12/19/2022] Open
Abstract
Recently, the clinical proof of concept for the first ultra-long oral insulin was reported, showing efficacy and safety similar to subcutaneously administered insulin glargine. Here, we report the molecular engineering as well as biological and pharmacological properties of these insulin analogues. Molecules were designed to have ultra-long pharmacokinetic profile to minimize variability in plasma exposure. Elimination plasma half-life of ~20 h in dogs and ~70 h in man is achieved by a strong albumin binding, and by lowering the insulin receptor affinity 500-fold to slow down receptor mediated clearance. These insulin analogues still stimulate efficient glucose disposal in rats, pigs and dogs during constant intravenous infusion and euglycemic clamp conditions. The albumin binding facilitates initial high plasma exposure with a concomitant delay in distribution to peripheral tissues. This slow appearance in the periphery mediates an early transient hepato-centric insulin action and blunts hypoglycaemia in dogs in response to overdosing.
Collapse
Affiliation(s)
| | | | | | - Peter Madsen
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | - Erica Nishimura
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | - Martin Münzel
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | | | | | | | - Erik Max Wulff
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | - Lone Pridal
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | - Ulla Ribel
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | | | - Trine Porsgaard
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | - Eva Johansson
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | | | - Lars Hovgaard
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | - Tine Glendorf
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | - Bo Falck Hansen
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | | | | | - Svend Ludvigsen
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | - Susanne Rugh
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark
| | | | | | | | - Thomas Kjeldsen
- Novo Nordisk A/S, Novo Nordisk Park 1, 2760, Maaloev, Denmark.
| |
Collapse
|
34
|
Chen KH, Miao YB, Shang CY, Huang TY, Yu YT, Yeh CN, Song HL, Chen CT, Mi FL, Lin KJ, Sung HW. A bubble bursting-mediated oral drug delivery system that enables concurrent delivery of lipophilic and hydrophilic chemotherapeutics for treating pancreatic tumors in rats. Biomaterials 2020; 255:120157. [PMID: 32535305 DOI: 10.1016/j.biomaterials.2020.120157] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 05/27/2020] [Indexed: 12/20/2022]
Abstract
The therapeutic outcome of pancreatic cancer remains unsatisfactory, despite many attempts to improve it. To address this challenge, an oral drug delivery system that spontaneously initiates an effervescent reaction to form gas-bubble carriers is proposed. These carriers concurrently deliver lipophilic paclitaxel (PTX) and hydrophilic gemcitabine (GEM) in the small intestine. The bursting of the bubbles promotes the intestinal absorption of the drugs. The antitumor efficacy of this proposed oral drug delivery system is evaluated in rats with experimentally created orthotopic pancreatic tumors. The combined administration of equivalent amounts of PTX and GEM via the intravenous (i.v.) route, which is clinically used for treating pancreatic cancer, serves as a control. Following oral administration, the lipophilic PTX is initially absorbed through the intestinal lymphatic system and then enters systemic circulation, whereas the hydrophilic GEM is directly taken up into the blood circulation, ultimately accumulating in the tumorous pancreatic tissues. A pharmacokinetic study reveals that the orally delivered formulation has none of the toxic side-effects that are associated with the i.v. injected formulation; changes the pharmacokinetic profiles of the drugs; and increases the bioavailability of PTX. The oral formulation has a greater impact than the i.v. formulation on tumor-specific stromal depletion, resulting in greater inhibition of tumor growth with no evidence of metastatic spread. As well as enhancing the therapeutic efficacy, this unique approach of oral chemotherapy has potential for use on outpatients, greatly improving their quality of life.
Collapse
Affiliation(s)
- Kuan-Hung Chen
- Department of Chemical Engineering/Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Yang-Bao Miao
- Department of Chemical Engineering/Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Chun-Yu Shang
- Department of Chemical Engineering/Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Tring-Yo Huang
- Department of Chemical Engineering/Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Tzu Yu
- Department of Chemical Engineering/Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan
| | - Chun-Nan Yeh
- Department of General Surgery, Liver Research Center, Linkou Chang Gung Memorial Hospital, And Chang Gung University, Taoyuan, Taiwan
| | - Hsiang-Lin Song
- Department of Pathology, National Taiwan University Hospital-Hsinchu Branch, Hsinchu, Taiwan
| | - Chiung-Tong Chen
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institute, Zhunan, Miaoli, Taiwan
| | - Fwu-Long Mi
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kun-Ju Lin
- Department of Nuclear Medicine and Molecular Imaging Center, Linkou Chang Gung Memorial Hospital, And Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan.
| | - Hsing-Wen Sung
- Department of Chemical Engineering/Institute of Biomedical Engineering and Frontier Research Center on Fundamental and Applied Sciences of Matters, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
35
|
Brayden D, Hill T, Fairlie D, Maher S, Mrsny R. Systemic delivery of peptides by the oral route: Formulation and medicinal chemistry approaches. Adv Drug Deliv Rev 2020; 157:2-36. [PMID: 32479930 DOI: 10.1016/j.addr.2020.05.007] [Citation(s) in RCA: 149] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 02/07/2023]
Abstract
In its 33 years, ADDR has published regularly on the po5tential of oral delivery of biologics especially peptides and proteins. In the intervening period, analysis of the preclinical and clinical trial failures of many purported platform technologies has led to reflection on the true status of the field and reigning in of expectations. Oral formulations of semaglutide, octreotide, and salmon calcitonin have completed Phase III trials, with oral semaglutide being approved by the FDA in 2019. The progress made with oral peptide formulations based on traditional permeation enhancers is against a background of low and variable oral bioavailability values of ~1%, leading to a current perception that only potent peptides with a viable cost of synthesis can be realistically considered. Desirable features of candidates should include a large therapeutic index, some stability in the GI tract, a long elimination half-life, and a relatively low clearance rate. Administration in nanoparticle formats have largely disappointed, with few prototypes reaching clinical trials: insufficient particle loading, lack of controlled release, low epithelial particle uptake, and lack of scalable synthesis being the main reasons for discontinuation. Disruptive technologies based on engineered devices promise improvements, but scale-up and toxicology aspects are issues to address. In parallel, medicinal chemists are synthesizing stable hydrophobic macrocyclic candidate peptides of lower molecular weight and with potential for greater oral bioavailability than linear peptides, but perhaps without the same requirement for elaborate drug delivery systems. In summary, while there have been advances in understanding the limitations of peptides for oral delivery, low membrane permeability, metabolism, and high clearance rates continue to hamper progress.
Collapse
|
36
|
Ibrahim YHE, Regdon G, Kristó K, Kelemen A, Adam ME, Hamedelniel EI, Sovány T. Design and characterization of chitosan/citrate films as carrier for oral macromolecule delivery. Eur J Pharm Sci 2020; 146:105270. [DOI: 10.1016/j.ejps.2020.105270] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 01/31/2020] [Accepted: 02/16/2020] [Indexed: 10/25/2022]
|
37
|
McCright JC, Maisel K. Engineering drug delivery systems to overcome mucosal barriers for immunotherapy and vaccination. Tissue Barriers 2019; 8:1695476. [PMID: 31775577 DOI: 10.1080/21688370.2019.1695476] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Mucosal surfaces protect our bodies from pathogens and external irritants using a system of biological barriers. Overcoming these barriers is a significant drug delivery challenge, particularly for immunotherapies that aim to modulate the local immune response. Reaching local lymphoid tissues and draining lymph nodes (LNs) requires crossing the mucus mesh, mucosal epithelium, and either targeting M cells covering lymphoid tissues or utilizing lymphatic transport that shuttles molecules and particulates from the periphery to the LN. We first highlight the barrier properties of mucus and mucosal epithelium, and the function of the mucosal immune system. We then dive into existing drug delivery technologies that have been engineered to overcome each of these barriers. We particularly focus on novel strategies for targeting lymphoid tissues, which has been shown to enhance immunotherapies and vaccinations, via directly targeting LNs, lymphatic vessels, and M cells that transport samples of mucosal content to the lymphoid tissues.
Collapse
Affiliation(s)
- Jacob C McCright
- Department of Bioengineering, University of Maryland College Park, College Park, MD, USA
| | - Katharina Maisel
- Department of Bioengineering, University of Maryland College Park, College Park, MD, USA
| |
Collapse
|
38
|
Microcontainers for oral insulin delivery – In vitro studies of permeation enhancement. Eur J Pharm Biopharm 2019; 143:98-105. [DOI: 10.1016/j.ejpb.2019.08.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 08/13/2019] [Accepted: 08/15/2019] [Indexed: 12/21/2022]
|
39
|
Feng K, Li C, Wei YS, Zong MH, Wu H, Han SY. Development of a polysaccharide based multi-unit nanofiber mat for colon-targeted sustained release of salmon calcitonin. J Colloid Interface Sci 2019; 552:186-195. [DOI: 10.1016/j.jcis.2019.05.037] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/08/2019] [Accepted: 05/10/2019] [Indexed: 01/13/2023]
|
40
|
Buckley ST, Bækdal TA, Vegge A, Maarbjerg SJ, Pyke C, Ahnfelt-Rønne J, Madsen KG, Schéele SG, Alanentalo T, Kirk RK, Pedersen BL, Skyggebjerg RB, Benie AJ, Strauss HM, Wahlund PO, Bjerregaard S, Farkas E, Fekete C, Søndergaard FL, Borregaard J, Hartoft-Nielsen ML, Knudsen LB. Transcellular stomach absorption of a derivatized glucagon-like peptide-1 receptor agonist. Sci Transl Med 2018; 10:10/467/eaar7047. [DOI: 10.1126/scitranslmed.aar7047] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 10/22/2018] [Indexed: 11/02/2022]
Abstract
Oral administration of therapeutic peptides is hindered by poor absorption across the gastrointestinal barrier and extensive degradation by proteolytic enzymes. Here, we investigated the absorption of orally delivered semaglutide, a glucagon-like peptide-1 analog, coformulated with the absorption enhancer sodiumN-[8-(2-hydroxybenzoyl) aminocaprylate] (SNAC) in a tablet. In contrast to intestinal absorption usually seen with small molecules, clinical and preclinical dog studies revealed that absorption of semaglutide takes place in the stomach, is confined to an area in close proximity to the tablet surface, and requires coformulation with SNAC. SNAC protects against enzymatic degradation via local buffering actions and only transiently enhances absorption. The mechanism of absorption is shown to be compound specific, transcellular, and without any evidence of effect on tight junctions. These data have implications for understanding how highly efficacious and specific therapeutic peptides could be transformed from injectable to tablet-based oral therapies.
Collapse
|
41
|
An intestinal paracellular pathway biased toward positively-charged macromolecules. J Control Release 2018; 288:111-125. [PMID: 30194947 DOI: 10.1016/j.jconrel.2018.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/03/2018] [Accepted: 09/03/2018] [Indexed: 12/16/2022]
Abstract
Lacking an effective mechanism to safely and consistently enhance macromolecular uptake across the intestinal epithelium, prospects for successful development of oral therapeutic peptide drugs remain unlikely. We previously addressed this challenge by identifying an endogenous mechanism that controls intestinal paracellular permeability that can be activated by a peptide, termed PIP 640, which can increase cellular levels of phosphorylated myosin light chain at position S19 (MLC-pS19). Apical application in vitro or luminal application in vivo was shown to increase macromolecular solute transport within minutes that recovered completely within a few hours after removal. We now examine the nature of PIP 640-mediated permeability changes. Confluent Caco-2 cell monolayers treated with PIP 640 enhanced apical-to-basolateral (AB) transport of 4-kDa, but not 10-kDa, dextran. Expression and/or cellular distribution changes of tight junction (TJ) proteins were restricted to increased claudin-2 over a time course that correlated with an apparent shift in its distribution from the nucleus to the membrane fraction of the cell. PIP 640-mediated epithelial changes were distinct from the combined actions of the pro-inflammatory cytokines tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ). While TNF-α/IFN-γ treatment also increased MLC-pS19 levels, these cytokines enhanced AB transport for 70-kDa dextran and decreased occludin expression at TJs. Claudin-2-dependent changes induced by PIP 640 resulted in an AB transport bias for positively-charged macromolecules demonstrated in vitro using charge variants of 4-kDa dextrans and by comparing transport of salmon calcitonin to exenatide. Comparable outcomes of increased TJ localization of claudin-2 and enhanced transport of these therapeutic peptides that biased toward cationic characteristics was demonstrated in vivo following after intra-luminal injection into rat jejunum. Together, these data have shown a potential mechanism for PIP 640 to enhance paracellular permeability of solutes in the size range of small therapeutic peptides that is biased toward positively-charged solutes.
Collapse
|
42
|
Werle M, Föger F. Peroral peptide delivery: Peptidase inhibition as a key concept for commercial drug products. Bioorg Med Chem 2018; 26:2906-2913. [DOI: 10.1016/j.bmc.2017.08.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 08/10/2017] [Indexed: 11/16/2022]
|
43
|
Almansour K, Taverner A, Eggleston IM, Mrsny RJ. Mechanistic studies of a cell-permeant peptide designed to enhance myosin light chain phosphorylation in polarized intestinal epithelia. J Control Release 2018; 279:208-219. [PMID: 29614254 DOI: 10.1016/j.jconrel.2018.03.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 12/15/2022]
Abstract
Tight junction (TJ) structures restrict the movement of solutes between adjacent epithelial cells to maintain homeostatic conditions. A peptide, termed PIP 640, with the capacity to regulate the transient opening of intestinal TJ structures through an endogenous mechanism involving the induction of myosin light chain (MLC) phosphorylation at serine 19 (MLC-pS19) has provided a promising new method to enhance the in vivo oral bioavailability of peptide therapeutics. PIP 640 is a decapeptide composed of all D-amino acids (rrdykvevrr-NH2) that contains a central sequence designed to emulates a specific domain of C-kinase potentiated protein phosphatase-1 inhibitor-17 kDa (CPI-17) surrounded by positively-charged amino acids that provide a cell penetrating peptide (CPP)-like character. Here, we examine compositional requirements of PIP 640 with regard to its actions on MLC phosphorylation, its intracellular localization to TJ structures, and its interactions with MLC phosphatase (MLCP) elements that correlate with enhanced solute uptake. These studies showed that a glutamic acid and tyrosine within this peptide are critical for PIP 640 to retain its ability to increase MLC-pS19 levels and enhance the permeability of macromolecular solutes of the size range of therapeutic peptides without detectable cytotoxicity. On the other hand, exchange of the aspartic acid for alanine and then arginine resulted in an increasingly greater bias toward protein phosphatase-1 (PP1) relative to MLCP inhibition, an outcome that resulted in increased paracellular permeability for solutes in the size range of therapeutic peptides, but with a significant increase in cytotoxicity. Together, these data further our understanding of the composition requirements of PIP 640 with respect to the desired goal of transiently altering the intestinal epithelial cell paracellular barrier properties through an endogenous mechanism, providing a novel approach to enhance the oral bioavailability of poorly absorbed therapeutic agents of < ~ 5 kDa.
Collapse
Affiliation(s)
- Khaled Almansour
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, UK
| | - Alistair Taverner
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, UK
| | - Ian M Eggleston
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, UK
| | - Randall J Mrsny
- Department of Pharmacy and Pharmacology, University of Bath, Bath BA2 7AY, UK.
| |
Collapse
|
44
|
Xing C, Xing JF, Ge ZQ. Inhibition of proteases activity in intestine needs a sustainable acidic environment rather than a transient. Drug Dev Ind Pharm 2017; 43:1648-1655. [PMID: 28541760 DOI: 10.1080/03639045.2017.1328433] [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: 10/19/2022]
Abstract
α-Chymotrypsin (α-CT) and trypsin are important components of the enzymatic barrier. They could degrade the therapeutic proteins and peptides, inhibit their activity consequently, and thereby reduce their oral bioavailability. Acidic agents, as one type of indirect protease inhibitors, have shown proof of concept in clinical trials. We report here the inactivated proteases due to acid influence can be reactivated immediately by environmental pH recovery regardless of how long the inactivation last. To keep the inactivation time of proteases for 4-5 h, we designed and prepared a sustained-release tablet containing citric acid (CA) which can effectively reduce the pH below 5.0 and maintain it for 5 h in the dissolution-reaction medium. The activity of α-CT and trypsin was quantified by analyzing the residual amount of their respective substrates BTEE and TAME. More than 80% of the substrates were survived in 5.0 h of incubation, whereas the common tablet inhibited the proteases activity for only two hours in the same experimental medium. It indicates that the sustained-release tablet loaded with CA can efficiently inhibit the α-CT and trypsin activity longer than the common tablet. The results will be beneficial for designing and formulating the peroral administration of peptide and protein drugs.
Collapse
Affiliation(s)
- Chang Xing
- a Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology , Tianjin University, Education Ministry Key Laboratory of Systems Bioengineering , Tianjin , PR China
| | - Jin-Feng Xing
- a Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology , Tianjin University, Education Ministry Key Laboratory of Systems Bioengineering , Tianjin , PR China
| | - Zhi-Qiang Ge
- a Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology , Tianjin University, Education Ministry Key Laboratory of Systems Bioengineering , Tianjin , PR China
| |
Collapse
|
45
|
Malhaire H, Gimel JC, Roger E, Benoît JP, Lagarce F. How to design the surface of peptide-loaded nanoparticles for efficient oral bioavailability? Adv Drug Deliv Rev 2016; 106:320-336. [PMID: 27058155 DOI: 10.1016/j.addr.2016.03.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 03/17/2016] [Accepted: 03/28/2016] [Indexed: 01/22/2023]
Abstract
The oral administration of proteins is a current challenge to be faced in the field of therapeutics. There is currently much interest in nanocarriers since they can enhance oral bioavailability. For lack of a clear definition, the key characteristics of nanoparticles have been highlighted. Specific surface area is one of these characteristics and represents a huge source of energy that can be used to control the biological fate of the carrier. The review discusses nanocarrier stability, mucus interaction and absorption through the intestinal epithelium. The protein corona, which has raised interest over the last decade, is also discussed. The universal ideal surface is a myth and over-coated carriers are not a solution either. Besides, common excipients can be useful on several targets. The suitable design should rather take into account the composition, structure and behavior of unmodified nanomaterials.
Collapse
|
46
|
Aguirre TAS, Teijeiro-Osorio D, Rosa M, Coulter IS, Alonso MJ, Brayden DJ. Current status of selected oral peptide technologies in advanced preclinical development and in clinical trials. Adv Drug Deliv Rev 2016; 106:223-241. [PMID: 26921819 DOI: 10.1016/j.addr.2016.02.004] [Citation(s) in RCA: 219] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 01/12/2023]
Abstract
The development of oral dosage forms that allows absorption of therapeutic peptides to the systemic circulation is one of the greatest challenges for the pharmaceutical industry. Currently, a number of technologies including either mixtures of penetration enhancers or protease inhibitors and/or nanotechnology-based products are under clinical development. Typically, these formulations are presented in the form of enteric-coated tablets or capsules. Systems undergoing preclinical investigation include further advances in nanotechnology, including intestinal microneedle patches, as well as their combination with regional delivery to the colon. This review critically examines four selected promising oral peptide technologies at preclinical stage and the twelve that have progressed to clinical trials, as indicated in www.clinicaltrials.gov. We examined these technologies under the criteria of peptide selection, formulation design, system components and excipients, intestinal mechanism of action, efficacy in man, and safety issues. The conclusion is that most of the technologies in clinical trials are incremental rather than paradigm-shifting and that even the more clinically advanced oral peptide drugs examples of oral bioavailability appear to yield oral bioavailability values of only 1-2% and are, therefore, only currently suitable for a limited range of peptides.
Collapse
Affiliation(s)
- T A S Aguirre
- Centro de Ciências Exatas e Tecnologia, Universidade de Caxias do Sul (UCS), Caxias do Sul, Brazil
| | - D Teijeiro-Osorio
- CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela, Spain
| | - M Rosa
- Sigmoid Pharma, Dublin City University, Invent Centre, Dublin 9, Ireland
| | - I S Coulter
- Sigmoid Pharma, Dublin City University, Invent Centre, Dublin 9, Ireland
| | - M J Alonso
- CIMUS Research Institute, University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - D J Brayden
- UCD School of Veterinary Medicine and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
47
|
Maher S, Mrsny RJ, Brayden DJ. Intestinal permeation enhancers for oral peptide delivery. Adv Drug Deliv Rev 2016; 106:277-319. [PMID: 27320643 DOI: 10.1016/j.addr.2016.06.005] [Citation(s) in RCA: 248] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 06/07/2016] [Accepted: 06/09/2016] [Indexed: 12/15/2022]
Abstract
Intestinal permeation enhancers (PEs) are one of the most widely tested strategies to improve oral delivery of therapeutic peptides. This article assesses the intestinal permeation enhancement action of over 250 PEs that have been tested in intestinal delivery models. In depth analysis of pre-clinical data is presented for PEs as components of proprietary delivery systems that have progressed to clinical trials. Given the importance of co-presentation of sufficiently high concentrations of PE and peptide at the small intestinal epithelium, there is an emphasis on studies where PEs have been formulated with poorly permeable molecules in solid dosage forms and lipoidal dispersions.
Collapse
|
48
|
Serre E, Boutin Y, Langevin ME, Lutin F, Pedneault K, Lacour S, Bazinet L. Deacidification of cranberry juice protects against disruption of in-vitro intestinal cell barrier integrity. J Funct Foods 2016. [DOI: 10.1016/j.jff.2016.06.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
49
|
Castro-Cabado M, Casado A, San Román J. Effect of CaO in the thermal crosslinking of maltodextrin and citric acid: A cooperative action of condensation and ionic interactions. J Appl Polym Sci 2016. [DOI: 10.1002/app.44203] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
| | - A.L. Casado
- URSA Insulation S.A; Paseo de Recoletos 3 Madrid 28004 Spain
| | - J. San Román
- Institute of Polymer Science and Technology, and CIBER-BBN; C/Juan de la Cierva 3 Madrid 28006 Spain
| |
Collapse
|
50
|
Aguirre TAS, Aversa V, Rosa M, Guterres SS, Pohlmann AR, Coulter I, Brayden DJ. Coated minispheres of salmon calcitonin target rat intestinal regions to achieve systemic bioavailability: Comparison between intestinal instillation and oral gavage. J Control Release 2016; 238:242-252. [PMID: 27480451 DOI: 10.1016/j.jconrel.2016.07.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/26/2016] [Accepted: 07/28/2016] [Indexed: 10/21/2022]
Abstract
Achieving oral peptide delivery is an elusive challenge. Emulsion-based minispheres of salmon calcitonin (sCT) were synthesized using single multiple pill (SmPill®) technology incorporating the permeation enhancers (PEs): sodium taurodeoxycholate (NaTDC), sodium caprate (C10), or coco-glucoside (CG), or the pH acidifier, citric acid (CA). Minispheres were coated with an outer layer of Eudragit® L30 D-55 (designed for jejunal release) or Surelease®/Pectin (designed for colonic release). The process was mild and in vitro biological activity of sCT was retained upon release from minispheres stored up to 4months. In vitro release profiles suggested that sCT was released from minispheres by diffusion through coatings due to swelling of gelatin and the polymeric matrix upon contact with PBS at pH6.8. X-ray analysis confirmed that coated minispheres dissolved at the intended intestinal region of rats following oral gavage. Uncoated minispheres at a dose of ~2000I.U.sCT/kg were administered to rats by intra-jejunal (i.j.) or intra-colonic (i.c.) instillation and caused hypocalcaemia. Notable sCT absolute bioavailability (F) values were: 5.5% from minispheres containing NaTDC (i.j), 17.3% with CG (i.c.) and 18.2% with C10 (i.c.). Coated minispheres administered by oral gavage at threefold higher doses also induced hypocalcaemia. A highly competitive F value of 2.7% was obtained for orally-administered sCT-minispheres containing CG (45μmol/kg) and coated with Eudragit®. In conclusion, the SmPill® technology is a potential dosage form for several peptides when formulated with PEs and coated for regional delivery. PK data from instillations over-estimates oral bioavailability and poorly predicts rank ordering of formulations.
Collapse
Affiliation(s)
- Tanira A S Aguirre
- UCD School of Veterinary Science and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland; Sigmoid Pharma, Dublin City University, Invent Centre, Dublin 9, Ireland
| | - Vincenzo Aversa
- Sigmoid Pharma, Dublin City University, Invent Centre, Dublin 9, Ireland
| | - Mónica Rosa
- Sigmoid Pharma, Dublin City University, Invent Centre, Dublin 9, Ireland
| | - Sílvia S Guterres
- Programa de pós Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Adriana R Pohlmann
- Programa de pós Graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Departamento de Química Orgânica, Instituto de Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ivan Coulter
- Sigmoid Pharma, Dublin City University, Invent Centre, Dublin 9, Ireland
| | - David J Brayden
- UCD School of Veterinary Science and UCD Conway Institute, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|