1
|
Zhu C, Mu J, Liang L. Nanocarriers for intracellular delivery of proteins in biomedical applications: strategies and recent advances. J Nanobiotechnology 2024; 22:688. [PMID: 39523313 PMCID: PMC11552240 DOI: 10.1186/s12951-024-02969-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
Protein drugs are of great importance in maintaining the normal functioning of living organisms. Indeed, they have been instrumental in combating tumors and genetic diseases for decades. Among these pharmaceutical agents, those that target intracellular components necessitate the use of therapeutic proteins to exert their effects within the targeted cells. However, the use of protein drugs is limited by their short half-life and potential adverse effects in the physiological environment. The advent of nanoparticles offers a promising avenue for prolonging the half-life of protein drugs. This is achieved by encapsulating proteins, thereby safeguarding their biological activity and ensuring precise delivery into cells. This nanomaterial-based intracellular protein drug delivery system mitigates the rapid hydrolysis and unwarranted diffusion of proteins, thereby minimizing potential side effects and circumventing the limitations inherent in traditional techniques like electroporation. This review examines established protein drug delivery systems, including those based on polymers, liposomes, and protein nanoparticles. We delve into the operational principles and transport mechanisms of nanocarriers, discussing the various considerations essential for designing cutting-edge delivery platforms. Additionally, we investigate innovative designs and applications of traditional cytosolic protein delivery systems in medical research and clinical practice, particularly in areas like tumor treatment, gene editing and fluorescence imaging. This review sheds light on the current restrictions of protein delivery systems and anticipates future research avenues, aiming to foster the continued advancement in this field.
Collapse
Affiliation(s)
- Chuanda Zhu
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China
| | - Jing Mu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen, 518036, P.R. China.
| | - Ling Liang
- Department of Biophysics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, P.R. China.
| |
Collapse
|
2
|
Li Q, Yang X, Xia X, Xia XX, Yan D. Affibody-Functionalized Elastin-like Peptide-Drug Conjugate Nanomicelle for Targeted Ovarian Cancer Therapy. Biomacromolecules 2024; 25:6474-6484. [PMID: 39235966 DOI: 10.1021/acs.biomac.4c00640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
Recombinant elastin-like polypeptides (ELPs) have emerged as an attractive nanoplatform for drug delivery due to their tunable genetically encoded sequence, biocompatibility, and stimuli-responsive self-assembly behaviors. Here, we designed and biosynthesized an HER2 (human epidermal growth factor receptor 2)-targeted affibody-ELP fusion protein (Z-ELP), which was subsequently conjugated with monomethyl auristatin E (MMAE) to build a protein-drug conjugate (Z-ELP-M). Due to its thermal response, Z-ELP-M can immediately self-assemble into a nanomicelle at physiological temperature. Benefiting from its active targeting and nanomorphology, Z-ELP-M exhibits enhanced cellular internalization and deep tumor penetration in vitro. Moreover, Z-ELP-M shows excellent tumor targeting and superior antitumor efficacy in HER2-positive ovarian cancer, demonstrating a relative tumor growth inhibition of 104.6%. These findings suggest that an affibody-functionalized elastin-like peptide-drug conjugate nanomicelle is an efficient strategy to improve antitumor efficacy and biosafety in cancer therapy.
Collapse
Affiliation(s)
- Qingrong Li
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xiaoyuan Yang
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xuelin Xia
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Xiao-Xia Xia
- State Key Laboratory of Microbial Metabolism, Joint International Research Laboratory of Metabolic & Developmental Sciences, and School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| | - Deyue Yan
- School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, People's Republic of China
| |
Collapse
|
3
|
Gusliakova OI, Kurochkin MA, Barmin RA, Prikhozhdenko ES, Estifeeva TM, Rudakovskaya PG, Sindeeva OA, Galushka VV, Vavaev ES, Komlev AS, Lyubin EV, Fedyanin AA, Dey KK, Gorin DA. Magnetically navigated microbubbles coated with albumin/polyarginine and superparamagnetic iron oxide nanoparticles. BIOMATERIALS ADVANCES 2024; 158:213759. [PMID: 38227987 DOI: 10.1016/j.bioadv.2024.213759] [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: 09/22/2023] [Revised: 12/31/2023] [Accepted: 01/01/2024] [Indexed: 01/18/2024]
Abstract
While microbubbles (MB) are routinely used for ultrasound (US) imaging, magnetic MB are increasingly explored as they can be guided to specific sites of interest by applied magnetic field gradient. This requires the MB shell composition tuning to prolong MB stability and provide functionalization capabilities with magnetic nanoparticles. Hence, we developed air-filled MB stabilized by a protein-polymer complex of bovine serum albumin (BSA) and poly-L-arginine (pArg) of different molecular weights, showing that pArg of moderate molecular weight distribution (15-70 kDa) enabled MB with greater stability and acoustic response while preserving MB narrow diameters and the relative viability of THP-1 cells after 48 h of incubation. After MB functionalization with superparamagnetic iron oxide nanoparticles (SPION), magnetic moment values provided by single MB confirmed the sufficient SPION deposition onto BSA + pArg MB shells. During MB magnetic navigation in a blood vessel mimicking phantom with magnetic tweezers and in a Petri dish with adherent mouse renal carcinoma cell line, we demonstrated the effectiveness of magnetic MB localization in the desired area by magnetic field gradient. Magnetic MB co-localization with cells was further exploited for effective doxorubicin delivery with drug-loaded MB. Taken together, these findings open new avenues in control over albumin MB properties and magnetic navigation of SPION-loaded MB, which can envisage their applications in diagnostic and therapeutic needs.
Collapse
Affiliation(s)
- Olga I Gusliakova
- Science Medical Center, Saratov State University, Saratov 410012, Russia; Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.
| | - Maxim A Kurochkin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Roman A Barmin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | | | - Tatyana M Estifeeva
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Polina G Rudakovskaya
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Olga A Sindeeva
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow 121205, Russia
| | - Victor V Galushka
- Education and Research Institute of Nanostructures and Biosystems, Saratov State University, Saratov 410012, Russia
| | - Evgeny S Vavaev
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Aleksei S Komlev
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Evgeny V Lyubin
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Andrey A Fedyanin
- Faculty of Physics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Krishna Kanti Dey
- Department of Physics, Indian Institute of Technology Gandhinagar, Palaj, Gujarat 382055, India
| | - Dmitry A Gorin
- Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology, Moscow 121205, Russia.
| |
Collapse
|
4
|
Gonzalez-Valdivieso J, Vallejo R, Rodriguez-Rojo S, Santos M, Schneider J, Arias FJ, Girotti A. CD44-targeted nanoparticles for co-delivery of docetaxel and an Akt inhibitor against colorectal cancer. BIOMATERIALS ADVANCES 2023; 154:213595. [PMID: 37639856 DOI: 10.1016/j.bioadv.2023.213595] [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: 02/20/2023] [Revised: 07/24/2023] [Accepted: 08/19/2023] [Indexed: 08/31/2023]
Abstract
New strategies to develop drug-loaded nanocarriers with improved therapeutic efficacy are needed for cancer treatment. Herein we report a novel drug-delivery nanosystem comprising encapsulation of the chemotherapeutic drug docetaxel (DTX) and recombinant fusion of a small peptide inhibitor of Akt kinase within an elastin-like recombinamer (ELR) vehicle. This combined approach is also precisely targeted to colorectal cancer cells by means of a chemically conjugated DNA aptamer specific for the CD44 tumor marker. This 53 nm dual-approach nanosystem was found to selectively affect cell viability (2.5 % survival) and proliferation of colorectal cancer cells in vitro compared to endothelial cells (50 % survival), and to trigger both apoptosis- and necrosis-mediated cell death. Our findings also show that the nanohybrid particles remain stable under physiological conditions, trigger sustained drug release and possess an adequate pharmacokinetic profile after systemic intravenous administration. In vivo assays showed that these dual-approach nanohybrids significantly reduced the number of tumor polyps along the colorectal tract in a murine colorectal cancer model. Furthermore, systemic administration of advanced nanohybrids induced tissue recovery by improving the morphology of gastrointestinal crypts and the tissue architecture. Taken together, these findings indicate that our strategy of an advanced dual-approach nanosystem allows us to achieve successful controlled release of chemotherapeutics in cancer cells and may have a promising potential for colorectal cancer treatment.
Collapse
Affiliation(s)
- Juan Gonzalez-Valdivieso
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain
| | - Reinaldo Vallejo
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; BioEcoUVa, Research Institute on Bioeconomy, High Pressure Process Group, University of Valladolid, Department of Chemical Engineering and Environmental Technology, Escuela de Ingenierías Industriales, Sede Mergelina, Valladolid, Spain
| | - Soraya Rodriguez-Rojo
- BioEcoUVa, Research Institute on Bioeconomy, High Pressure Process Group, University of Valladolid, Department of Chemical Engineering and Environmental Technology, Escuela de Ingenierías Industriales, Sede Mergelina, Valladolid, Spain
| | - Mercedes Santos
- BIOFORGE Research Group (Group for Advanced Materials and Nanobiotechnology), University of Valladolid, CIBER-BBN, LUCIA Building, Valladolid, Spain
| | - Jose Schneider
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Department of Obstetrics & Gynecology, University of Valladolid, School of Medicine, Valladolid, Spain
| | - Francisco Javier Arias
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Unidad de excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), University of Valladolid CSIC, Valladolid, Spain.
| | - Alessandra Girotti
- Smart Devices for NanoMedicine Group, University of Valladolid, LUCIA Building, Valladolid, Spain; Unidad de excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM), University of Valladolid CSIC, Valladolid, Spain.
| |
Collapse
|
5
|
Hwang J, Huang H, Sullivan MO, Kiick KL. Controlled Delivery of Vancomycin from Collagen-tethered Peptide Vehicles for the Treatment of Wound Infections. Mol Pharm 2023; 20:1696-1708. [PMID: 36707500 PMCID: PMC10197141 DOI: 10.1021/acs.molpharmaceut.2c00898] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Despite the great promise of antibiotic therapy in wound infections, antibiotic resistance stemming from frequent dosing diminishes drug efficacy and contributes to recurrent infection. To identify improvements in antibiotic therapies, new antibiotic delivery systems that maximize pharmacological activity and minimize side effects are needed. In this study, we developed elastin-like peptide and collagen-like peptide nanovesicles (ECnVs) tethered to collagen-containing matrices to control vancomycin delivery and provide extended antibacterial effects against methicillin-resistant Staphylococcus aureus (MRSA). We observed that ECnVs showed enhanced entrapment efficacy of vancomycin by 3-fold as compared to liposome formulations. Additionally, ECnVs enabled the controlled release of vancomycin at a constant rate with zero-order kinetics, whereas liposomes exhibited first-order release kinetics. Moreover, ECnVs could be retained on both collagen-fibrin (co-gel) matrices and collagen-only matrices, with differential retention on the two biomaterials resulting in different local concentrations of released vancomycin. Overall, the biphasic release profiles of vancomycin from ECnVs/co-gel and ECnVs/collagen more effectively inhibited the growth of MRSA for 18 and 24 h, respectively, even after repeated bacterial inoculation, as compared to matrices containing free vancomycin, which just delayed the growth of MRSA. Thus, this newly developed antibiotic delivery system exhibited distinct advantages for controlled vancomycin delivery and prolonged antibacterial activity relevant to the treatment of wound infections.
Collapse
Affiliation(s)
- Jeongmin Hwang
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19713, USA
| | - Haofu Huang
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Millicent O. Sullivan
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19713, USA
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, 19716, USA
| | - Kristi L. Kiick
- Department of Biomedical Engineering, University of Delaware, Newark, DE, 19713, USA
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, 19716, USA
| |
Collapse
|
6
|
Hou J, Li N, Zhang W, Zhang W. Exploring the impact of PEGylation on the cell-nanomicelle interactions by AFM-based single-molecule force spectroscopy and force tracing. Acta Biomater 2023; 157:310-320. [PMID: 36535567 DOI: 10.1016/j.actbio.2022.12.020] [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: 09/04/2022] [Revised: 11/15/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
PEGylation has been considered the gold standard method for the modification of various drug delivery systems since the last century. However, the impact of PEGylation on the dynamic interaction between drug carriers and cell membranes has not been quantitatively clarified. Herein, the cellular binding and receptor-mediated endocytosis of a model PEGylated polypeptide nanomicelle were systematically investigated at the single-particle level using AFM-based single-molecule force spectroscopy (SMFS) and force tracing. A self-assembled elastin-like polypeptide (ELP) nanomicelle, which is capable of cross-linking, gastrin-releasing peptide (GRP) modification, and PEGylation was prepared. The cross-linked ELP-based nanomicelles exhibited outstanding stability in a broad temperature range of 4-40 °C, which facilitate the drug loading, as well as our cell-nanomicelle study at the single particle level. The unbinding force between the cross-linked ELP-based nanomicelles and the GRP receptor (GRPR)-containing cell (PC-3) membranes was quantitatively measured by AFM-SMFS. It is found that the PEGylated GRP-displaying nanomicelles exhibit the highest unbinding force, indicating the enhanced specific binding effect of PEGylation. Furthermore, the receptor-mediated endocytosis of the cross-linked ELP-based nanomicelles was monitored with the help of force tracing based on AFM-SMFS. Our results show that PEGylation decreases the endocytic force, duration, and engulfment depth of the PEGylated GRP-displaying nanomicelles, but increases their endocytic velocity, which results from the elimination of non-specific interactions during endocytosis. These observations demonstrate the diverse and complex roles of PEGylation on the interaction of polypeptide nanomicelles to cell membranes and may shed light on the rational design of organic polymer-based drug delivery systems aiming for active and passive targeting strategies. STATEMENT OF SIGNIFICANCE: A self-assembled elastin-like polypeptide (ELP) nanomicelle, which can be easily cross-linked, gastrin-releasing peptide (GRP) modified, and PEGylated, is designed. The AFM-SMFS experiment shows that PEGylation can enhance specific binding of the nanomicelles to the receptors on cell membranes. The force tracing experiment indicates that PEGylation decreases the endocytic force as well as engulfment depth of the nanomicelles through the elimination of non-specific interactions. PEGylation can benefit the drug delivery systems aiming at active targeting, while might not be an ideal modification for drug carriers designed for passive targeting, whose cellular uptake mainly depends on non-specific interactions.
Collapse
Affiliation(s)
- Jue Hou
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Nan Li
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Wei Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, PR China; College of Chemistry, Jilin University, Changchun 130012, PR China.
| | - Wenke Zhang
- State Key Laboratory of Supramolecular Structure and Materials, Center for Supramolecular Chemical Biology, College of Chemistry, Jilin University, Changchun, 130012, PR China.
| |
Collapse
|
7
|
Pradhan R, Dey A, Taliyan R, Puri A, Kharavtekar S, Dubey SK. Recent Advances in Targeted Nanocarriers for the Management of Triple Negative Breast Cancer. Pharmaceutics 2023; 15:pharmaceutics15010246. [PMID: 36678877 PMCID: PMC9866847 DOI: 10.3390/pharmaceutics15010246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/01/2023] [Accepted: 01/04/2023] [Indexed: 01/13/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a life-threatening form of breast cancer which has been found to account for 15% of all the subtypes of breast cancer. Currently available treatments are significantly less effective in TNBC management because of several factors such as poor bioavailability, low specificity, multidrug resistance, poor cellular uptake, and unwanted side effects being the major ones. As a rapidly growing field, nano-therapeutics offers promising alternatives for breast cancer treatment. This platform provides a suitable pathway for crossing biological barriers and allowing sustained systemic circulation time and an improved pharmacokinetic profile of the drug. Apart from this, it also provides an optimized target-specific drug delivery system and improves drug accumulation in tumor cells. This review provides insights into the molecular mechanisms associated with the pathogenesis of TNBC, along with summarizing the conventional therapy and recent advances of different nano-carriers for the management of TNBC.
Collapse
Affiliation(s)
- Rajesh Pradhan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
| | - Anuradha Dey
- Medical Research, R&D Healthcare Division, Emami Ltd., Kolkata 700056, India
| | - Rajeev Taliyan
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
- Correspondence: (R.T.); (S.K.D.); Tel.: +91-6378-364-745 (R.T.); +91-8239-703-734 (S.K.D.)
| | - Anu Puri
- RNA Structure and Design Section, RNA Biology Laboratory (RBL), Center for Cancer Research, National Cancer Institute—Frederick, Frederick, MD 21702, USA
| | - Sanskruti Kharavtekar
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
| | - Sunil Kumar Dubey
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani 333031, India
- Medical Research, R&D Healthcare Division, Emami Ltd., Kolkata 700056, India
- Correspondence: (R.T.); (S.K.D.); Tel.: +91-6378-364-745 (R.T.); +91-8239-703-734 (S.K.D.)
| |
Collapse
|
8
|
Jiang A, Guan X, He L, Guan X. Engineered elastin-like polypeptides: An efficient platform for enhanced cancer treatment. Front Pharmacol 2023; 13:1113079. [PMID: 36699056 PMCID: PMC9868590 DOI: 10.3389/fphar.2022.1113079] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
Drug delivery systems (DDSs) have recently gained widespread attention for improving drug loading and delivery efficiency in treating many cancers. Elastin-like polypeptides (ELPs) are synthetic peptides derived from a precursor of elastin (tropoelastin), reserving similar structural and physicochemical properties. ELPs have gained a variety of applications in tissue engineering and cancer therapy due to their excellent biocompatibility, complete degradability, temperature-responsive property, controllable sequence and length, and precisely tuned structure and function. ELPs-based drug delivery systems can improve the pharmacokinetics and biodistribution of therapeutic reagents, leading to enhanced antitumor efficacy. In this review, we summarize the recent application of ELPs in cancer treatment, focusing on the delivery of functional peptides, therapeutic proteins, small molecule drugs, and photosensitizers.
Collapse
Affiliation(s)
- Aiguo Jiang
- Department of Respiratory Medicine, Taizhou University Affiliated Wenling Hospital, Taizhou University, Taizhou, China
| | - Xinqiang Guan
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Lianping He
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, China
| | - Xingang Guan
- Department of Respiratory Medicine, Taizhou University Affiliated Wenling Hospital, Taizhou University, Taizhou, China
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, China
| |
Collapse
|
9
|
Elastin-like polypeptide-based micelles as a promising platform in nanomedicine. J Control Release 2023; 353:713-726. [PMID: 36526018 DOI: 10.1016/j.jconrel.2022.12.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
New and improved nanomaterials are constantly being developed for biomedical purposes. Nanomaterials based on elastin-like polypeptides (ELPs) have increasingly shown potential over the past two decades. These polymers are artificial proteins of which the design is based on human tropoelastin. Due to this similarity, ELP-based nanomaterials are biodegradable and therefore well suited to drug delivery. The assembly of ELP molecules into nanoparticles spontaneously occurs at temperatures above a transition temperature (Tt). The ELP sequence influences both the Tt and the physicochemical properties of the assembled nanomaterial. Nanoparticles with desired properties can hence be designed by choosing the appropriate sequence. A promising class of ELP nanoparticles are micelles assembled from amphiphilic ELP diblock copolymers. Such micelles are generally uniform and well defined. Furthermore, site-specific attachment of cargo to the hydrophobic block results in micelles with the cargo shielded inside their core, while conjugation to the hydrophilic block causes the cargo to reside in the corona where it is available for interactions. Such control over particle design is one of the main contributing factors for the potential of ELP-based micelles as a drug delivery system. Additionally, the micelles are easily loaded with protein or peptide-based cargo by expressing it as a fusion protein. Small molecule drugs and other cargo types can be either covalently conjugated to ELP domains or physically entrapped inside the micelle core. This review aims to give an overview of ELP-based micelles and their applications in nanomedicine.
Collapse
|
10
|
The application of elastin-like peptides in cancer, tissue engineering and ocular disease. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
11
|
Levêque M, Xiao Y, Durand L, Massé L, Garanger E, Lecommandoux S. Aqueous synthesis and self-assembly of bioactive and thermo-responsive HA- b-ELP bioconjugates. Biomater Sci 2022; 10:6365-6376. [PMID: 36168976 DOI: 10.1039/d2bm01149b] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The design of synthetic (bio)macromolecules that combine biocompatibility, self-assembly and bioactivity properties at the molecular level is an intense field of research for biomedical applications such as (nano)medicine. In this contribution, we have designed and synthesized a library of bioactive and thermo-responsive bioconjugates from elastin-like polypeptides (ELPs) and hyaluronic acid (HA) in order to access bioactive self-assembled nanoparticles. These were prepared by a simple synthetic and purification strategy, compatible with the requirements for biological applications and industrial scale-up. A series of 9 HA-b-ELP bioconjugates with different compositions and block lengths was synthesized under aqueous conditions by strain-promoted azide-alkyne cycloaddition (SPAAC), avoiding the use of catalysts, co-reactants and organic solvents, and isolated by a simple centrifugation step. An extensive physico-chemical study was then performed on the whole library of bioconjugates in an attempt to establish structure-property relationships. In particular, the determination of the critical conditions for thermally driven self-assembly was carried out upon temperature (CMT) and concentration (CMC) gradients, leading to a phase diagram for each of these bioconjugates. These parameters and the size of nanoparticles were found to depend on the chemical composition of the bioconjugates, namely on the respective size of individual blocks. Understanding the mechanism underlying this dependency is a real asset for designing more effective experiments: with key criteria defined (e.g. concentration, temperature, salinity, and biological target), the composition of the best candidates can be rationalized. In particular, four of the bioconjugates (HA4.6k-ELPn80 or n100 and HA24k-ELPn80 or n100) were found to self-assemble into well-defined spherical core-shell nanoparticles, with a negative surface charge due to the HA block exposed at the surface, a hydrodynamic diameter between 40 and 200 nm under physiological conditions and a good stability over time at 37 °C. We therefore propose here a versatile and simple design of smart, controllable, and bioactive nanoparticles that present different behaviors depending on the diblocks' composition.
Collapse
Affiliation(s)
- Manon Levêque
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France.
| | - Ye Xiao
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France.
| | - Laura Durand
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France.
| | - Louise Massé
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France.
| | - Elisabeth Garanger
- Université de Bordeaux, CNRS, Bordeaux INP, LCPO, UMR 5629, Pessac F-33600, France.
| | | |
Collapse
|
12
|
Avila H, Yu J, Boddu G, Phan A, Truong A, Peddi S, Guo H, Lee SJ, Alba M, Canfield E, Yamamoto V, Paton JC, Paton AW, Lee AS, MacKay JA. Hydra-Elastin-like Polypeptides Increase Rapamycin Potency When Targeting Cell Surface GRP78. Biomacromolecules 2022; 23:3116-3129. [PMID: 35786858 PMCID: PMC10231879 DOI: 10.1021/acs.biomac.2c00048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Rapalogues are powerful therapeutic modalities for breast cancer; however, they suffer from low solubility and dose-limiting side effects. To overcome these challenges, we developed a long-circulating multiheaded drug carrier called 5FA, which contains rapamycin-binding domains linked with elastin-like polypeptides (ELPs). To target these "Hydra-ELPs" toward breast cancer, we here linked 5FA with four distinct peptides which are reported to engage the cell surface form of the 78 kDa glucose-regulated protein (csGRP78). To determine if these peptides affected the carrier solubility, this library was characterized by light scattering and mass spectrometry. To guide in vitro selection of the most potent functional carrier for rapamycin, its uptake and inhibition of mTORC1 were monitored in a ductal breast cancer model (BT474). Using flow cytometry to track cellular association, it was found that only the targeted carriers enhanced cellular uptake and were susceptible to proteolysis by SubA, which specifically targets csGRP78. The functional inhibition of mTOR was monitored by Western blot for pS6K, whereby the best carrier L-5FA reduced mTOR activity by 3-fold compared to 5FA or free rapamycin. L-5FA was further visualized using super-resolution confocal laser scanning microscopy, which revealed that targeting increased exposure to the carrier by ∼8-fold. This study demonstrates how peptide ligands for GRP78, such as the L peptide (RLLDTNRPLLPY), may be incorporated into protein-based drug carriers to enhance targeting.
Collapse
Affiliation(s)
- Hugo Avila
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Jingmei Yu
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Geetha Boddu
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Alvin Phan
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Anh Truong
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Santosh Peddi
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Hao Guo
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Shin-Jae Lee
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
- Department of Biomedical Engineering, USC Viterbi School of Engineering, Los Angeles, California 90089, United States
| | - Mario Alba
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Ethan Canfield
- Mass Spectrometry Core, USC School of Pharmacy, Los Angeles, California 90089, United States
| | - Vicky Yamamoto
- Department of Biochemistry and Molecular Medicine, USC Keck School of Medicine, Los Angeles, California 90033, United States
| | - James C Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide 5005, Australia
| | - Adrienne W Paton
- Research Centre for Infectious Diseases, Department of Molecular and Biomedical Science, University of Adelaide, Adelaide 5005, Australia
| | - Amy S Lee
- Department of Biochemistry and Molecular Medicine, USC Keck School of Medicine, Los Angeles, California 90033, United States
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California 90089, United States
- Department of Biomedical Engineering, USC Viterbi School of Engineering, Los Angeles, California 90089, United States
- Department of Ophthalmology, USC Keck School of Medicine, Los Angeles, California 90033, United States
| |
Collapse
|
13
|
Chae YK, Um Y, Kim H. A simple and sensitive detection of the binding ligands by using the receptor aggregation and NMR spectroscopy: a test case of the maltose binding protein. JOURNAL OF BIOMOLECULAR NMR 2021; 75:371-381. [PMID: 34524563 PMCID: PMC8441238 DOI: 10.1007/s10858-021-00381-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/02/2021] [Indexed: 06/13/2023]
Abstract
Protein-ligand interaction is one of the highlights of molecular recognition. The most popular application of this type of interaction is drug development which requires a high throughput screening of a ligand that binds to the target protein. Our goal was to find a binding ligand with a simple detection, and once this type of ligand was found, other methods could then be used to measure the detailed kinetic or thermodynamic parameters. We started with the idea that the ligand NMR signal would disappear if it was bound to the non-tumbling mass. In order to create the non-tumbling mass, we tried the aggregates of a target protein, which was fused to the elastin-like polypeptide. We chose the maltose binding proteinas a test case, and we tried it with several sugars, which included maltose, glucose, sucrose, lactose, galactose, maltotriose, and β-cyclodextrin. The maltose signal in the H-1 NMR spectrum disappeared completely as hoped around the protein to ligand ratio of 1:3 at 298 K where the proteins aggregated. The protein signals also disappeared upon aggregation except for the fast-moving part, which resulted in a cleaner background than the monomeric form. Since we only needed to look for a disappearing signal amongst those from the mixture, it should be useful in high throughput screening. Other types of sugars except for the maltotriose and β-cyclodextrin, which are siblings of the maltose, did not seem to bind at all. We believe that our system would be especially more effective when dealing with a smaller target protein, so both the protein and the bound ligand would lose their signals only when the aggregates formed. We hope that our proposed method would contribute to accelerating the development of the potent drug candidates by simultaneously identifying several binders directly from a mixture.
Collapse
Affiliation(s)
- Young Kee Chae
- Department of Chemistry, Sejong University, 209 Neungdong-Ro, Gwangjin-Gu, Seoul, 05006, Korea.
| | - Yoonjin Um
- Department of Chemistry, Sejong University, 209 Neungdong-Ro, Gwangjin-Gu, Seoul, 05006, Korea
| | - Hakbeom Kim
- Department of Chemistry, Sejong University, 209 Neungdong-Ro, Gwangjin-Gu, Seoul, 05006, Korea
| |
Collapse
|
14
|
Gonzalez-Valdivieso J, Garcia-Sampedro A, Hall AR, Girotti A, Arias FJ, Pereira SP, Acedo P. Smart Nanoparticles as Advanced Anti-Akt Kinase Delivery Systems for Pancreatic Cancer Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:55790-55805. [PMID: 34788541 DOI: 10.1021/acsami.1c14592] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Pancreatic cancer is one of the deadliest cancers partly due to late diagnosis, poor drug delivery to the target site, and acquired resistance to therapy. Therefore, more effective therapies are urgently needed to improve the outcome of patients. In this work, we have tested self-assembling genetically engineered polymeric nanoparticles formed by elastin-like recombinamers (ELRs), carrying a small peptide inhibitor of the protein kinase Akt, in both PANC-1 and patient-derived pancreatic cancer cells (PDX models). Nanoparticle cell uptake was measured by flow cytometry, and subcellular localization was determined by confocal microscopy, which showed a lysosomal localization of these nanoparticles. Furthermore, metabolic activity and cell viability were significantly reduced after incubation with nanoparticles carrying the Akt inhibitor in a time- and dose-dependent fashion. Self-assembling 73 ± 3.2 nm size nanoparticles inhibited phosphorylation and consequent activation of Akt protein, blocked the NF-κB signaling pathway, and triggered caspase 3-mediated apoptosis. Furthermore, in vivo assays showed that ELR-based nanoparticles were suitable devices for drug delivery purposes with long circulating time and minimum toxicity. Hence, the use of these smart nanoparticles could lead to the development of more effective treatment options for pancreatic cancer based on the inhibition of Akt.
Collapse
Affiliation(s)
- Juan Gonzalez-Valdivieso
- Smart Biodevices for NanoMed Group, University of Valladolid, Paseo Belén, Valladolid 47011, Spain
| | - Andres Garcia-Sampedro
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Andrew R Hall
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
- Sheila Sherlock Liver Centre, Royal Free London NHS Foundation Trust, London NW3 2QG, United Kingdom
| | - Alessandra Girotti
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, University of Valladolid, Paseo Belén, Valladolid 47011, Spain
| | - Francisco Javier Arias
- Smart Biodevices for NanoMed Group, University of Valladolid, Paseo Belén, Valladolid 47011, Spain
| | - Stephen P Pereira
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| | - Pilar Acedo
- Institute for Liver and Digestive Health, Royal Free Hospital Campus, University College London, Pond Street, London NW3 2QG, United Kingdom
| |
Collapse
|
15
|
Bidwell GL. Novel Protein Therapeutics Created Using the Elastin-Like Polypeptide Platform. Physiology (Bethesda) 2021; 36:367-381. [PMID: 34486397 DOI: 10.1152/physiol.00026.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Elastin-like polypeptides (ELPs) are bioengineered proteins that have a unique physical property, a thermally triggered inverse phase transition, that can be exploited for drug delivery. ELP-fusion proteins can be used as soluble biologics, thermally targeted drug carriers, self-assembling nanoparticles, and slow-release drug depots. Because of their unique physical characteristics and versatility for delivery of nearly any type of therapeutic, ELP-based drug delivery systems represent a promising platform for biologics development.
Collapse
Affiliation(s)
- Gene L Bidwell
- Departments of Neurology, Cell and Molecular Biology, and Pharmacology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
16
|
Chowdhury P, Ghosh U, Samanta K, Jaggi M, Chauhan SC, Yallapu MM. Bioactive nanotherapeutic trends to combat triple negative breast cancer. Bioact Mater 2021; 6:3269-3287. [PMID: 33778204 PMCID: PMC7970221 DOI: 10.1016/j.bioactmat.2021.02.037] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/27/2021] [Accepted: 02/28/2021] [Indexed: 02/09/2023] Open
Abstract
The management of aggressive breast cancer, particularly, triple negative breast cancer (TNBC) remains a formidable challenge, despite treatment advancement. Although newer therapies such as atezolizumab, olaparib, and sacituzumab can tackle the breast cancer prognosis and/or progression, but achieved limited survival benefit(s). The current research efforts are aimed to develop and implement strategies for improved bioavailability, targetability, reduce systemic toxicity, and enhance therapeutic outcome of FDA-approved treatment regimen. This review presents various nanoparticle technology mediated delivery of chemotherapeutic agent(s) for breast cancer treatment. This article also documents novel strategies to employ cellular and cell membrane cloaked (biomimetic) nanoparticles for effective clinical translation. These technologies offer a safe and active targeting nanomedicine for effective management of breast cancer, especially TNBC.
Collapse
Affiliation(s)
- Pallabita Chowdhury
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Upasana Ghosh
- Department of Biomedical Engineering, School of Engineering, Rutgers University, The State University of New Jersey, Piscataway, NJ, 08854, USA
| | - Kamalika Samanta
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Meena Jaggi
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C. Chauhan
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M. Yallapu
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| |
Collapse
|
17
|
Jenkins IC, Milligan JJ, Chilkoti A. Genetically Encoded Elastin-Like Polypeptides for Drug Delivery. Adv Healthc Mater 2021; 10:e2100209. [PMID: 34080796 DOI: 10.1002/adhm.202100209] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/14/2021] [Indexed: 12/19/2022]
Abstract
Elastin-like polypeptides (ELPs) are thermally responsive biopolymers that consist of a repeated amino acid motif derived from human tropoelastin. These peptides exhibit temperature-dependent phase behavior that can be harnessed to produce stimuli-responsive biomaterials, such as nanoparticles or injectable drug delivery depots. As ELPs are genetically encoded, the properties of ELP-based biomaterials can be controlled with a precision that is unattainable with synthetic polymers. Unique ELP architectures, such as spherical or rod-like micelles or injectable coacervates, can be designed by manipulating the ELP amino acid sequence and length. ELPs can be loaded with drugs to create controlled, intelligent drug delivery systems. ELPs are biodegradable, nonimmunogenic, and tolerant of therapeutic additives. These qualities make ELPs exquisitely well-suited to address current challenges in drug delivery and have spurred the development of ELP-based therapeutics to treat diseases-such as cancer and diabetes-and to promote wound healing. This review focuses on the use of ELPs in drug delivery systems.
Collapse
Affiliation(s)
- Irene C. Jenkins
- Department of Biomedical Engineering Duke University Durham NC 277018 USA
| | - Joshua J. Milligan
- Department of Biomedical Engineering Duke University Durham NC 277018 USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering Duke University Durham NC 277018 USA
| |
Collapse
|
18
|
Romero EL, Morilla MJ. Preclinical autophagy modulatory nanomedicines: big challenges, slow advances. Expert Opin Drug Deliv 2021; 18:1415-1434. [PMID: 34030559 DOI: 10.1080/17425247.2021.1933428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Autophagy is a critical housekeeping pathway to remove toxic protein aggregates, damaged organelles, providing cells with bioenergetic substrates needed to survive under adverse conditions. Since altered autophagy is associated with diverse diseases, its pharmacological modulation is considered of therapeutic interest. Nanomedicines may reduce the toxicity and improve the activity of toxic autophagy modulatory drugs (amd). AREAS COVERED The status of the most relevant anti-tumor, anti-inflammatory, and anti-infectious treatments mediated by autophagy modulatory nanomedicines (amN) published in the last 5 years is discussed. EXPERT OPINION Antitumor and anti-inflammatory treatments may be improved by administering amN for selective, massive, and targeted delivery of amd to diseased tissues. The use of amN as antimicrobial agent remains almost underexploited. Assessing the effect of amN on the complex autophagy machinery operating under different basal diseases, however, is not a trivial task. Besides structural reproducibility, nanomedicines must grant higher efficiency, and lower adverse effects than conventional medication. Simplicity of design, carefully chosen (scalable) preparation techniques, and rigorous monitoring of preclinical efficacy and nanotoxicity will improve the chances of clinical success. Currently, available data are not sufficient to envisage a fast-succeeding translation. Application of quality by design criteria would help to reach such milestones.
Collapse
Affiliation(s)
- Eder Lilia Romero
- Department of Science and Technology, Nanomedicines Research and Development Center, Quilmes National University, Bernal, Buenos Aires, Argentina
| | - Maria Jose Morilla
- Department of Science and Technology, Nanomedicines Research and Development Center, Quilmes National University, Bernal, Buenos Aires, Argentina
| |
Collapse
|
19
|
Alvisi N, Gutiérrez-Mejía FA, Lokker M, Lin YT, de Jong AM, van Delft F, de Vries R. Self-Assembly of Elastin-like Polypeptide Brushes on Silica Surfaces and Nanoparticles. Biomacromolecules 2021; 22:1966-1979. [PMID: 33871996 PMCID: PMC8154268 DOI: 10.1021/acs.biomac.1c00067] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Control over the placement and activity of biomolecules on solid surfaces is a key challenge in bionanotechnology. While covalent approaches excel in performance, physical attachment approaches excel in ease of processing, which is equally important in many applications. We show how the precision of recombinant protein engineering can be harnessed to design and produce protein-based diblock polymers with a silica-binding and highly hydrophilic elastin-like domain that self-assembles on silica surfaces and nanoparticles to form stable polypeptide brushes that can be used as a scaffold for later biofunctionalization. From atomic force microscopy-based single-molecule force spectroscopy, we find that individual silica-binding peptides have high unbinding rates. Nevertheless, from quartz crystal microbalance measurements, we find that the self-assembled polypeptide brushes cannot easily be rinsed off. From atomic force microscopy imaging and bulk dynamic light scattering, we find that the binding to silica induces fibrillar self-assembly of the peptides. Hence, we conclude that the unexpected stability of these self-assembled polypeptide brushes is at least in part due to peptide-peptide interactions of the silica-binding blocks at the silica surface.
Collapse
Affiliation(s)
- Nicolò Alvisi
- Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, Wageningen 6708 WE, The Netherlands
| | - Fabiola A Gutiérrez-Mejía
- Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, Wageningen 6708 WE, The Netherlands
| | - Meike Lokker
- Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, Wageningen 6708 WE, The Netherlands
| | - Yu-Ting Lin
- Department of Applied Physics and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven 5600 MB, The Netherlands
| | - Arthur M de Jong
- Department of Applied Physics and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven 5600 MB, The Netherlands
| | - Floris van Delft
- Laboratory of Organic Chemistry, Wageningen University and Research, Stippeneng 4, Wageningen 6708 WE, The Netherlands
| | - Renko de Vries
- Physical Chemistry and Soft Matter, Wageningen University and Research, Stippeneng 4, Wageningen 6708 WE, The Netherlands
| |
Collapse
|
20
|
Séhédic D, Roncali L, Djoudi A, Buchtova N, Avril S, Chérel M, Boury F, Lacoeuille F, Hindré F, Garcion E. Rapamycin-Loaded Lipid Nanocapsules Induce Selective Inhibition of the mTORC1-Signaling Pathway in Glioblastoma Cells. Front Bioeng Biotechnol 2021; 8:602998. [PMID: 33718332 PMCID: PMC7947795 DOI: 10.3389/fbioe.2020.602998] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 12/29/2020] [Indexed: 11/21/2022] Open
Abstract
Inhibition of the PI3K/Akt/mTOR signaling pathway represents a potential issue for the treatment of cancer, including glioblastoma. As such, rapamycin that inhibits the mechanistic target of rapamycin (mTOR), the downstream effector of this signaling pathway, is of great interest. However, clinical development of rapamycin has floundered due to the lack of a suitable formulation of delivery systems. In the present study, a novel method for the formulation of safe rapamycin nanocarriers is investigated. A phase inversion process was adapted to prepare lipid nanocapsules (LNCs) loaded with the lipophilic and temperature sensitive rapamycin. Rapamycin-loaded LNCs (LNC-rapa) are ~110 nm in diameter with a low polydispersity index (<0.05) and the zeta potential of about −5 mV. The encapsulation efficiency, determined by spectrophotometry conjugated with filtration/exclusion, was found to be about 69%, which represents 0.6 wt% of loading capacity. Western blot analysis showed that LNC-rapa do not act synergistically with X-ray beam radiation in U87MG glioblastoma model in vitro. Nevertheless, it demonstrated the selective inhibition of the phosphorylation of mTORC1 signaling pathway on Ser2448 at a concentration of 1 μM rapamycin in serum-free medium. Interestingly, cells cultivated in normoxia (21% O2) seem to be more sensitive to mTOR inhibition by rapamycin than those cultivated in hypoxia (0.4% O2). Finally, we also established that mTOR phosphorylation inhibition by LNC-rapa induced a negative feedback through the activation of Akt phosphorylation. This phenomenon was more noticeable after stabilization of HIF-1α in hypoxia.
Collapse
Affiliation(s)
- Delphine Séhédic
- Univ Angers, Université de Nantes, Inserm, CRCINA, SFR ICAT, Angers, France
| | - Loris Roncali
- Univ Angers, Université de Nantes, Inserm, CRCINA, SFR ICAT, Angers, France
| | - Amel Djoudi
- Univ Angers, Université de Nantes, Inserm, CRCINA, SFR ICAT, Angers, France
| | - Nela Buchtova
- Univ Angers, Université de Nantes, Inserm, CRCINA, SFR ICAT, Angers, France
| | - Sylvie Avril
- Univ Angers, Université de Nantes, Inserm, CRCINA, SFR ICAT, Angers, France
| | - Michel Chérel
- Université de Nantes, Inserm, CNRS, CRCINA, Nantes, France
| | - Frank Boury
- Univ Angers, Université de Nantes, Inserm, CRCINA, SFR ICAT, Angers, France
| | - Franck Lacoeuille
- Univ Angers, Université de Nantes, Inserm, CRCINA, SFR ICAT, Angers, France
| | - François Hindré
- Univ Angers, Université de Nantes, Inserm, CRCINA, SFR ICAT, Angers, France
| | - Emmanuel Garcion
- Univ Angers, Université de Nantes, Inserm, CRCINA, SFR ICAT, Angers, France
| |
Collapse
|
21
|
Georgilis E, Abdelghani M, Pille J, Aydinlioglu E, van Hest JC, Lecommandoux S, Garanger E. Nanoparticles based on natural, engineered or synthetic proteins and polypeptides for drug delivery applications. Int J Pharm 2020; 586:119537. [DOI: 10.1016/j.ijpharm.2020.119537] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 06/03/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022]
|
22
|
Abstract
Elastin-like polypeptides (ELPs) are stimulus-responsive biopolymers derived from human elastin. Their unique properties—including lower critical solution temperature phase behavior and minimal immunogenicity—make them attractive materials for a variety of biomedical applications. ELPs also benefit from recombinant synthesis and genetically encoded design; these enable control over the molecular weight and precise incorporation of peptides and pharmacological agents into the sequence. Because their size and sequence are defined, ELPs benefit from exquisite control over their structure and function, qualities that cannot be matched by synthetic polymers. As such, ELPs have been engineered to assemble into unique architectures and display bioactive agents for a variety of applications. This review discusses the design and representative biomedical applications of ELPs, focusing primarily on their use in tissue engineering and drug delivery.
Collapse
Affiliation(s)
- Anastasia K. Varanko
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Jonathan C. Su
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, USA
| |
Collapse
|
23
|
Hsueh PY, Ju Y, Vega A, Edman MC, MacKay JA, Hamm-Alvarez SF. A Multivalent ICAM-1 Binding Nanoparticle which Inhibits ICAM-1 and LFA-1 Interaction Represents a New Tool for the Investigation of Autoimmune-Mediated Dry Eye. Int J Mol Sci 2020; 21:ijms21082758. [PMID: 32326657 PMCID: PMC7216292 DOI: 10.3390/ijms21082758] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/11/2020] [Accepted: 04/12/2020] [Indexed: 12/31/2022] Open
Abstract
The autoimmune disorder, Sjögren’s syndrome (SS), is characterized by lymphocytic infiltration and loss of function of exocrine glands such as the lacrimal gland (LG) and salivary gland. SS-associated changes in the LG are associated with the development of autoimmune-mediated dry eye disease. We have previously reported the accumulation of intercellular adhesion molecule 1 (ICAM-1) in the LG of Non-Obese Diabetic (NOD) mice, a murine model of autoimmune-mediated dry eye in SS, in both LG acinar cells and infiltrating lymphocytes. ICAM-1 initiates T-cell activation and can trigger T-cell migration through binding to lymphocyte function-associated 1 antigen (LFA). To modulate this interaction, this study introduces a new tool, a multivalent biopolymeric nanoparticle assembled from a diblock elastin-like polypeptide (ELP) using the S48I48 (SI) ELP scaffold fused with a mouse ICAM-1 targeting peptide to form IBP-SI. IBP-SI forms a multivalent, monodisperse nanoparticle with a radius of 21.9 nm. Unlike the parent SI, IBP-SI binds mouse ICAM-1 and is internalized by endocytosis into transfected HeLa cells before it accumulates in lysosomes. In vitro assays measuring lymphocyte adhesion to Tumor Necrosis Factor TNF-α-treated bEnd.3 cells, which express high levels of ICAM-1, show that adhesion is inhibited by IBP-SI but not by SI, with IC50 values of 62.7 μM and 81.2 μM, respectively, in two different assay formats. IBP-SI, but not SI, also blocked T-cell proliferation in a mixed lymphocyte reaction by 74% relative to proliferation in an untreated mixed cell reaction. These data suggest that a biopolymeric nanoparticle with affinity for ICAM-1 can disrupt ICAM-1 and LFA interactions in vitro and may have further utility as an in vivo tool or potential therapeutic.
Collapse
Affiliation(s)
- Pang-Yu Hsueh
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; (P.-Y.H.); (Y.J.); (A.V.)
| | - Yaping Ju
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; (P.-Y.H.); (Y.J.); (A.V.)
| | - Adrianna Vega
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; (P.-Y.H.); (Y.J.); (A.V.)
| | - Maria C. Edman
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA 90033, USA;
| | - J. Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; (P.-Y.H.); (Y.J.); (A.V.)
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
- Correspondence: (J.A.M.); (S.F.H.-A.)
| | - Sarah F. Hamm-Alvarez
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90033, USA; (P.-Y.H.); (Y.J.); (A.V.)
- Department of Ophthalmology, USC Roski Eye Institute, University of Southern California, Los Angeles, CA 90033, USA;
- Correspondence: (J.A.M.); (S.F.H.-A.)
| |
Collapse
|
24
|
Peddi S, Roberts SK, MacKay JA. Nanotoxicology of an Elastin-like Polypeptide Rapamycin Formulation for Breast Cancer. Biomacromolecules 2020; 21:1091-1102. [PMID: 31927993 PMCID: PMC7219203 DOI: 10.1021/acs.biomac.9b01431] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The clinical utility of rapamycin (Rapa) is limited by solubility, bioavailability, and side effects. To overcome this, our team recently reported an elastin-like polypeptide (ELP) nanoparticle with high affinity, noncovalent drug binding, and integrin-mediated cellular uptake. Given the scarcity of pharmacology/toxicology studies of ELP-based drug carriers, this article explores safety and efficacy of ELP-Rapa. ELP-Rapa nanoparticles tested negative for hemolysis, did not interfere in plasma coagulation nor in platelet function, and did not activate the complement. Upon incubation with HepG2 cells, ELP-Rapa revealed significant cellular uptake and trafficking to acidic organelles, consistent with lysosomes. Internalized ELP-Rapa nanoparticles increased oxidative stress 4-fold compared to free drug or free ELP controls. However, mice bearing orthotopic hormone receptor positive BT-474 breast tumors, given a high dose (∼10-fold above therapeutic dose) of 1 month administration of ELP-Rapa, did not induce hepatotoxicity. On the other hand, tumor growth and mTOR signaling were suppressed without affecting body weight. Nanoparticles assembled using ELP technology appear to be a safe and efficient strategy for delivering Rapa.
Collapse
Affiliation(s)
- Santosh Peddi
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy at the University of Southern California, Los Angeles, California 90033-9121, United States
| | - S Kenny Roberts
- Eunoia Biotech LLC, Wynnewood, Pennsylvania 19096, United States
| | - John Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy at the University of Southern California, Los Angeles, California 90033-9121, United States
- Department of Biomedical Engineering, Viterbi School of Engineering at the University of Southern California, Los Angeles, California 90089, United States
- Department of Ophthalmology, Keck School of Medicine at the University of Southern California, Los Angeles, California 90089-9020, United States
| |
Collapse
|
25
|
Human plasma protein adsorption to elastin-like polypeptide nanoparticles. Biointerphases 2020; 15:021007. [DOI: 10.1116/6.0000027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
26
|
Santos M, Serrano-Dúcar S, González-Valdivieso J, Vallejo R, Girotti A, Cuadrado P, Arias FJ. Genetically Engineered Elastin-based Biomaterials for Biomedical Applications. Curr Med Chem 2020; 26:7117-7146. [PMID: 29737250 DOI: 10.2174/0929867325666180508094637] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/28/2018] [Accepted: 04/13/2018] [Indexed: 01/31/2023]
Abstract
Protein-based polymers are some of the most promising candidates for a new generation of innovative biomaterials as recent advances in genetic-engineering and biotechnological techniques mean that protein-based biomaterials can be designed and constructed with a higher degree of complexity and accuracy. Moreover, their sequences, which are derived from structural protein-based modules, can easily be modified to include bioactive motifs that improve their functions and material-host interactions, thereby satisfying fundamental biological requirements. The accuracy with which these advanced polypeptides can be produced, and their versatility, self-assembly behavior, stimuli-responsiveness and biocompatibility, means that they have attracted increasing attention for use in biomedical applications such as cell culture, tissue engineering, protein purification, surface engineering and controlled drug delivery. The biopolymers discussed in this review are elastin-derived protein-based polymers which are biologically inspired and biomimetic materials. This review will also focus on the design, synthesis and characterization of these genetically encoded polymers and their potential utility for controlled drug and gene delivery, as well as in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Mercedes Santos
- BIOFORGE Research Group, CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - Sofía Serrano-Dúcar
- BIOFORGE Research Group, CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | | | - Reinaldo Vallejo
- BIOFORGE Research Group, CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - Alessandra Girotti
- BIOFORGE Research Group, CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | - Purificación Cuadrado
- BIOFORGE Research Group, CIBER-BBN, University of Valladolid, 47011 Valladolid, Spain
| | | |
Collapse
|
27
|
Varanko A, Saha S, Chilkoti A. Recent trends in protein and peptide-based biomaterials for advanced drug delivery. Adv Drug Deliv Rev 2020; 156:133-187. [PMID: 32871201 PMCID: PMC7456198 DOI: 10.1016/j.addr.2020.08.008] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/14/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023]
Abstract
Engineering protein and peptide-based materials for drug delivery applications has gained momentum due to their biochemical and biophysical properties over synthetic materials, including biocompatibility, ease of synthesis and purification, tunability, scalability, and lack of toxicity. These biomolecules have been used to develop a host of drug delivery platforms, such as peptide- and protein-drug conjugates, injectable particles, and drug depots to deliver small molecule drugs, therapeutic proteins, and nucleic acids. In this review, we discuss progress in engineering the architecture and biological functions of peptide-based biomaterials -naturally derived, chemically synthesized and recombinant- with a focus on the molecular features that modulate their structure-function relationships for drug delivery.
Collapse
Affiliation(s)
| | | | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, USA.
| |
Collapse
|
28
|
Wang Z, Guo J, Sun J, Liang P, Wei Y, Deng X, Gao W. Thermoresponsive and Protease-Cleavable Interferon-Polypeptide Conjugates with Spatiotemporally Programmed Two-Step Release Kinetics for Tumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1900586. [PMID: 31453069 PMCID: PMC6702759 DOI: 10.1002/advs.201900586] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/06/2019] [Indexed: 05/11/2023]
Abstract
Protein-polymer conjugates show improved pharmacokinetics but reduced bioactivity and tumor penetration as compared to native proteins, resulting in limited antitumor efficacy. To address this dilemma, genetic engineering of a body temperature-responsive and matrix metalloproteinase (MMP)-cleavable conjugate of interferon alpha (IFNα) and elastin-like polypeptide (ELP) is reported with spatiotemporally programmed two-step release kinetics for tumor therapy. Notably, the conjugate could phase separate to form a depot postsubcutaneous injection, leading to 1-month zero-order release kinetics. Furthermore, it could selectively be cleaved by MMPs that are overexpressed in tumors to release IFNα from ELP and thus to recover the bioactivity of IFNα. Consequently, it exhibits dramatically enhanced tumor accumulation, tumor penetration, and antitumor efficacy as compared to free IFNα in two mouse models of melanoma and ovarian tumor. These findings may provide an intelligent technology of thermoresponsive and protease-cleavable protein-polymer conjugates with spatiotemporally programmed two-step release kinetics for tumor treatment.
Collapse
Affiliation(s)
- Zhuoran Wang
- Department of Biomedical EngineeringSchool of MedicineTsinghua UniversityBeijing100084P. R. China
| | - Jianwen Guo
- Department of Biomedical EngineeringSchool of MedicineTsinghua UniversityBeijing100084P. R. China
| | - Jiawei Sun
- Department of Biomedical EngineeringSchool of MedicineTsinghua UniversityBeijing100084P. R. China
| | - Ping Liang
- Department of NeurosurgeryBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijing102218P. R. China
| | - Yan Wei
- Department of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P.R. China
| | - Xuliang Deng
- Department of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Biomedical Engineering DepartmentPeking UniversityBeijing100191P. R. China
| | - Weiping Gao
- Department of Geriatric DentistryBeijing Laboratory of Biomedical MaterialsPeking University School and Hospital of StomatologyBeijing100081P. R. China
- Biomedical Engineering DepartmentPeking UniversityBeijing100191P. R. China
| |
Collapse
|
29
|
Katyal P, Meleties M, Montclare JK. Self-Assembled Protein- and Peptide-Based Nanomaterials. ACS Biomater Sci Eng 2019; 5:4132-4147. [PMID: 33417774 DOI: 10.1021/acsbiomaterials.9b00408] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Considerable effort has been devoted to generating novel protein- and peptide-based nanomaterials with their applications in a wide range of fields. Specifically, the unique property of proteins to self-assemble has been utilized to create a variety of nanoassemblies, which offer significant possibilities for next-generation biomaterials. In this minireview, we describe self-assembled protein- and peptide-based nanomaterials with focus on nanofibers and nanoparticles. Their applications in delivering therapeutic drugs and genes are discussed.
Collapse
Affiliation(s)
- Priya Katyal
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, Brooklyn, New York 11201, United States
| | - Michael Meleties
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, Brooklyn, New York 11201, United States
| | - Jin K Montclare
- Department of Chemical and Biomolecular Engineering, Tandon School of Engineering, New York University, Brooklyn, New York 11201, United States.,Department of Radiology, New York University Langone Health, New York, New York 10016, United States.,Department of Biomaterials, College of Dentistry, New York University, New York, New York 10010, United States.,Department of Chemistry, New York University, New York, New York 10003, United States
| |
Collapse
|
30
|
Rapamycin-loaded polysorbate 80-coated PLGA nanoparticles: Optimization of formulation variables and in vitro anti-glioma assessment. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.05.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
31
|
Kim J, Narayana A, Patel S, Sahay G. Advances in intracellular delivery through supramolecular self-assembly of oligonucleotides and peptides. Theranostics 2019; 9:3191-3212. [PMID: 31244949 PMCID: PMC6567962 DOI: 10.7150/thno.33921] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 04/09/2019] [Indexed: 12/15/2022] Open
Abstract
Cells utilize natural supramolecular assemblies to maintain homeostasis and biological functions. Naturally inspired modular assembly of biomaterials are now being exploited for understanding or manipulating cell biology for treatment, diagnosis, and detection of diseases. Supramolecular biomaterials, in particular peptides and oligonucleotides, can be precisely tuned to have diverse structural, mechanical, physicochemical and biological properties. These merits of oligonucleotides and peptides as building blocks have given rise to the evolution of numerous nucleic acid- and peptide-based self-assembling nanomaterials for various medical applications, including drug delivery, tissue engineering, regenerative medicine, and immunotherapy. In this review, we provide an extensive overview of the intracellular delivery approaches using supramolecular self-assembly of DNA, RNA, and peptides. Furthermore, we discuss the current challenges related to subcellular delivery and provide future perspectives of the application of supramolecular biomaterials for intracellular delivery in theranostics.
Collapse
Affiliation(s)
- Jeonghwan Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR
| | - Ashwanikumar Narayana
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR
| | - Siddharth Patel
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR
| | - Gaurav Sahay
- Department of Pharmaceutical Sciences, College of Pharmacy, Robertson Life Sciences Building, Oregon State University, Portland, OR
- Department of Biomedical Engineering, Robertson Life Sciences Building, Oregon Health Science University, Portland, OR
| |
Collapse
|
32
|
Nurunnabi M, Khatun Z, Badruddoza AZM, McCarthy JR, Lee YK, Huh KM. Biomaterials and Bioengineering Approaches for Mitochondria and Nuclear Targeting Drug Delivery. ACS Biomater Sci Eng 2019. [DOI: 10.1021/acsbiomaterials.8b01615] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Md Nurunnabi
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129 United States
| | - Zehedina Khatun
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts 02111 United States
| | - Abu Zayed Md Badruddoza
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, Richmond, Virginia 23219 United States
| | - Jason R. McCarthy
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129 United States
| | - Yong-kyu Lee
- Department of Chemical and Biological Engineering, Korea National University of Transportation, Chungju 380-706, Republic of Korea
| | - Kang Moo Huh
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon 305-764, Republic of Korea
| |
Collapse
|
33
|
A novel elastin-like polypeptide drug carrier for cyclosporine A improves tear flow in a mouse model of Sjögren's syndrome. J Control Release 2018; 292:183-195. [PMID: 30359668 DOI: 10.1016/j.jconrel.2018.10.026] [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: 06/30/2018] [Revised: 10/05/2018] [Accepted: 10/22/2018] [Indexed: 12/16/2022]
Abstract
As a potent macrolide immunosuppressant, cyclosporine A (CsA) is used to treat multiple autoimmune diseases, including non-autoimmune and autoimmune-mediated dry eye disease, rheumatoid arthritis and psoriasis. Despite its potency, CsA has poor solubility, poor bioavailability, and can cause serious adverse reactions such as nephrotoxicity and neurotoxicity. To overcome these limitations, we invented a new strategy to carry CsA by fusing its cognate human receptor, cyclophilin A (CypA), to a 73 kDa elastin-like polypeptide (ELP) termed A192 using recombinant protein expression. Derived from human tropoelastin, ELPs are characterized by the ability to phase separate above a temperature that is a function of variables including concentration, molecular weight, and hydrophobicity. The resultant fusion protein, termed CA192, which assembles into a dimeric species in solution, effectively binds and solubilizes CsA with a Kd of 189 nM, comparable to that of endogenous CypA with a Kd of 35.5 nM. The release profile of CsA from CA192 follows a one phase decay model with a half-life of 957.3 h without a burst release stage. Moreover, CA192-CsA inhibited IL-2 expression induced in Jurkat cells through the calcineurin-NFAT signaling pathway with an IC50 of 1.2 nM, comparable to that of free CsA with an IC50 of 0.5 nM. The intravenous pharmacokinetics of CA192 followed a two-compartment model with a mean residence time of 7.3 h. Subcutaneous administration revealed a bioavailability of 30% and a mean residence time of 15.9 h. When given subcutaneously for 2 weeks starting at 14 weeks in male non-obese diabetic (NOD) mice, a model of autoimmune dacryoadenitis used to study Sjögren's syndrome (SS), CA192-CsA (2.5 mg/kg, every other day) significantly (p = 0.014) increased tear production relative to CA192 alone. Moreover, CA192 delivery reduced indications of CsA nephrotoxicity relative to free CsA. CA192 represents a viable new approach to deliver this effective but nephrotoxic agent in a modality that preserves therapeutic efficacy but suppresses drug toxicity.
Collapse
|
34
|
Zhang W, Song Y, Eldi P, Guo X, Hayball JD, Garg S, Albrecht H. Targeting prostate cancer cells with hybrid elastin-like polypeptide/liposome nanoparticles. Int J Nanomedicine 2018; 13:293-305. [PMID: 29391790 PMCID: PMC5768422 DOI: 10.2147/ijn.s152485] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Prostate cancer cells frequently overexpress the gastrin-releasing peptide receptor, and various strategies have been applied in preclinical settings to target this receptor for the specific delivery of anticancer compounds. Recently, elastin-like polypeptide (ELP)-based self-assembling micelles with tethered GRP on the surface have been suggested to actively target prostate cancer cells. Poorly soluble chemotherapeutics such as docetaxel (DTX) can be loaded into the hydrophobic cores of ELP micelles, but only limited drug retention times have been achieved. Herein, we report the generation of hybrid ELP/liposome nanoparticles which self-assembled rapidly in response to temperature change, encapsulated DTX at high concentrations with slow release, displayed the GRP ligand on the surface, and specifically bound to GRP receptor expressing PC-3 cells as demonstrated by flow cytometry. This novel type of drug nanocarrier was successfully used to reduce cell viability of prostate cancer cells in vitro through the specific delivery of DTX.
Collapse
Affiliation(s)
- Wei Zhang
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Yunmei Song
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Preethi Eldi
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Xiuli Guo
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, Jinan, China
| | - John D Hayball
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia.,Robinson Research Institute and Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Sanjay Garg
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| | - Hugo Albrecht
- Centre for Pharmaceutical Innovation and Development, Centre for Drug Discovery and Development, Experimental Therapeutics Laboratory, Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, SA, Australia
| |
Collapse
|
35
|
Abstract
The protein elastin imparts extensibility, elastic recoil, and resilience to tissues including arterial walls, skin, lung alveoli, and the uterus. Elastin and elastin-like peptides are hydrophobic, disordered, and undergo liquid-liquid phase separation upon self-assembly. Despite extensive study, the structure of elastin remains controversial. We use molecular dynamics simulations on a massive scale to elucidate the structural ensemble of aggregated elastin-like peptides. Consistent with the entropic nature of elastic recoil, the aggregated state is stabilized by the hydrophobic effect. However, self-assembly does not entail formation of a hydrophobic core. The polypeptide backbone forms transient, sparse hydrogen-bonded turns and remains significantly hydrated even as self-assembly triples the extent of non-polar side chain contacts. Individual chains in the assembly approach a maximally-disordered, melt-like state which may be called the liquid state of proteins. These findings resolve long-standing controversies regarding elastin structure and function and afford insight into the phase separation of disordered proteins.
Collapse
Affiliation(s)
- Sarah Rauscher
- Molecular MedicineThe Hospital for Sick ChildrenTorontoCanada
- Department of BiochemistryUniversity of TorontoTorontoCanada
| | - Régis Pomès
- Molecular MedicineThe Hospital for Sick ChildrenTorontoCanada
- Department of BiochemistryUniversity of TorontoTorontoCanada
| |
Collapse
|
36
|
Dhandhukia JP, Shi P, Peddi S, Li Z, Aluri S, Ju Y, Brill D, Wang W, Janib SM, Lin YA, Liu S, Cui H, MacKay JA. Bifunctional Elastin-like Polypeptide Nanoparticles Bind Rapamycin and Integrins and Suppress Tumor Growth in Vivo. Bioconjug Chem 2017; 28:2715-2728. [PMID: 28937754 DOI: 10.1021/acs.bioconjchem.7b00469] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Recombinant protein-polymer scaffolds such as elastin-like polypeptides (ELPs) offer drug-delivery opportunities including biocompatibility, monodispersity, and multifunctionality. We recently reported that the fusion of FK-506 binding protein 12 (FKBP) to an ELP nanoparticle (FSI) increases rapamycin (Rapa) solubility, suppresses tumor growth in breast cancer xenografts, and reduces side effects observed with free-drug controls. This new report significantly advances this carrier strategy by demonstrating the coassembly of two different ELP diblock copolymers containing drug-loading and tumor-targeting domains. A new ELP nanoparticle (ISR) was synthesized that includes the canonical integrin-targeting ligand (Arg-Gly-Asp, RGD). FSI and ISR mixed in a 1:1 molar ratio coassemble into bifunctional nanoparticles containing both the FKBP domain for Rapa loading and the RGD ligand for integrin binding. Coassembled nanoparticles were evaluated for bifunctionality by performing in vitro cell-binding and drug-retention assays and in vivo MDA-MB-468 breast tumor regression and tumor-accumulation studies. The bifunctional nanoparticle demonstrated superior cell target binding and similar drug retention to FSI; however, it enhanced the formulation potency, such that tumor growth was suppressed at a 3-fold lower dose compared to an untargeted FSI-Rapa control. This data suggests that ELP-mediated scaffolds are useful tools for generating multifunctional nanomedicines with potential activity in cancer.
Collapse
Affiliation(s)
- Jugal P Dhandhukia
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy , Los Angeles, California 90089, United States
| | - Pu Shi
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy , Los Angeles, California 90089, United States
| | - Santosh Peddi
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy , Los Angeles, California 90089, United States
| | - Zhe Li
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy , Los Angeles, California 90089, United States
| | - Suhaas Aluri
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy , Los Angeles, California 90089, United States
| | - Yaping Ju
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy , Los Angeles, California 90089, United States
| | - Dab Brill
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy , Los Angeles, California 90089, United States
| | - Wan Wang
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy , Los Angeles, California 90089, United States
| | - Siti M Janib
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy , Los Angeles, California 90089, United States
| | - Yi-An Lin
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University , Baltimore, Maryland 21218, United States
| | | | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University , Baltimore, Maryland 21218, United States
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California School of Pharmacy , Los Angeles, California 90089, United States.,Department of Biomedical Engineering, University of Southern California Viterbi School of Engineering , Los Angeles, California 90089, United States
| |
Collapse
|
37
|
Dhandhukia JP, Li Z, Peddi S, Kakan S, Mehta A, Tyrpak D, Despanie J, MacKay JA. Berunda Polypeptides: Multi-Headed Fusion Proteins Promote Subcutaneous Administration of Rapamycin to Breast Cancer In Vivo. Theranostics 2017; 7:3856-3872. [PMID: 29109782 PMCID: PMC5667409 DOI: 10.7150/thno.19981] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 08/01/2017] [Indexed: 11/05/2022] Open
Abstract
Recombinant Elastin-Like Polypeptides (ELPs) serve as attractive scaffolds for nanoformulations because they can be charge-neutral, water soluble, high molecular weight, monodisperse, biodegradable, and decorated with functional proteins. We recently reported that fusion of the FK-506 binding protein 12 (FKBP) to an ELP nanoparticle (FSI) reduces rapamycin (Rapa) toxicity and enables intravenous (IV) therapy in both a xenograft breast cancer model and a murine autoimmune disease model. Rapa has poor solubility, which leads to variable oral bioavailability or drug precipitation following parenteral administration. While IV administration is routine during chemotherapy, cytostatic molecules like Rapa would require repeat administrations in clinical settings. To optimize FKBP/Rapa for subcutaneous (SC) administration, this manuscript expands upon first-generation FSI nanoparticles (Rh ~ 25 nm) and compares them with two second-generation carriers (FA and FAF) that: i) do not self-assemble; ii) retain a hydrodynamic radius (Rh ~ 7 nm) above the renal filtration cutoff; iii) increase tumor accumulation; and iv) have either one (FA) or two (FAF) drug-binding FKBP domains per ELP protein. Methods: The carriers were compared and evaluated for temperature-concentration phase behavior by UV-Vis spectrophotometry; equilibrium binding and thermodynamics by Isothermal Titration Calorimetry; drug retention and formulation stability by Dialysis and Dynamic Light Scattering; in vitro efficacy using a cell proliferation assay; in vivo efficacy in human MDA-MB-468 orthotopic breast cancer xenografts; downstream target inhibition using western blot; tissue histopathology; and bio-distribution via optical imaging in the orthotopic xenograft mouse model. Results: Named after the two-headed bird in Hindu mythology, the 'Berunda polypeptide' FAF with molecular weight of 97 kDa and particle size, Rh ~ 7 nm demonstrated polypeptide conformation of a soluble hydrated coiled polymer, retained formulation stability for one month post Rapa loading, eliminated toxicity observed with free Rapa after SC administration, suppressed tumor growth, decreased phosphorylation of a downstream target, and increased tumor accumulation in orthotopic breast tumor xenografts. Conclusion: This comprehensive manuscript demonstrates the versatility of recombinant protein-polymers to investigate drug carrier architectures. Furthermore, their facilitation of SC administration of poorly soluble drugs, like Rapa, may enable chronic self-administration in patients.
Collapse
|
38
|
Dhandhukia JP, Brill DA, Kouhi A, Pastuszka MK, MacKay JA. Elastin-like polypeptide switches: A design strategy to detect multimeric proteins. Protein Sci 2017. [PMID: 28639381 DOI: 10.1002/pro.3215] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Elastin-Like Polypeptides (ELPs) reversibly phase separate in response to changes in temperature, pressure, concentration, pH, and ionic species. While powerful triggers, biological microenvironments present a multitude of more specific biological cues, such as antibodies, cytokines, and cell-surface receptors. To develop better biosensors and bioresponsive drug carriers, rational strategies are required to sense and respond to these target proteins. We recently reported that noncovalent association of two ELP fusion proteins to a "chemical inducer of dimerization" small molecule (1.5 kDa) induces phase separation at physiological temperatures. Having detected a small molecule, here we present the first evidence that ELP multimerization can also detect a much larger (60 kDa) protein target. To demonstrate this strategy, ELPs were biotinylated at their amino terminus and mixed with tetrameric streptavidin. At a stoichiometric ratio of [4:1], two to three biotin-ELPs associate with streptavidin into multimeric complexes with an apparent Kd of 5 nM. The increased ELP density around a streptavidin core strongly promotes isothermal phase separation, which was tuned to occur at physiological temperature. This phase separation reverses upon saturation with excess streptavidin, which only favors [1:1] complexes. Together, these findings suggest that ELP association with multimeric biomolecules is a viable strategy to deliberately engineer ELPs that respond to multimeric protein substrates.
Collapse
Affiliation(s)
- Jugal P Dhandhukia
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Dab A Brill
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Aida Kouhi
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - Martha K Pastuszka
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, California.,Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, California, 90089-9121
| |
Collapse
|
39
|
Yin L, Yuvienco C, Montclare JK. Protein based therapeutic delivery agents: Contemporary developments and challenges. Biomaterials 2017; 134:91-116. [PMID: 28458031 DOI: 10.1016/j.biomaterials.2017.04.036] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 04/18/2017] [Accepted: 04/21/2017] [Indexed: 12/15/2022]
Abstract
As unique biopolymers, proteins can be employed for therapeutic delivery. They bear important features such as bioavailability, biocompatibility, and biodegradability with low toxicity serving as a platform for delivery of various small molecule therapeutics, gene therapies, protein biologics and cells. Depending on size and characteristic of the therapeutic, a variety of natural and engineered proteins or peptides have been developed. This, coupled to recent advances in synthetic and chemical biology, has led to the creation of tailor-made protein materials for delivery. This review highlights strategies employing proteins to facilitate the delivery of therapeutic matter, addressing the challenges for small molecule, gene, protein and cell transport.
Collapse
Affiliation(s)
- Liming Yin
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY 11201, United States
| | - Carlo Yuvienco
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY 11201, United States
| | - Jin Kim Montclare
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY 11201, United States; Department of Chemistry, New York University, New York, NY 10003, United States; Department of Biomaterials, NYU College of Dentistry, New York, NY 10010, United States; Department of Biochemistry, SUNY Downstate Medical Center, Brooklyn, NY 11203, United States.
| |
Collapse
|
40
|
MacEwan SR, Weitzhandler I, Hoffmann I, Genzer J, Gradzielski M, Chilkoti A. Phase Behavior and Self-Assembly of Perfectly Sequence-Defined and Monodisperse Multiblock Copolypeptides. Biomacromolecules 2017; 18:599-609. [PMID: 28094978 PMCID: PMC5558835 DOI: 10.1021/acs.biomac.6b01759] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
This paper investigates how the properties of multiblock copolypeptides can be tuned by their block architecture, defined by the size and distribution of blocks along the polymer chain. These parameters were explored by the precise, genetically encoded synthesis of recombinant elastin-like polypeptides (ELPs). A family of ELPs was synthesized in which the composition and length were conserved while the block length and distribution were varied, thus creating 11 ELPs with unique block architectures. To our knowledge, these polymers are unprecedented in their intricately and precisely varied architectures. ELPs exhibit lower critical solution temperature (LCST) behavior and micellar self-assembly, both of which impart easily measured physicochemical properties to the copolymers, providing insight into polymer hydrophobicity and self-assembly into higher order structures, as a function of solution temperature. Even subtle variation in block architecture changed the LCST phase behavior and morphology of these ELPs, measured by their temperature-triggered phase transition and nanoscale self-assembly. Size and morphology of polypeptide micelles could be tuned solely by controlling the block architecture, thus demonstrating that when sequence can be precisely controlled, nanoscale self-assembly of polypeptides can be modulated by block architecture.
Collapse
Affiliation(s)
- Sarah R MacEwan
- Department of Biomedical Engineering, Duke University , Durham, North Carolina 27708, United States
- Research Triangle Materials Research Science and Engineering Center , Durham, North Carolina 27708, United States
| | - Isaac Weitzhandler
- Department of Biomedical Engineering, Duke University , Durham, North Carolina 27708, United States
- Research Triangle Materials Research Science and Engineering Center , Durham, North Carolina 27708, United States
| | - Ingo Hoffmann
- Stranski-Laboratorium fur Physikalische und Theoretische Chemie, Institut fur Chemie, Technische Universität Berlin , 10623, Berlin, Germany
| | - Jan Genzer
- Research Triangle Materials Research Science and Engineering Center , Durham, North Carolina 27708, United States
- Department of Chemical and Biomolecular Engineering, North Carolina State University , Raleigh, North Carolina 27695, United States
| | - Michael Gradzielski
- Stranski-Laboratorium fur Physikalische und Theoretische Chemie, Institut fur Chemie, Technische Universität Berlin , 10623, Berlin, Germany
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Duke University , Durham, North Carolina 27708, United States
- Research Triangle Materials Research Science and Engineering Center , Durham, North Carolina 27708, United States
| |
Collapse
|
41
|
Zhang W, Garg S, Eldi P, Zhou FHH, Johnson IR, Brooks DA, Lam F, Rychkov G, Hayball J, Albrecht H. Targeting prostate cancer cells with genetically engineered polypeptide-based micelles displaying gastrin-releasing peptide. Int J Pharm 2016; 513:270-279. [PMID: 27633281 DOI: 10.1016/j.ijpharm.2016.09.039] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/12/2016] [Accepted: 09/10/2016] [Indexed: 01/15/2023]
|
42
|
Despanie J, Dhandhukia JP, Hamm-Alvarez SF, MacKay JA. Elastin-like polypeptides: Therapeutic applications for an emerging class of nanomedicines. J Control Release 2016; 240:93-108. [PMID: 26578439 PMCID: PMC5767577 DOI: 10.1016/j.jconrel.2015.11.010] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 11/06/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023]
Abstract
Elastin-like polypeptides (ELPs) constitute a genetically engineered class of 'protein polymers' derived from human tropoelastin. They exhibit a reversible phase separation whereby samples remain soluble below a transition temperature (Tt) but form amorphous coacervates above Tt. Their phase behavior has many possible applications in purification, sensing, activation, and nanoassembly. As humanized polypeptides, they are non-immunogenic, substrates for proteolytic biodegradation, and can be decorated with pharmacologically active peptides, proteins, and small molecules. Recombinant synthesis additionally allows precise control over ELP architecture and molecular weight, resulting in protein polymers with uniform physicochemical properties suited to the design of multifunctional biologics. As such, ELPs have been employed for various uses including as anti-cancer agents, ocular drug delivery vehicles, and protein trafficking modulators. This review aims to offer the reader a catalogue of ELPs, their various applications, and potential for commercialization across a broad spectrum of fields.
Collapse
Affiliation(s)
- Jordan Despanie
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90033-9121, USA
| | - Jugal P Dhandhukia
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90033-9121, USA
| | - Sarah F Hamm-Alvarez
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90033-9121, USA; Department of Ophthalmology, University of Southern California, Los Angeles, CA, 90033, USA
| | - J Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90033-9121, USA; Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA.
| |
Collapse
|
43
|
Rodríguez-Cabello JC, Arias FJ, Rodrigo MA, Girotti A. Elastin-like polypeptides in drug delivery. Adv Drug Deliv Rev 2016; 97:85-100. [PMID: 26705126 DOI: 10.1016/j.addr.2015.12.007] [Citation(s) in RCA: 109] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 12/03/2015] [Accepted: 12/07/2015] [Indexed: 12/12/2022]
Abstract
The use of recombinant elastin-like materials, or elastin-like recombinamers (ELRs), in drug-delivery applications is reviewed in this work. Although ELRs were initially used in similar ways to other, more conventional kinds of polymeric carriers, their unique properties soon gave rise to systems of unparalleled functionality and efficiency, with the stimuli responsiveness of ELRs and their ability to self-assemble readily allowing the creation of advanced systems. However, their recombinant nature is likely the most important factor that has driven the current breakthrough properties of ELR-based delivery systems. Recombinant technology allows an unprecedented degree of complexity in macromolecular design and synthesis. In addition, recombinant materials easily incorporate any functional domain present in natural proteins. Therefore, ELR-based delivery systems can exhibit complex interactions with both their drug load and the tissues and cells towards which this load is directed. Selected examples, ranging from highly functional nanocarriers to macrodepots, will be presented.
Collapse
|
44
|
Su H, Koo JM, Cui H. One-component nanomedicine. J Control Release 2015; 219:383-395. [PMID: 26423237 PMCID: PMC4656119 DOI: 10.1016/j.jconrel.2015.09.056] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/24/2015] [Accepted: 09/25/2015] [Indexed: 01/06/2023]
Abstract
One-component nanomedicine (OCN) represents an emerging class of therapeutic nanostructures that contain only one type of chemical substance. This one-component feature allows for fine-tuning and optimization of the drug loading and physicochemical properties of nanomedicine in a precise manner through molecular engineering of the underlying building blocks. Using a precipitation procedure or effective molecular assembly strategies, molecularly crafted therapeutic agents (e.g. polymer-drug conjugates, small molecule prodrugs, or drug amphiphiles) could involuntarily aggregate, or self-assemble into nanoscale objects of well-defined sizes and shapes. Unlike traditional carrier-based nanomedicines that are inherently multicomponent systems, an OCN does not require the use of additional carriers and could itself possess desired physicochemical features for preferential accumulation at target sites. We review here recent progress in the molecular design, conjugation methods, and fabrication strategies of OCN, and analyze the opportunities that this emerging platform could open for the new and improved treatment of devastating diseases such as cancer.
Collapse
Affiliation(s)
- Hao Su
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
| | - Jin Mo Koo
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA
| | - Honggang Cui
- Department of Chemical and Biomolecular Engineering, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA; Institute for NanoBioTechnology, The Johns Hopkins University, 3400 N Charles Street, Baltimore, MD 21218, USA; Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Nanomedicine, The Wilmer Eye Institute, Johns Hopkins University School of Medicine, 400 North Broadway, Baltimore, MD 21231, USA.
| |
Collapse
|
45
|
Taniguchi S, Watanabe N, Nose T, Maeda I. Development of short and highly potent self-assembling elastin-derived pentapeptide repeats containing aromatic amino acid residues. J Pept Sci 2015; 22:36-42. [PMID: 26662843 DOI: 10.1002/psc.2837] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 10/11/2015] [Accepted: 10/21/2015] [Indexed: 11/07/2022]
Abstract
Tropoelastin is the primary component of elastin, which forms the elastic fibers that make up connective tissues. The hydrophobic domains of tropoelastin are thought to mediate the self-assembly of elastin into fibers, and the temperature-mediated self-assembly (coacervation) of one such repetitive peptide sequence (VPGVG) has been utilized in various bio-applications. To elucidate a mechanism for coacervation activity enhancement and to develop more potent coacervatable elastin-derived peptides, we synthesized two series of peptide analogs containing an aromatic amino acid, Trp or Tyr, in addition to Phe-containing analogs and tested their functional characteristics. Thus, position 1 of the hydrophobic pentapeptide repeat of elastin (X(1)P(2)G(3)V(4)G(5)) was substituted by Trp or Tyr. Eventually, we acquired a novel, short Trp-containing elastin-derived peptide analog (WPGVG)3 with potent coacervation ability. From the results obtained during this process, we determined the importance of aromaticity and hydrophobicity for the coacervation potency of elastin-derived peptide analogs. Generally, however, the production of long-chain synthetic polypeptides in quantities sufficient for commercial use remain cost-prohibitive. Therefore, the identification of (WPGVG)3, which is a 15-mer short peptide consisting simply of five natural amino acids and shows temperature-dependent self-assembly activity, might serve as a foundation for the development of various kinds of biomaterials.
Collapse
Affiliation(s)
- Suguru Taniguchi
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka, 820-8502, Japan
| | - Noriko Watanabe
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka, 820-8502, Japan
| | - Takeru Nose
- Faculty of Arts and Science, Kyushu University, Fukuoka, 819-0395, Japan
| | - Iori Maeda
- Department of Bioscience and Bioinformatics, Kyushu Institute of Technology, Iizuka, Fukuoka, 820-8502, Japan
| |
Collapse
|
46
|
Yeo GC, Aghaei-Ghareh-Bolagh B, Brackenreg EP, Hiob MA, Lee P, Weiss AS. Fabricated Elastin. Adv Healthc Mater 2015; 4:2530-2556. [PMID: 25771993 PMCID: PMC4568180 DOI: 10.1002/adhm.201400781] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 02/09/2015] [Indexed: 12/18/2022]
Abstract
The mechanical stability, elasticity, inherent bioactivity, and self-assembly properties of elastin make it a highly attractive candidate for the fabrication of versatile biomaterials. The ability to engineer specific peptide sequences derived from elastin allows the precise control of these physicochemical and organizational characteristics, and further broadens the diversity of elastin-based applications. Elastin and elastin-like peptides can also be modified or blended with other natural or synthetic moieties, including peptides, proteins, polysaccharides, and polymers, to augment existing capabilities or confer additional architectural and biofunctional features to compositionally pure materials. Elastin and elastin-based composites have been subjected to diverse fabrication processes, including heating, electrospinning, wet spinning, solvent casting, freeze-drying, and cross-linking, for the manufacture of particles, fibers, gels, tubes, sheets and films. The resulting materials can be tailored to possess specific strength, elasticity, morphology, topography, porosity, wettability, surface charge, and bioactivity. This extraordinary tunability of elastin-based constructs enables their use in a range of biomedical and tissue engineering applications such as targeted drug delivery, cell encapsulation, vascular repair, nerve regeneration, wound healing, and dermal, cartilage, bone, and dental replacement.
Collapse
Affiliation(s)
- Giselle C. Yeo
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
- School of Molecular Bioscience, The University of Sydney, NSW 2006, Australia
| | - Behnaz Aghaei-Ghareh-Bolagh
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
- School of Molecular Bioscience, The University of Sydney, NSW 2006, Australia
| | - Edwin P. Brackenreg
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
- School of Molecular Bioscience, The University of Sydney, NSW 2006, Australia
| | - Matti A. Hiob
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
- School of Molecular Bioscience, The University of Sydney, NSW 2006, Australia
| | - Pearl Lee
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
- School of Molecular Bioscience, The University of Sydney, NSW 2006, Australia
| | - Anthony S. Weiss
- Charles Perkins Centre, The University of Sydney, NSW 2006, Australia
- School of Molecular Bioscience, The University of Sydney, NSW 2006, Australia
- Bosch Institute, The University of Sydney, NSW 2006, Australia
| |
Collapse
|
47
|
Mendes TFS, Kluskens LD, Rodrigues LR. Triple Negative Breast Cancer: Nanosolutions for a Big Challenge. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2015; 2:1500053. [PMID: 27980912 PMCID: PMC5115335 DOI: 10.1002/advs.201500053] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 06/03/2015] [Indexed: 05/11/2023]
Abstract
Triple negative breast cancer (TNBC) is a particular immunopathological subtype of breast cancer that lacks expression of estrogen and progesterone receptors (ER/PR) and amplification of the human epidermal growth factor receptor 2 (HER2) gene. Characterized by aggressive and metastatic phenotypes and high rates of relapse, TNBC is the only breast cancer subgroup still lacking effective therapeutic options, thus presenting the worst prognosis. The development of targeted therapies, as well as early diagnosis methods, is vital to ensure an adequate and timely therapeutic intervention in patients with TNBC. This review intends to discuss potentially emerging approaches for the diagnosis and treatment of TNBC patients, with a special focus on nano-based solutions that actively target these particular tumors.
Collapse
Affiliation(s)
| | - Leon D Kluskens
- Centre of Biological Engineering University of Minho 4710-057 Braga Portugal
| | | |
Collapse
|
48
|
Brill DA, MacKay JA. Image-driven pharmacokinetics: nanomedicine concentration across space and time. Nanomedicine (Lond) 2015; 10:2861-79. [PMID: 26370694 DOI: 10.2217/nnm.15.127] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Clinical pharmacokinetics (PK) primarily measures the concentration of drugs in the blood. For nanomedicines it may be more relevant to determine concentration within a target tissue. The emerging field of image-driven PK, which utilizes clinically accepted molecular imaging technology, empirically and noninvasively, measures concentration in multiple tissues. Image-driven PK represents the intersection of PK and biodistribution, combining to provide models of concentration across space and time. Image-driven PK can be used both as a research tool and in the clinic. This review explores the history of pharmacokinetics, technologies used in molecular imaging (especially positron emission tomography) and research using image-driven pharmacokinetic analysis. When standardized, image-driven PK may have significant implications in preclinical development as well as clinical optimization of targeted nanomedicines.
Collapse
Affiliation(s)
- Dab A Brill
- Department of Pharmacology & Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA
| | - J Andrew MacKay
- Department of Pharmacology & Pharmaceutical Sciences, University of Southern California, Los Angeles, CA, USA.,Department of Biomedical Engineering, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
49
|
Abstract
Polymeric nanomaterials have extensively been applied for the preparation of targeted and controlled release drug/gene delivery systems. However, problems involved in the formulation of synthetic polymers such as using of the toxic solvents and surfactants have limited their desirable applications. In this regard, natural biomolecules including proteins and polysaccharide are suitable alternatives due to their safety. According to literature, protein-based nanoparticles possess many advantages for drug and gene delivery such as biocompatibility, biodegradability and ability to functionalize with targeting ligands. This review provides a general sight on the application of biodegradable protein-based nanoparticles in drug/gene delivery based on their origins. Their unique physicochemical properties that help them to be formulated as pharmaceutical carriers are also discussed.
Collapse
|
50
|
Cho S, Dong S, Parent KN, Chen M. Immune-tolerant elastin-like polypeptides (iTEPs) and their application as CTL vaccine carriers. J Drug Target 2015; 24:328-39. [PMID: 26307138 PMCID: PMC4813525 DOI: 10.3109/1061186x.2015.1077847] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 07/27/2015] [Indexed: 01/19/2023]
Abstract
BACKGROUND Cytotoxic T lymphocyte (CTL) vaccine carriers are known to enhance the efficacy of vaccines, but a search for more effective carriers is warranted. Elastin-like polypeptides (ELPs) have been examined for many medical applications but not as CTL vaccine carriers. PURPOSE We aimed to create immune tolerant ELPs using a new polypeptide engineering practice and create CTL vaccine carriers using the ELPs. RESULTS Four sets of novel ELPs, termed immune-tolerant elastin-like polypeptide (iTEP) were generated according to the principles dictating humoral immunogenicity of polypeptides and phase transition property of ELPs. The iTEPs were non-immunogenic in mice. Their phase transition feature was confirmed through a turbidity assay. An iTEP nanoparticle (NP) was assembled from an amphiphilic iTEP copolymer plus a CTL peptide vaccine, SIINFEKL. The NP facilitated the presentation of the vaccine by dendritic cells (DCs) and enhanced vaccine-induced CTL responses. DISCUSSION A new ELP design and development practice was established. The non-canonical motif and the immune tolerant nature of the iTEPs broaden our insights about ELPs. ELPs, for the first time, were successfully used as carriers for CTL vaccines. CONCLUSION It is feasible to concurrently engineer both immune-tolerant and functional peptide materials. ELPs are a promising type of CTL vaccine carriers.
Collapse
Affiliation(s)
- S. Cho
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 E 2000 S, Salt Lake City, Utah 84112
| | - S. Dong
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 E 2000 S, Salt Lake City, Utah 84112
| | - K. N. Parent
- Dept. of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, East Lansing, MI 48824
| | - M. Chen
- Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, 30 E 2000 S, Salt Lake City, Utah 84112
| |
Collapse
|