1
|
Pierzynowska K, Morcinek-Orłowska J, Gaffke L, Jaroszewicz W, Skowron PM, Węgrzyn G. Applications of the phage display technology in molecular biology, biotechnology and medicine. Crit Rev Microbiol 2024; 50:450-490. [PMID: 37270791 DOI: 10.1080/1040841x.2023.2219741] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 10/17/2022] [Accepted: 05/25/2023] [Indexed: 06/06/2023]
Abstract
The phage display technology is based on the presentation of peptide sequences on the surface of virions of bacteriophages. Its development led to creation of sophisticated systems based on the possibility of the presentation of a huge variability of peptides, attached to one of proteins of bacteriophage capsids. The use of such systems allowed for achieving enormous advantages in the processes of selection of bioactive molecules. In fact, the phage display technology has been employed in numerous fields of biotechnology, as diverse as immunological and biomedical applications (in both diagnostics and therapy), the formation of novel materials, and many others. In this paper, contrary to many other review articles which were focussed on either specific display systems or the use of phage display in selected fields, we present a comprehensive overview of various possibilities of applications of this technology. We discuss an usefulness of the phage display technology in various fields of science, medicine and the broad sense of biotechnology. This overview indicates the spread and importance of applications of microbial systems (exemplified by the phage display technology), pointing to the possibility of developing such sophisticated tools when advanced molecular methods are used in microbiological studies, accompanied with understanding of details of structures and functions of microbial entities (bacteriophages in this case).
Collapse
Affiliation(s)
- Karolina Pierzynowska
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | | | - Lidia Gaffke
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Weronika Jaroszewicz
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdańsk, Poland
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Gdansk, Poland
| |
Collapse
|
2
|
Subramanian A. Emerging roles of bacteriophage-based therapeutics in combating antibiotic resistance. Front Microbiol 2024; 15:1384164. [PMID: 39035437 PMCID: PMC11257900 DOI: 10.3389/fmicb.2024.1384164] [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: 02/08/2024] [Accepted: 06/06/2024] [Indexed: 07/23/2024] Open
Abstract
Amid the growing challenge of antibiotic resistance on a global scale, there has been a notable resurgence in bacteriophage-based treatments, signaling a shift in our approach to managing infections. Bacteriophages (BPs), bacterial predators of nature, present a promising alternative for tackling infections caused by antibiotic-resistant pathogens. This review delves into the intricate relationship between bacteriophages and resistant bacteria, exploring various treatment strategies. Drawing upon both preclinical and clinical studies, the review highlights the effectiveness of bacteriophage therapy, particularly when integrated synergistically with conventional antibiotics. It discusses various treatment approaches for systemic and localized infections, demonstrating the adaptability of bacteriophage therapy across different clinical scenarios. Furthermore, the formulation and delivery of bacteriophages shed light on the various methods used to encapsulate and administer them effectively. It also acknowledges the challenge of bacterial resistance to bacteriophages and the ongoing efforts to overcome this hurdle. In addition, this review highlights the importance of the bacteriophage sensitivity profile (phagogram), which helps tailor treatment regimens to individual patients and specific pathogens. By surpassing the limitations of traditional antibiotics, bacteriophage-based therapies offer a personalized and potent solution against antibiotic resistance, promising to reshape the future of infectious disease management.
Collapse
|
3
|
Huang Z, Meng H, Xu L, Pei X, Xiong J, Wang Y, Zhan X, Li S, He Y. Liposomes in the cosmetics: present and outlook. J Liposome Res 2024:1-13. [PMID: 38712581 DOI: 10.1080/08982104.2024.2341139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 04/04/2024] [Indexed: 05/08/2024]
Abstract
Liposomes are small spherical vesicles composed of phospholipid bilayers capable of encapsulating a variety of ingredients, including water- and oil-soluble compound, which are one of the most commonly used piggybacking and delivery techniques for many active ingredients and different compounds in biology, medicine and cosmetics. With the increasing number of active cosmetic ingredients, the concomitant challenge is to effectively protect, transport, and utilize these substances in a judicious manner. Many cosmetic ingredients are ineffective both topically and systemically when applied to the skin, thus changing the method of delivery and interaction with the skin of the active ingredients is a crucial step toward improving their effectiveness. Liposomes can improve the delivery of active ingredients to the skin, enhance their stability, and ultimately, improve the efficacy of cosmetics and and pharmaceuticals. In this review, we summarized the basic properties of liposomes and their recent advances of functionalities in cosmetics and and pharmaceuticals. Also, the current state of the art in the field is discussed and the prospects for future research areas are highlighted. We hope that this review will provide ideas and inspiration on the application and development of cosmetics and pharmaceuticals.
Collapse
Affiliation(s)
- Zhaohe Huang
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Hong Meng
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Li Xu
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Xiaojing Pei
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Jie Xiong
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Yanan Wang
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Xin Zhan
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Shujing Li
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| | - Yifan He
- College of Chemistry and Materials Engineering and Institute of Cosmetic Regulatory Science, Beijing Technology and Business University, Beijing, P. R. China
| |
Collapse
|
4
|
Aljabali AAA, Aljbaly MBM, Obeid MA, Shahcheraghi SH, Tambuwala MM. The Next Generation of Drug Delivery: Harnessing the Power of Bacteriophages. Methods Mol Biol 2024; 2738:279-315. [PMID: 37966606 DOI: 10.1007/978-1-0716-3549-0_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2023]
Abstract
The use of biomaterials, such as bacteriophages, as drug delivery vehicles (DDVs) has gained increasing interest in recent years due to their potential to address the limitations of conventional drug delivery systems. Bacteriophages offer several advantages as drug carriers, such as high specificity for targeting bacterial cells, low toxicity, and the ability to be engineered to express specific proteins or peptides for enhanced targeting and drug delivery. In addition, bacteriophages have been shown to reduce the development of antibiotic resistance, which is a major concern in the field of antimicrobial therapy. Many initiatives have been taken to take up various payloads selectively and precisely by surface functionalization of the outside or interior of self-assembling viral protein capsids. Bacteriophages have emerged as a promising platform for the targeted delivery of therapeutic agents, including drugs, genes, and imaging agents. They possess several properties that make them attractive as drug delivery vehicles, including their ability to specifically target bacterial cells, their structural diversity, their ease of genetic manipulation, and their biocompatibility. Despite the potential advantages of using bacteriophages as drug carriers, several challenges and limitations need to be addressed. One of the main challenges is the limited host range of bacteriophages, which restricts their use to specific bacterial strains. However, this can also be considered as an advantage, as it allows for precise and targeted drug delivery to the desired bacterial cells. The use of biomaterials, including bacteriophages, as drug delivery vehicles has shown promising potential to address the limitations of conventional drug delivery systems. Further research is needed to fully understand the potential of these biomaterials and address the challenges and limitations associated with their use.
Collapse
Affiliation(s)
- Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan.
| | | | - Mohammad A Obeid
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Yarmouk University, Irbid, Jordan
| | - Seyed Hossein Shahcheraghi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Murtaza M Tambuwala
- Lincoln Medical School, Brayford Pool Campus, University of Lincoln, Lincoln, UK.
| |
Collapse
|
5
|
Ragothaman M, Yoo SY. Engineered Phage-Based Cancer Vaccines: Current Advances and Future Directions. Vaccines (Basel) 2023; 11:vaccines11050919. [PMID: 37243023 DOI: 10.3390/vaccines11050919] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/22/2023] [Accepted: 04/27/2023] [Indexed: 05/28/2023] Open
Abstract
Bacteriophages have emerged as versatile tools in the field of bioengineering, with enormous potential in tissue engineering, vaccine development, and immunotherapy. The genetic makeup of phages can be harnessed for the development of novel DNA vaccines and antigen display systems, as they can provide a highly organized and repetitive presentation of antigens to immune cells. Bacteriophages have opened new possibilities for the targeting of specific molecular determinants of cancer cells. Phages can be used as anticancer agents and carriers of imaging molecules and therapeutics. In this review, we explored the role of bacteriophages and bacteriophage engineering in targeted cancer therapy. The question of how the engineered bacteriophages can interact with the biological and immunological systems is emphasized to comprehend the underlying mechanism of phage use in cancer immunotherapy. The effectiveness of phage display technology in identifying high-affinity ligands for substrates, such as cancer cells and tumor-associated molecules, and the emerging field of phage engineering and its potential in the development of effective cancer treatments are discussed. We also highlight phage usage in clinical trials as well as the related patents. This review provides a new insight into engineered phage-based cancer vaccines.
Collapse
Affiliation(s)
- Murali Ragothaman
- BIO-IT Foundry Technology Institute, Pusan National University, Busan 46241, Republic of Korea
| | - So Young Yoo
- BIO-IT Foundry Technology Institute, Pusan National University, Busan 46241, Republic of Korea
| |
Collapse
|
6
|
Azari M, Bahreini F, Uversky VN, Rezaei N. Current therapeutic approaches and promising perspectives of using bioengineered peptides in fighting chemoresistance in triple-negative breast cancer. Biochem Pharmacol 2023; 210:115459. [PMID: 36813121 DOI: 10.1016/j.bcp.2023.115459] [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: 10/04/2022] [Revised: 02/11/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023]
Abstract
Breast cancer is a collation of malignancies that manifest in the mammary glands at the early stages. Among breast cancer subtypes, triple-negative breast cancer (TNBC) shows the most aggressive behavior, with apparent stemness features. Owing to the lack of response to hormone therapy and specific targeted therapies, chemotherapy remains the first line of the TNBC treatment. However, the acquisition of resistance to chemotherapeutic agents increase therapy failure, and promotes cancer recurrence and distant metastasis. Invasive primary tumors are the birthplace of cancer burden, though metastasis is a key attribute of TNBC-associated morbidity and mortality. Targeting the chemoresistant metastases-initiating cells via specific therapeutic agents with affinity to the upregulated molecular targets is a promising step in the TNBC clinical management. Exploring the capacity of peptides as biocompatible entities with the specificity of action, low immunogenicity, and robust efficacy provides a principle for designing peptide-based drugs capable of increasing the efficacy of current chemotherapy agents for selective targeting of the drug-tolerant TNBC cells. Here, we first focus on the resistance mechanisms that TNBC cells acquire to evade the effect of chemotherapeutic agents. Next, the novel therapeutic approaches employing tumor-targeting peptides to exploit the mechanisms of drug resistance in chemorefractory TNBC are described.
Collapse
Affiliation(s)
- Mandana Azari
- School of Chemical Engineering-Biotechnology, College of Engineering, University of Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farbod Bahreini
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran; Research Center for Immunodeficiencies (RCID), Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
7
|
One-Step Pharmaceutical Preparation of PEG-Modified Exosomes Encapsulating Anti-Cancer Drugs by a High-Pressure Homogenization Technique. Pharmaceuticals (Basel) 2023; 16:ph16010108. [PMID: 36678605 PMCID: PMC9865360 DOI: 10.3390/ph16010108] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/04/2023] [Accepted: 01/07/2023] [Indexed: 01/12/2023] Open
Abstract
The use of exosomes encapsulating therapeutic agents for the treatment of diseases is of increasing interest. However, some concerns such as limited efficiency and scalability of conventional drug encapsulation methods to exosomes have still remained; thus, a new approach that enables encapsulation of therapeutic agents with superior efficiency and scalability is required. Herein, we used RAW264 macrophage cell-derived exosomes (RAW-Exos) and demonstrated that high-pressure homogenization (HPH) using a microfluidizer decreased their particle size without changing their morphology, the amount of exosomal marker proteins, and cellular uptake efficiency into RAW264 and colon-26 cancer cells. Moreover, HPH allowed for modification of polyethylene glycol (PEG)-conjugated lipids onto RAW-Exos, as well as encapsulation of the anti-cancer agent doxorubicin. Importantly, the doxorubicin encapsulation efficiency became higher upon increasing the process pressure and simultaneous HPH with PEG-lipids. Moreover, treatment with PEG-modified RAW-Exos encapsulating doxorubicin significantly suppressed tumor growth in colon-26-bearing mice. Taken together, these results suggest that HPH using a microfluidizer could be useful to prepare PEG-modified Exos encapsulating anti-cancer drugs via a one-step pharmaceutical process, and that the prepared functional Exos could be applied for the treatment of cancer in vivo.
Collapse
|
8
|
Ling H, Lou X, Luo Q, He Z, Sun M, Sun J. Recent advances in bacteriophage-based therapeutics: Insight into the post-antibiotic era. Acta Pharm Sin B 2022; 12:4348-4364. [PMID: 36561998 PMCID: PMC9764073 DOI: 10.1016/j.apsb.2022.05.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/07/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
Antibiotic resistance is one of the biggest threats to global health, as it can make the treatment of bacterial infections in humans difficult owing to their high incidence rate, mortality, and treatment costs. Bacteriophage, which constitutes a type of virus that can kill bacteria, is a promising alternative strategy against antibiotic-resistant bacterial infections. Although bacteriophage therapy was first used nearly a century ago, its development came to a standstill after introducing the antibiotics. Nowadays, with the rise in antibiotic resistance, bacteriophage therapy is in the spotlight again. As bacteriophage therapy is safe and has significant anti-bacterial activity, some specific types of bacteriophages (such as bacteriophage phiX174 and Pyo bacteriophage complex liquid) entered into phase III clinical trials. Herein, we review the key points of the antibiotic resistance crisis and illustrate the factors that support the renewal of bacteriophage applications. By summarizing recent state-of-the-art studies and clinical data on bacteriophage treatment, we introduced (i) the pharmacological mechanisms and advantages of antibacterial bacteriophages, (ii) bacteriophage preparations with clinical potential and bacteriophage-derived anti-bacterial treatment strategies, and (iii) bacteriophage therapeutics aimed at multiple infection types and infection-induced cancer treatments. Finally, we highlighted the challenges and critical perspectives of bacteriophage therapy for future clinical development.
Collapse
Affiliation(s)
- Hao Ling
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinyu Lou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qiuhua Luo
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang 110001, China
| | - Zhonggui He
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Mengchi Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China,Corresponding authors.
| | - Jin Sun
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China,Corresponding authors.
| |
Collapse
|
9
|
Fukuta T, Oku N, Kogure K. Application and Utility of Liposomal Neuroprotective Agents and Biomimetic Nanoparticles for the Treatment of Ischemic Stroke. Pharmaceutics 2022; 14:pharmaceutics14020361. [PMID: 35214092 PMCID: PMC8877231 DOI: 10.3390/pharmaceutics14020361] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 12/04/2022] Open
Abstract
Ischemic stroke is still one of the leading causes of high mortality and severe disability worldwide. Therapeutic options for ischemic stroke and subsequent cerebral ischemia/reperfusion injury remain limited due to challenges associated with drug permeability through the blood-brain barrier (BBB). Neuroprotectant delivery with nanoparticles, including liposomes, offers a promising solution to address this problem, as BBB disruption following ischemic stroke allows nanoparticles to pass through the intercellular gaps between endothelial cells. To ameliorate ischemic brain damage, a number of nanotherapeutics encapsulating neuroprotective agents, as well as surface-modified nanoparticles with specific ligands targeting the injured brain regions, have been developed. Combination therapy with nanoparticles encapsulating neuroprotectants and tissue plasminogen activator (t-PA), a globally approved thrombolytic agent, has been demonstrated to extend the narrow therapeutic time window of t-PA. In addition, the design of biomimetic drug delivery systems (DDS) employing circulating cells (e.g., leukocytes, platelets) with unique properties has recently been investigated to overcome the injured BBB, utilizing these cells’ inherent capability to penetrate the ischemic brain. Herein, we review recent findings on the application and utility of nanoparticle DDS, particularly liposomes, and various approaches to developing biomimetic DDS functionalized with cellular membranes/membrane proteins for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Tatsuya Fukuta
- Department of Physical Pharmaceutics, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichiban-cho, Wakayama 640-8156, Japan
| | - Naoto Oku
- Faculty of Pharma-Science, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Kentaro Kogure
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima 770-8505, Japan
| |
Collapse
|
10
|
Kim SH, Lee EH, Kim HJ, Kim AR, Kim YE, Lee JH, Yoon MY, Koh SH. Development of a Low-Molecular-Weight Aβ42 Detection System Using a Enzyme-Linked Peptide Assay. Biomolecules 2021; 11:1818. [PMID: 34944462 PMCID: PMC8699310 DOI: 10.3390/biom11121818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/29/2021] [Accepted: 12/01/2021] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) is a degenerative brain disease that is the most common cause of dementia. The incidence of AD is rapidly rising because of the aging of the world population. Because AD is presently incurable, early diagnosis is very important. The disease is characterized by pathological changes such as deposition of senile plaques and decreased concentration of the amyloid-beta 42 (Aβ42) peptide in the cerebrospinal fluid (CSF). The concentration of Aβ42 in the CSF is a well-studied AD biomarker. The specific peptide probe was screened through four rounds of biopanning, which included the phage display process. The screened peptide showed strong binding affinity in the micromolar range, and the enzyme-linked peptide assay was optimized using the peptide we developed. This diagnostic method showed specificity toward Aβ42 in the presence of other proteins. The peptide-binding site was also estimated using molecular docking analysis. Finally, the diagnostic method we developed could significantly distinguish patients who were classified based on amyloid PET images.
Collapse
Affiliation(s)
- Sang-Heon Kim
- Department of Chemistry and Research, Institute of Natural Sciences, Hanyang University, Seoul 04763, Korea; (S.-H.K.); (A.-R.K.)
| | - Eun-Hye Lee
- Departments of Neurology, Hanyang University Guri Hospital, Guri 11923, Korea; (E.-H.L.); (Y.-E.K.)
| | - Hyung-Ji Kim
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea;
| | - A-Ru Kim
- Department of Chemistry and Research, Institute of Natural Sciences, Hanyang University, Seoul 04763, Korea; (S.-H.K.); (A.-R.K.)
| | - Ye-Eun Kim
- Departments of Neurology, Hanyang University Guri Hospital, Guri 11923, Korea; (E.-H.L.); (Y.-E.K.)
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Korea
| | - Jae-Hong Lee
- Department of Neurology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea;
| | - Moon-Young Yoon
- Department of Chemistry and Research, Institute of Natural Sciences, Hanyang University, Seoul 04763, Korea; (S.-H.K.); (A.-R.K.)
| | - Seong-Ho Koh
- Departments of Neurology, Hanyang University Guri Hospital, Guri 11923, Korea; (E.-H.L.); (Y.-E.K.)
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, Seoul 04763, Korea
| |
Collapse
|
11
|
Kashapov R, Ibragimova A, Pavlov R, Gabdrakhmanov D, Kashapova N, Burilova E, Zakharova L, Sinyashin O. Nanocarriers for Biomedicine: From Lipid Formulations to Inorganic and Hybrid Nanoparticles. Int J Mol Sci 2021; 22:7055. [PMID: 34209023 PMCID: PMC8269010 DOI: 10.3390/ijms22137055] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Encapsulation of cargoes in nanocontainers is widely used in different fields to solve the problems of their solubility, homogeneity, stability, protection from unwanted chemical and biological destructive effects, and functional activity improvement. This approach is of special importance in biomedicine, since this makes it possible to reduce the limitations of drug delivery related to the toxicity and side effects of therapeutics, their low bioavailability and biocompatibility. This review highlights current progress in the use of lipid systems to deliver active substances to the human body. Various lipid compositions modified with amphiphilic open-chain and macrocyclic compounds, peptide molecules and alternative target ligands are discussed. Liposome modification also evolves by creating new hybrid structures consisting of organic and inorganic parts. Such nanohybrid platforms include cerasomes, which are considered as alternative nanocarriers allowing to reduce inherent limitations of lipid nanoparticles. Compositions based on mesoporous silica are beginning to acquire no less relevance due to their unique features, such as advanced porous properties, well-proven drug delivery efficiency and their versatility for creating highly efficient nanomaterials. The types of silica nanoparticles, their efficacy in biomedical applications and hybrid inorganic-polymer platforms are the subject of discussion in this review, with current challenges emphasized.
Collapse
Affiliation(s)
- Ruslan Kashapov
- A.E. Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of RAS, Arbuzov Street 8, 420088 Kazan, Russia; (A.I.); (R.P.); (D.G.); (N.K.); (E.B.); (L.Z.); (O.S.)
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Bacteriophages as Therapeutic and Diagnostic Vehicles in Cancer. Pharmaceuticals (Basel) 2021; 14:ph14020161. [PMID: 33671476 PMCID: PMC7923149 DOI: 10.3390/ph14020161] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/11/2022] Open
Abstract
Evolution of nanomedicine is the re-design of synthetic and biological carriers to implement novel theranostic platforms. In recent years, bacteriophage research favors this process, which has opened up new roads in drug and gene delivery studies. By displaying antibodies, peptides, or proteins on the surface of different bacteriophages through the phage display technique, it is now possible to unravel specific molecular determinants of both cancer cells and tumor-associated microenvironmental molecules. Downstream applications are manifold, with peptides being employed most of the times to functionalize drug carriers and improve their therapeutic index. Bacteriophages themselves were proven, in this scenario, to be good carriers for imaging molecules and therapeutics as well. Moreover, manipulation of their genetic material to stably vehiculate suicide genes within cancer cells substantially changed perspectives in gene therapy. In this review, we provide examples of how amenable phages can be used as anticancer agents, especially because their systemic administration is possible. We also provide some insights into how their immunogenic profile can be modulated and exploited in immuno-oncology for vaccine production.
Collapse
|
13
|
Fukuta T, Yoshimi S, Kogure K. Leukocyte-Mimetic Liposomes Penetrate Into Tumor Spheroids and Suppress Spheroid Growth by Encapsulated Doxorubicin. J Pharm Sci 2020; 110:1701-1709. [PMID: 33129835 DOI: 10.1016/j.xphs.2020.10.049] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/11/2020] [Accepted: 10/26/2020] [Indexed: 11/18/2022]
Abstract
As leukocytes can penetrate into deep regions of a tumor mass, leukocyte-mimetic liposomes (LM-Lipo) containing leukocyte membrane proteins are also expected to penetrate into tumors by exerting properties of those membrane proteins. The aim of the present study was to examine whether LM-Lipo, which were recently demonstrated to actively pass through inflamed endothelial layers, can penetrate into tumor spheroids, and to investigate the potential of LM-Lipo for use as an anticancer drug carrier. We prepared LM-Lipo via intermembrane protein transfer from human leukemia cells; transfer of leukocyte membrane proteins onto the liposomes was determined by Western blotting. LM-Lipo demonstrated a significantly high association with human lung cancer A549 cells compared with plain liposomes, which contributed to effective anti-proliferative action by encapsulated doxorubicin hydrochloride (DOX). Confocal microscopic images showed that LM-Lipo, but not plain liposomes, could efficiently penetrate into A549 tumor spheroids. Moreover, DOX-encapsulated LM-Lipo significantly suppressed tumor spheroid growth. Thus, leukocyte membrane proteins transferred onto LM-Lipo retained their unique function, which allowed for efficient penetration of the liposomes into tumor spheroids, similar to leukocytes. In conclusion, these results suggest that LM-Lipo could be a useful tumor-penetrating drug delivery system for cancer treatment.
Collapse
Affiliation(s)
- Tatsuya Fukuta
- Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima 770-8505, Japan.
| | - Shintaro Yoshimi
- Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima 770-8505, Japan
| | - Kentaro Kogure
- Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima 770-8505, Japan
| |
Collapse
|
14
|
Kim SH, Lee EH, Lee SC, Kim AR, Park HH, Son JW, Koh SH, Yoon MY. Development of peptide aptamers as alternatives for antibody in the detection of amyloid-beta 42 aggregates. Anal Biochem 2020; 609:113921. [PMID: 32828793 DOI: 10.1016/j.ab.2020.113921] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/13/2020] [Accepted: 08/15/2020] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) causes cognitive impairment and serious social isolation. However, there are no effective treatments and even no established confirmatory diagnostic tools for the disease. Amyloid beta (Aβ) aggregation in the brain is the best-known pathognomonic mechanism of AD, so various methods for Aβ detection have been developed for the diagnosis of this disease. We synthesized two novel, ultra-sensitive peptide probes specialized in detecting Aβ aggregates, and examined their potential for future diagnostic application. The peptides are produced through phage high-throughput screening (HTS) and amplified through a serial process called biopanning, which is a repeating method of elution and amplification of probes. We picked phages specific for amyloid from two kinds of phage display. The synthesized peptides were confirmed to have excellent binding affinity to Aβ aggregates, by immunohistochemical staining and western blotting using the brains of 3X transgenic (Tg) AD mice at different stages (5-7, 12-17 months old) of AD severity. In the present study, it was confirmed that newly developed amyloid-binding peptides could be used as novel probes for the detection of Aβ aggregates, which can be used for clinical diagnosis of AD in the future.
Collapse
Affiliation(s)
- Sang-Heon Kim
- Department of Chemistry and Research Institute of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Eun-Hye Lee
- Department of Neurology, Hanyang University College of Medicine, 11923, Gyeongchun-ro, Guri-Si, Gyeonggi-do, Republic of Korea
| | - Sang-Choon Lee
- Department of Chemistry, Georgia State University, Atlanta, GA, 30303, USA
| | - A-Ru Kim
- Department of Chemistry and Research Institute of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea
| | - Hyun-Hee Park
- Department of Neurology, Hanyang University College of Medicine, 11923, Gyeongchun-ro, Guri-Si, Gyeonggi-do, Republic of Korea
| | - Jeong-Woo Son
- Department of Neurology, Hanyang University College of Medicine, 11923, Gyeongchun-ro, Guri-Si, Gyeonggi-do, Republic of Korea
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University College of Medicine, 11923, Gyeongchun-ro, Guri-Si, Gyeonggi-do, Republic of Korea; Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science & Engineering, 11923, Gyeongchun-ro, Guri-Si, Gyeonggi-do, Republic of Korea.
| | - Moon-Young Yoon
- Department of Chemistry and Research Institute of Natural Sciences, Hanyang University, Seoul, 04763, Republic of Korea.
| |
Collapse
|
15
|
Targeting Tumor Endothelial Cells with Nanoparticles. Int J Mol Sci 2019; 20:ijms20235819. [PMID: 31756900 PMCID: PMC6928777 DOI: 10.3390/ijms20235819] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/14/2019] [Accepted: 11/18/2019] [Indexed: 12/12/2022] Open
Abstract
Because angiogenesis is a major contributor to cancer progression and metastasis, it is an attractive target for cancer therapy. Although a diverse number of small compounds for anti-angiogenic therapy have been developed, severe adverse effects commonly occur, since small compounds can affect not only tumor endothelial cells (TECs), but also normal endothelial cells. This low selectivity for TECs has motivated researchers to develop alternate types of drug delivery systems (DDSs). In this review, we summarize the current state of knowledge concerning the delivery of nano DDSs to TECs. Their payloads range from small compounds to nucleic acids. Perspectives regarding new therapeutic targets are also mentioned.
Collapse
|
16
|
Rajabi M, Adeyeye M, Mousa SA. Peptide-Conjugated Nanoparticles as Targeted Anti-angiogenesis Therapeutic and Diagnostic in Cancer. Curr Med Chem 2019; 26:5664-5683. [DOI: 10.2174/0929867326666190620100800] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 03/11/2019] [Accepted: 03/21/2019] [Indexed: 12/25/2022]
Abstract
:Targeting angiogenesis in the microenvironment of a tumor can enable suppression of tumor angiogenesis and delivery of anticancer drugs into the tumor. Anti-angiogenesis targeted delivery systems utilizing passive targeting such as Enhanced Permeability and Retention (EPR) and specific receptor-mediated targeting (active targeting) should result in tumor-specific targeting. One targeted anti-angiogenesis approach uses peptides conjugated to nanoparticles, which can be loaded with anticancer agents. Anti-angiogenesis agents can suppress tumor angiogenesis and thereby affect tumor growth progression (tumor growth arrest), which may be further reduced with the targetdelivered anticancer agent. This review provides an update of tumor vascular targeting for therapeutic and diagnostic applications, with conventional or long-circulating nanoparticles decorated with peptides that target neovascularization (anti-angiogenesis) in the tumor microenvironment.
Collapse
Affiliation(s)
- Mehdi Rajabi
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| | - Mary Adeyeye
- Department of Chemistry, University of Albany, State University of New York, Albany, NY 12222, United States
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, United States
| |
Collapse
|
17
|
Jiang Z, Guan J, Qian J, Zhan C. Peptide ligand-mediated targeted drug delivery of nanomedicines. Biomater Sci 2019; 7:461-471. [PMID: 30656305 DOI: 10.1039/c8bm01340c] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Targeted drug delivery is emerging as a promising strategy to achieve better clinical outcomes. Actively targeted drug delivery that utilizes overexpressed receptors or antigens on diseased tissues is receiving increasing scrutiny, especially due to the uncertainty of existence of the enhanced permeability and retention (EPR) effect in cancer patients. Peptide ligands are advantageous over other classes of targeting ligands due to their accessibility of high-throughput screening, ease of synthesis, high specificity and affinity, etc. In this review, we briefly summarize the resources of peptide ligands and discuss the pitfalls and perspectives of peptide ligand-mediated targeted delivery of nanomedicines.
Collapse
Affiliation(s)
- Zhuxuan Jiang
- Department of Pharmacology, School of Basic Medical Sciences & State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200032, P.R. China.
| | | | | | | |
Collapse
|
18
|
Fan Q, Zhang Y, Hou X, Li Z, Zhang K, Shao Q, Feng N. Improved oral bioavailability of notoginsenoside R1 with sodium glycocholate-mediated liposomes: Preparation by supercritical fluid technology and evaluation in vitro and in vivo. Int J Pharm 2018; 552:360-370. [PMID: 30292894 DOI: 10.1016/j.ijpharm.2018.10.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 09/18/2018] [Accepted: 10/02/2018] [Indexed: 01/12/2023]
Abstract
The chief objective of this research was to appraise liposomes embodying a bile salt, sodium glycocholate (SGC), as oral nanoscale drug delivery system to strengthen the bioavailability of a water-soluble and weakly penetrable pharmaceutical, notoginsenoside R1 (NGR1). NGR1-loaded liposomes were prepared with the improved supercritical reverse evaporation (ISCRPE) method and the preparation conditions were optimized with response surface methodology (RSM). The mean encapsulation efficiency (EE), particle size, and polydispersity index (PDI) of the optimized liposomal formulation (NGR1@Liposomes) were 49.49%, 308.3 nm, and 0.229, respectively. SGC-mediated liposomes (NGR1@SGC-Liposomes) were formulated based on the optimal preparation conditions and the mean EE, particle size, and PDI were 41.51%, 200.1 nm, and 0.130, respectively. The in vitro Caco-2 cellular uptake of fluorescent marker was increased by loading into NGR1@SGC-Liposomes as compared with the conventional liposomes. Furthermore, the intestinal permeability as well as the intestinal absorption of NGR1 were both significantly improved with NGR1@SGC-Liposomes as the nanovesicles. The in vivo pharmacokinetic study results showed that AUC0-t value of NGR1@SGC-Liposomes and NGR1@Liposomes was 2.68- and 2.03-fold higher than that of NGR1 aqueous solution, respectively. The AUC0-t of the NGR1@SGC-Liposomes group was significantly higher than that of NGR1@Liposomes. Thus, ISCRPE method is a feasible method for the preparation of water-soluble drug-loaded liposomes and bile salt-mediated liposomes may enhance the oral absorption of water-soluble and poorly permeable drugs.
Collapse
Affiliation(s)
- Qiangyuan Fan
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yongtai Zhang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xuefeng Hou
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhe Li
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Kai Zhang
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Qun Shao
- Open Innovation, University of Bradford, West Yorkshire BD7 1DP, UK
| | - Nianping Feng
- Department of Pharmaceutical Sciences, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
19
|
Zhang H, Guo Z, He B, Dai W, Zhang H, Wang X, Zhang Q. The Improved Delivery to Breast Cancer Based on a Novel Nanocarrier Modified with High-Affinity Peptides Discovered by Phage Display. Adv Healthc Mater 2018; 7:e1800269. [PMID: 29956504 DOI: 10.1002/adhm.201800269] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/04/2018] [Indexed: 01/06/2023]
Abstract
Ligand-targeted nanosystems have the potential to realize site-specific tumor therapy and alleviate unwanted side effects of many chemotherapeutic agents, and one of the most key issues seems to be the construction of an effective nanocarrier. Based on different processes of phage display techniques, 38 cell-binding peptides and 32 cell-internalizing peptides are discovered. Four of these ligand peptides [FIPFDPMSMRWE (FIP), NASSFPTNSRWA (NAS), GLHTSATNLYLH (GLH), and ALAVAPSRWWNE (ALA), respectively] exhibit high affinity to MCF7 human breast cancer cells. Among them, NAS and ALA are reported for the first time, whose affinities are 20.6 and 76.3 times that of the random peptide control, respectively. Both NAS and ALA modifications to doxorubicin-loaded lipid nanosytems [LP(DOX)] show stronger tumor inhibition, longer animal survival time, and less body weight loss, compared to unmodified or control peptide modified nanosystems, on an MCF7 tumor-bearing mouse model. In conclusion, the cell-binding peptide NAS and cell-internalizing peptide ALA can be used for ligand-targeted delivery of antitumor drugs. It seems that the in vivo antitumor effect of these ligand-targeted nanosystems is closely related to their ligand-cell affinity, but fairly tolerant of the ligand types.
Collapse
Affiliation(s)
- Haoran Zhang
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University Health Science Center; Beijing 100191 China
| | - Zhaoming Guo
- School of Life Science and Medicine; Dalian University of Technology; Liaoning 124221 China
| | - Bing He
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University Health Science Center; Beijing 100191 China
| | - Wenbing Dai
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University Health Science Center; Beijing 100191 China
| | - Hua Zhang
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University Health Science Center; Beijing 100191 China
| | - Xueqing Wang
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University Health Science Center; Beijing 100191 China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs; School of Pharmaceutical Sciences; Peking University Health Science Center; Beijing 100191 China
| |
Collapse
|
20
|
Lopes RS, Queiroz MAF, Gomes STM, Vallinoto ACR, Goulart LR, Ishak R. Phage display: an important tool in the discovery of peptides with anti-HIV activity. Biotechnol Adv 2018; 36:1847-1854. [PMID: 30012540 DOI: 10.1016/j.biotechadv.2018.07.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 06/14/2018] [Accepted: 07/12/2018] [Indexed: 10/28/2022]
Abstract
Human immunodeficiency virus (HIV) remains a worldwide health problem despite huge investments and research breakthroughs, and no single drug is effective in killing the virus yet. Among new strategies to control HIV infection, the phage display (PD) technology has become a promising tool in the discovery of peptides that can be used as new drugs, or also as possible vaccine candidates. This review discusses basic aspects of PD and its use to advance two main objectives related to combating HIV-1 infection: the identification of peptides that inhibit virus replication and the identification of peptides that induce the production of neutralizing antibodies. We will cover the different approaches used for mapping and selection of mimotopes, and discuss the promising results of these biologicals as antiviral agents.
Collapse
Affiliation(s)
- Ronaldo Souza Lopes
- Biological Sciences Institute, Federal University of Para (Instituto de Ciências Biológicas/Universidade Feral do Pará - ICB/UFPA), Rua Augusto Corrêa, 1 - Guamá, Belém, PA 66075-110, Brazil.
| | - Maria Alice Freitas Queiroz
- Biological Sciences Institute, Federal University of Para (Instituto de Ciências Biológicas/Universidade Feral do Pará - ICB/UFPA), Rua Augusto Corrêa, 1 - Guamá, Belém, PA 66075-110, Brazil
| | - Samara Tatielle Monteiro Gomes
- Biological Sciences Institute, Federal University of Para (Instituto de Ciências Biológicas/Universidade Feral do Pará - ICB/UFPA), Rua Augusto Corrêa, 1 - Guamá, Belém, PA 66075-110, Brazil
| | - Antonio Carlos Rosário Vallinoto
- Biological Sciences Institute, Federal University of Para (Instituto de Ciências Biológicas/Universidade Feral do Pará - ICB/UFPA), Rua Augusto Corrêa, 1 - Guamá, Belém, PA 66075-110, Brazil.
| | - Luiz Ricardo Goulart
- Institute of Biotechnology, Federal University of Uberlândia (Universidade Federal de Uberlândia - UFU), Laboratory of Nanobiotechnology, Av. Amazonas s/n, Bloco 2E, Sala 248 - Campus Umuarama, Uberlândia, MG, CEP 38400-902, Brazil.
| | - Ricardo Ishak
- Biological Sciences Institute, Federal University of Para (Instituto de Ciências Biológicas/Universidade Feral do Pará - ICB/UFPA), Rua Augusto Corrêa, 1 - Guamá, Belém, PA 66075-110, Brazil.
| |
Collapse
|
21
|
Newman MR, Benoit DSW. In Vivo Translation of Peptide-Targeted Drug Delivery Systems Discovered by Phage Display. Bioconjug Chem 2018; 29:2161-2169. [PMID: 29889510 DOI: 10.1021/acs.bioconjchem.8b00285] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Therapeutic compounds with narrow therapeutic windows and significant systemic side effects benefit from targeted drug delivery strategies. Peptide-protein interactions are often exploited for targeting, with phage display a primary method to identify high-affinity peptide ligands that bind cell surface and matrix bound receptors preferentially expressed in target tissues. After isolating and sequencing high-binding phages, peptides are easily synthesized and chemically modified for incorporation into drug delivery systems, including peptide-drug conjugates, polymers, and nanoparticles. This review describes the phage display methodology to identify targeting peptide sequences, strategies to functionalize drug carriers with phage-derived peptides, specific examples of drug carriers with in vivo translation, and limitations and future applications of phage display to drug delivery.
Collapse
Affiliation(s)
- Maureen R Newman
- Center for Musculoskeletal Research, Department of Orthopaedics , University of Rochester Medical Center , Rochester , New York 14642 , United States
| | - Danielle S W Benoit
- Center for Musculoskeletal Research, Department of Orthopaedics , University of Rochester Medical Center , Rochester , New York 14642 , United States
| |
Collapse
|