1
|
Yadav K, Ebenezer Gnanakani SP, Kumar Sahu K, Sucheta, Dubey A, Minz S, Raza W, Pradhan M. Unleashing the potential of natural protein based nanoparticles for the delivery of therapeutic nucleic Acid: A comprehensive review. Int J Pharm 2025; 669:125049. [PMID: 39674384 DOI: 10.1016/j.ijpharm.2024.125049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 11/23/2024] [Accepted: 12/04/2024] [Indexed: 12/16/2024]
Abstract
Nucleic acid-based therapeutics represent a revolutionary approach in treating genetic disorders, offering unprecedented potential for addressing pathologies at their molecular level. However, effective cellular delivery remains a critical challenge hindering their clinical implementation. While existing delivery systems, including viral vectors and lipid nanoparticles, have shown utility, they face limitations in immunogenicity, cargo capacity, and manufacturing complexity. Natural protein-based nanoparticles, derived from proteins such as albumin, ferritin, and elastin, have emerged as promising alternative delivery systems. These carriers offer distinct advantages including reduced immunogenicity, enhanced biocompatibility, and optimal biodegradation profiles. Their engineerable nature enables precise control over particle size, surface charge, and ligand conjugation, facilitating selective cellular targeting and improved pharmacokinetics. Recent technological advances have expanded the application of protein nanoparticles across various nucleic acid modalities, including mRNA, siRNA, and plasmid DNA. Extensive research has characterized these systems through rigorous in vitro and in vivo studies, advancing our understanding of their biological behavior and clinical potential. Advanced engineering methodologies have further enhanced their optimization for specific therapeutic applications. This review examines the development and potential of protein-based nanoparticles in nucleic acid delivery, highlighting their advantages and addressing current challenges. By analyzing recent advances and clinical progress, we underscore their significant potential to enhance the safety, specificity, and efficacy of nucleic acid therapeutics, potentially revolutionizing the treatment of genetic disorders.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kurud Road, Kohka, Bhilai 490024, Chhattisgarh, India
| | - S Princely Ebenezer Gnanakani
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Limda, Waghodia, Vadodara, Gujarat 391760, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | - Akhilesh Dubey
- Nitte (Deemed to be University), NGSM Institute of Pharmaceutical Sciences, Department of Pharmaceutics, Mangaluru 575018, Karnataka, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | - Wasim Raza
- Central Laboratory Facility, Chhattisgarh Council of Science and Technology, Vigyan Bhawan, Raipur, Chhattisgarh, India
| | | |
Collapse
|
2
|
Panahipour L, Imani A, dos Santos Sanches N, Kühtreiber H, Mildner M, Gruber R. RNA Sequencing Revealed a Weak Response of Gingival Fibroblasts Exposed to Hyaluronic Acid. Bioengineering (Basel) 2024; 11:1307. [PMID: 39768125 PMCID: PMC11726844 DOI: 10.3390/bioengineering11121307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Hyaluronic acid was proposed to support soft tissue recession surgery and guided tissue regeneration. The molecular mechanisms through which hyaluronic acid modulates the response of connective tissue cells remain elusive. To elucidate the impact of hyaluronic acid on the connective tissue cells, we used bulk RNA sequencing to determine the changes in the genetic signature of gingival fibroblasts exposed to 1.6% cross-linked hyaluronic acid and 0.2% natural hyaluronic acid. Transcriptome-wide changes were modest. Even when implementing a minimum of 1.5 log2 fold-change and a significance threshold of 1.0 -log10, only a dozenth of genes were differentially expressed. Upregulated genes were PLK3, SLC16A6, IL6, HBEGF, DGKE, DUSP4, PTGS2, FOXC2, ATAD2B, NFATC2, and downregulated genes were MMP24 and PLXNA2. RT-PCR analysis supported the impact of hyaluronic acid on increasing the expression of a selected gene panel. The findings from bulk RNA sequencing suggest that gingival fibroblasts experience weak changes in their transcriptome when exposed to hyaluronic acid.
Collapse
Affiliation(s)
- Layla Panahipour
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (A.I.); (N.d.S.S.)
| | - Atefe Imani
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (A.I.); (N.d.S.S.)
| | - Natália dos Santos Sanches
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (A.I.); (N.d.S.S.)
- Department of Diagnosis and Surgery, Araçatuba Dental School of Sao Paulo, Sao Paulo 16015-050, Brazil
| | - Hannes Kühtreiber
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria; (H.K.); (M.M.)
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, 1090 Vienna, Austria; (H.K.); (M.M.)
| | - Reinhard Gruber
- Department of Oral Biology, University Clinic of Dentistry, Medical University of Vienna, Sensengasse 2a, 1090 Vienna, Austria; (L.P.); (A.I.); (N.d.S.S.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3010 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
| |
Collapse
|
3
|
Adhikari B, Stager MA, Collins EG, Fischenich KM, Olusoji J, Ruble AF, Payne KA, Krebs MD. Sustained release of MAPK14-targeting siRNA from polyelectrolyte complex hydrogels mitigates MSC osteogenesis in vitro with potential application in growth plate injury. J Biomed Mater Res A 2024; 112:2346-2357. [PMID: 39145460 DOI: 10.1002/jbm.a.37784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 07/13/2024] [Accepted: 07/30/2024] [Indexed: 08/16/2024]
Abstract
The growth plate is a cartilage structure at the end of long bones which mediates growth in children. When fractured, the formation of bony repair tissue known as a "bony bar" can occur and cause limb deformities. There are currently no effective clinical solutions for the prevention of the bony bar formation or regeneration of healthy growth plate cartilage after a fracture. This study employs previously developed alginate/chitosan polyelectrolyte complex (PEC) hydrogels as a sustained release vehicle for the delivery of short-interfering RNA (siRNA). Specifically, the siRNA targets the p38-MAPK pathway in mesenchymal stem cells (MSCs) to prevent their osteogenic differentiation. In vitro experimental findings show sustained release of siRNA from the hydrogels for 6 months. Flow cytometry and confocal imaging indicate that the hydrogels release siRNA to effectively knockdown GFP expression over a sustained period. MAPK-14 targeting siRNA was used to knockdown the expression of MAPK-14 and correspondingly decrease the expression of other osteogenic genes in MSCs in vitro over the span of 21 days. These hydrogels were used in a rat model of growth plate injury to determine whether siMAPK-14 released from the gels could inhibit bony bar formation. No significant reduction of bony bar formation was seen in vivo at the one concentration of siRNA examined. This PEC hydrogel represents a significant advancement for siRNA sustained delivery, and presents an interesting potential therapeutic delivery system for growth plate injuries and other regenerative medicine applications.
Collapse
Affiliation(s)
- Bikram Adhikari
- Quantitative Biosciences and Bioengineering, Colorado School of Mines, Golden, Colorado, USA
| | - Michael A Stager
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Elise G Collins
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| | - Kristine M Fischenich
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Jesutomisin Olusoji
- Quantitative Biosciences and Bioengineering, Colorado School of Mines, Golden, Colorado, USA
| | - Ana Ferreira Ruble
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Karin A Payne
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Melissa D Krebs
- Quantitative Biosciences and Bioengineering, Colorado School of Mines, Golden, Colorado, USA
- Chemical and Biological Engineering, Colorado School of Mines, Golden, Colorado, USA
| |
Collapse
|
4
|
Liu S, Guo H, Li D, Wang C. Immunologically effective biomaterials enhance immunotherapy of prostate cancer. J Mater Chem B 2024; 12:9821-9834. [PMID: 39239675 DOI: 10.1039/d3tb03044j] [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
Prostate cancer (PCa) is one of the most common malignant neoplasms affecting the male population. The onset of the disease is insidious and often associated with severe consequences, such as bone metastases at the time of initial diagnosis. Once it advances to metastatic castration-resistant PCa (mCRPC), conventional treatment methods become ineffective. As research on the mechanism of tumor therapy advances, immunotherapy has been evolving rapidly. However, PCa is a solid tumor type that primarily faces the challenges of poor immunogenicity and inhibitory tumor microenvironment (TME). Fortunately, the extensive use of biomaterials has led to continuous advancement in PCa immunotherapy. These innovative materials aim to address intractable issues, such as immune escape and immune desert, to inhibit tumor progression and metastasis. This detailed review focuses on the regulation of different aspects of tumor immunity by immunologically effective biomaterials, including modulating adaptive immunity, innate immunity, and the immune microenvironment, to enhance the efficacy of PCa immunotherapy. In addition, this review provides a perspective on the future prospects of immunotherapeutic nanoplatforms based on biomaterials in the treatment of PCa.
Collapse
Affiliation(s)
- Siqi Liu
- Department of Urology, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| | - Hui Guo
- Department of Urology, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| | - Di Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| | - Chunxi Wang
- Department of Urology, General Surgery Center, The First Hospital of Jilin University, 1 Xinmin Street, Changchun 130061, P. R. China
| |
Collapse
|
5
|
Kucharczyk K, Florczak A, Kaminska A, Guzniczak N, Sikorska A, Deptuch T, Dams-Kozlowska H. MMPs-responsive silk spheres for controlled drug release within tumor microenvironment. Int J Biol Macromol 2024; 269:132016. [PMID: 38697442 DOI: 10.1016/j.ijbiomac.2024.132016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/20/2024] [Accepted: 04/29/2024] [Indexed: 05/05/2024]
Abstract
Silk is a biocompatible and biodegradable material that enables the formation of various morphological forms, including nanospheres. The functionalization of bioengineered silk makes it possible to produce particles with specific properties. In addition to tumor cells, the tumor microenvironment (TME) includes stromal, immune, endothelial cells, signaling molecules, and the extracellular matrix (ECM). Matrix metalloproteinases (MMPs) are overexpressed in TME. We investigated bioengineered spider silks functionalized with MMP-responsive peptides to obtain targeted drug release from spheres within TME. Soluble silks MS12.2MS1, MS12.9MS1, and MS22.9MS2 and the corresponding silk spheres carrying MMP-2 or MMP-2/9 responsive peptides were produced, loaded with doxorubicin (Dox), and analyzed for their susceptibility to MMP-2/9 digestion. Although all variants of functionalized silks and spheres were specifically degraded by MMP-2/9, the MS22.9MS2 nanospheres showed the highest levels of degradation and release of Dox after enzyme treatment. Moreover, functionalized spheres were degraded in the presence of cancer cells releasing MMP-2/9. In the 2D and 3D spheroid cancer models, the MMP-2/9-responsive substrate was degraded and released from spheres when loaded into MS22.9MS2 particles but not into the control MS2 spheres. The present study demonstrated that a silk-based MMP-responsive delivery system could be used for controlled drug release within the tumor microenvironment.
Collapse
Affiliation(s)
- Kamil Kucharczyk
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Anna Florczak
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Alicja Kaminska
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
| | - Natalia Guzniczak
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland
| | - Agata Sikorska
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Tomasz Deptuch
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Hanna Dams-Kozlowska
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland.
| |
Collapse
|
6
|
Li M, Yao H, Yi K, Lao YH, Shao D, Tao Y. Emerging nanoparticle platforms for CpG oligonucleotide delivery. Biomater Sci 2024; 12:2203-2228. [PMID: 38293828 DOI: 10.1039/d3bm01970e] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Unmethylated cytosine-phosphate-guanine (CpG) oligodeoxynucleotides (ODNs), which were therapeutic DNA with high immunostimulatory activity, have been applied in widespread applications from basic research to clinics as therapeutic agents for cancer immunotherapy, viral infection, allergic diseases and asthma since their discovery in 1995. The major factors to consider for clinical translation using CpG motifs are the protection of CpG ODNs from DNase degradation and the delivery of CpG ODNs to the Toll-like receptor-9 expressed human B-cells and plasmacytoid dendritic cells. Therefore, great efforts have been devoted to the advances of efficient delivery systems for CpG ODNs. In this review, we outline new horizons and recent developments in this field, providing a comprehensive summary of the nanoparticle-based CpG delivery systems developed to improve the efficacy of CpG-mediated immune responses, including DNA nanostructures, inorganic nanoparticles, polymer nanoparticles, metal-organic-frameworks, lipid-based nanosystems, proteins and peptides, as well as exosomes and cell membrane nanoparticles. Moreover, future challenges in the establishment of CpG delivery systems for immunotherapeutic applications are discussed. We expect that the continuously growing interest in the development of CpG-based immunotherapy will certainly fuel the excitement and stimulation in medicine research.
Collapse
Affiliation(s)
- Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Haochen Yao
- Hepatobiliary and Pancreatic Surgery Department, General Surgery Center, First Hospital of Jilin University, No. 1 Xinmin Street, Changchun, 130021, Jilin, China
| | - Ke Yi
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Yeh-Hsing Lao
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14214, USA
| | - Dan Shao
- Institutes of Life Sciences, School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Yu Tao
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
7
|
Peng X, Liu Z, Gao J, Zhang Y, Wang H, Li C, Lv X, Gao Y, Deng H, Zhao B, Gao T, Li H. Influence of Spider Silk Protein Structure on Mechanical and Biological Properties for Energetic Material Detection. Molecules 2024; 29:1025. [PMID: 38474537 PMCID: PMC10934110 DOI: 10.3390/molecules29051025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 03/14/2024] Open
Abstract
Spider silk protein, renowned for its excellent mechanical properties, biodegradability, chemical stability, and low immune and inflammatory response activation, consists of a core domain with a repeat sequence and non-repeating sequences at the N-terminal and C-terminal. In this review, we focus on the relationship between the silk structure and its mechanical properties, exploring the potential applications of spider silk materials in the detection of energetic materials.
Collapse
Affiliation(s)
- Xinying Peng
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Zhiyong Liu
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Junhong Gao
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Yuhao Zhang
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Hong Wang
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Cunzhi Li
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Xiaoqiang Lv
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Yongchao Gao
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Hui Deng
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Bin Zhao
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Ting Gao
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| | - Huan Li
- Toxicology Research Center, Institute for Hygiene of Ordnance Industry, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China (Z.L.)
- Xi’an Key Laboratory of Toxicology and Biological Effects, NO. 12 Zhangbadong Road, Yanta District, Xi’an 710065, China
| |
Collapse
|
8
|
Molenda S, Sikorska A, Florczak A, Lorenc P, Dams-Kozlowska H. Oligonucleotide-Based Therapeutics for STAT3 Targeting in Cancer-Drug Carriers Matter. Cancers (Basel) 2023; 15:5647. [PMID: 38067351 PMCID: PMC10705165 DOI: 10.3390/cancers15235647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 09/08/2024] Open
Abstract
High expression and phosphorylation of signal transducer and transcription activator 3 (STAT3) are correlated with progression and poor prognosis in various types of cancer. The constitutive activation of STAT3 in cancer affects processes such as cell proliferation, apoptosis, metastasis, angiogenesis, and drug resistance. The importance of STAT3 in cancer makes it a potential therapeutic target. Various methods of directly and indirectly blocking STAT3 activity at different steps of the STAT3 pathway have been investigated. However, the outcome has been limited, mainly by the number of upstream proteins that can reactivate STAT3 or the relatively low specificity of the inhibitors. A new branch of molecules with significant therapeutic potential has emerged thanks to recent developments in the regulatory function of non-coding nucleic acids. Oligonucleotide-based therapeutics can silence target transcripts or edit genes, leading to the modification of gene expression profiles, causing cell death or restoring cell function. Moreover, they can reach untreatable targets, such as transcription factors. This review briefly describes oligonucleotide-based therapeutics that found application to target STAT3 activity in cancer. Additionally, this review comprehensively summarizes how the inhibition of STAT3 activity by nucleic acid-based therapeutics such as siRNA, shRNA, ASO, and ODN-decoy affected the therapy of different types of cancer in preclinical and clinical studies. Moreover, due to some limitations of oligonucleotide-based therapeutics, the importance of carriers that can deliver nucleic acid molecules to affect the STAT3 in cancer cells and cells of the tumor microenvironment (TME) was pointed out. Combining a high specificity of oligonucleotide-based therapeutics toward their targets and functionalized nanoparticles toward cell type can generate very efficient formulations.
Collapse
Affiliation(s)
- Sara Molenda
- Department of Cancer Immunology, Poznan University of Medical Sciences, 15 Garbary St., 61-866 Poznan, Poland; (S.M.); (A.S.); (A.F.); (P.L.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St., 61-866 Poznan, Poland
| | - Agata Sikorska
- Department of Cancer Immunology, Poznan University of Medical Sciences, 15 Garbary St., 61-866 Poznan, Poland; (S.M.); (A.S.); (A.F.); (P.L.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St., 61-866 Poznan, Poland
| | - Anna Florczak
- Department of Cancer Immunology, Poznan University of Medical Sciences, 15 Garbary St., 61-866 Poznan, Poland; (S.M.); (A.S.); (A.F.); (P.L.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St., 61-866 Poznan, Poland
| | - Patryk Lorenc
- Department of Cancer Immunology, Poznan University of Medical Sciences, 15 Garbary St., 61-866 Poznan, Poland; (S.M.); (A.S.); (A.F.); (P.L.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St., 61-866 Poznan, Poland
| | - Hanna Dams-Kozlowska
- Department of Cancer Immunology, Poznan University of Medical Sciences, 15 Garbary St., 61-866 Poznan, Poland; (S.M.); (A.S.); (A.F.); (P.L.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St., 61-866 Poznan, Poland
| |
Collapse
|
9
|
Trossmann VT, Lentz S, Scheibel T. Factors Influencing Properties of Spider Silk Coatings and Their Interactions within a Biological Environment. J Funct Biomater 2023; 14:434. [PMID: 37623678 PMCID: PMC10455157 DOI: 10.3390/jfb14080434] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/26/2023] Open
Abstract
Biomaterials are an indispensable part of biomedical research. However, although many materials display suitable application-specific properties, they provide only poor biocompatibility when implanted into a human/animal body leading to inflammation and rejection reactions. Coatings made of spider silk proteins are promising alternatives for various applications since they are biocompatible, non-toxic and anti-inflammatory. Nevertheless, the biological response toward a spider silk coating cannot be generalized. The properties of spider silk coatings are influenced by many factors, including silk source, solvent, the substrate to be coated, pre- and post-treatments and the processing technique. All these factors consequently affect the biological response of the environment and the putative application of the appropriate silk coating. Here, we summarize recently identified factors to be considered before spider silk processing as well as physicochemical characterization methods. Furthermore, we highlight important results of biological evaluations to emphasize the importance of adjustability and adaption to a specific application. Finally, we provide an experimental matrix of parameters to be considered for a specific application and a guided biological response as exemplarily tested with two different fibroblast cell lines.
Collapse
Affiliation(s)
- Vanessa T. Trossmann
- Chair of Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof.-Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany; (V.T.T.); (S.L.)
| | - Sarah Lentz
- Chair of Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof.-Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany; (V.T.T.); (S.L.)
| | - Thomas Scheibel
- Chair of Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof.-Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany; (V.T.T.); (S.L.)
- Bayreuth Center for Colloids and Interfaces (BZKG), University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bavarian Polymer Institute (BPI), University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuth Center for Molecular Biosciences (BZMB), University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuth Materials Center (BayMAT), University of Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Faculty of Medicine, University of Würzburg, Pleicherwall 2, 97070 Würzburg, Germany
| |
Collapse
|
10
|
Zhong R, Talebian S, Mendes BB, Wallace G, Langer R, Conde J, Shi J. Hydrogels for RNA delivery. NATURE MATERIALS 2023; 22:818-831. [PMID: 36941391 PMCID: PMC10330049 DOI: 10.1038/s41563-023-01472-w] [Citation(s) in RCA: 84] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 01/09/2023] [Indexed: 06/18/2023]
Abstract
RNA-based therapeutics have shown tremendous promise in disease intervention at the genetic level, and some have been approved for clinical use, including the recent COVID-19 messenger RNA vaccines. The clinical success of RNA therapy is largely dependent on the use of chemical modification, ligand conjugation or non-viral nanoparticles to improve RNA stability and facilitate intracellular delivery. Unlike molecular-level or nanoscale approaches, macroscopic hydrogels are soft, water-swollen three-dimensional structures that possess remarkable features such as biodegradability, tunable physiochemical properties and injectability, and recently they have attracted enormous attention for use in RNA therapy. Specifically, hydrogels can be engineered to exert precise spatiotemporal control over the release of RNA therapeutics, potentially minimizing systemic toxicity and enhancing in vivo efficacy. This Review provides a comprehensive overview of hydrogel loading of RNAs and hydrogel design for controlled release, highlights their biomedical applications and offers our perspectives on the opportunities and challenges in this exciting field of RNA delivery.
Collapse
Affiliation(s)
- Ruibo Zhong
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sepehr Talebian
- Faculty of Engineering, School of Chemical and Biomolecular Engineering, The University of Sydney, Sydney, New South Wales, Australia
- Nano Institute (Sydney Nano), The University of Sydney, Sydney, New South Wales, Australia
| | - Bárbara B Mendes
- ToxOmics, NOVA Medical School Faculdade de Ciências Médicas, NMS FCM, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Gordon Wallace
- ARC Centre of Excellence for Electromaterials Science, Intelligent Polymer Research Institute, AIIM, Innovation Campus, University of Wollongong, North Wollongong, New South Wales, Australia
| | - Robert Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - João Conde
- ToxOmics, NOVA Medical School Faculdade de Ciências Médicas, NMS FCM, Universidade NOVA de Lisboa, Lisbon, Portugal.
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Zhang X, Hai L, Gao Y, Yu G, Sun Y. Lipid nanomaterials-based RNA therapy and cancer treatment. Acta Pharm Sin B 2023; 13:903-915. [PMID: 36970213 PMCID: PMC10031258 DOI: 10.1016/j.apsb.2022.10.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/04/2022] [Accepted: 09/18/2022] [Indexed: 11/01/2022] Open
Abstract
We summarize the most important advances in RNA delivery and nanomedicine. We describe lipid nanoparticle-based RNA therapeutics and the impacts on the development of novel drugs. The fundamental properties of the key RNA members are described. We introduced recent advances in the nanoparticles to deliver RNA to defined targets, with a focus on lipid nanoparticles (LNPs). We review recent advances in biomedical therapy based on RNA drug delivery and state-of-the-art RNA application platforms, including the treatment of different types of cancer. This review presents an overview of current LNPs based RNA therapies in cancer treatment and provides deep insight into the development of future nanomedicines sophisticatedly combining the unparalleled functions of RNA therapeutics and nanotechnology.
Collapse
Affiliation(s)
- Xingcai Zhang
- School of Engineering and Applied Sciences, Harvard University, Cambridge 02138, MA, USA
| | - Luo Hai
- Central Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Yibo Gao
- Central Laboratory, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- Laboratory of Translational Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Guocan Yu
- Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Yingli Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
- University of Chinese Academy of Sciences, Beijing 100101, China
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing 100101, China
| |
Collapse
|
12
|
Gonzalez-Obeso C, Jane Hartzell E, Albert Scheel R, Kaplan DL. Delivering on the promise of recombinant silk-inspired proteins for drug delivery. Adv Drug Deliv Rev 2023; 192:114622. [PMID: 36414094 PMCID: PMC9812964 DOI: 10.1016/j.addr.2022.114622] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 11/06/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
Abstract
Effective drug delivery is essential for the success of a medical treatment. Polymeric drug delivery systems (DDSs) are preferred over systemic administration of drugs due to their protection capacity, directed release, and reduced side effects. Among the numerous polymer sources, silks and recombinant silks have drawn significant attention over the past decade as DDSs. Native silk is produced from a variety of organisms, which are then used as sources or guides of genetic material for heterologous expression or engineered designs. Recombinant silks bear the outstanding properties of natural silk, such as processability in aqueous solution, self-assembly, drug loading capacity, drug stabilization/protection, and degradability, while incorporating specific properties beneficial for their success as DDS, such as monodispersity and tailored physicochemical properties. Moreover, the on-demand inclusion of sequences that customize the DDS for the specific application enhances efficiency. Often, inclusion of a drug into a DDS is achieved by simple mixing or diffusion and stabilized by non-specific molecular interactions; however, these interactions can be improved by the incorporation of drug-binding peptide sequences. In this review we provide an overview of native sources for silks and silk sequences, as well as the design and formulation of recombinant silk biomaterials as drug delivery systems in a variety of formats, such as films, hydrogels, porous sponges, or particles.
Collapse
Affiliation(s)
- Constancio Gonzalez-Obeso
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, USA
| | - Emily Jane Hartzell
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, USA
| | - Ryan Albert Scheel
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, USA.
| |
Collapse
|
13
|
Goncalves AG, Hartzell EJ, Sullivan MO, Chen W. Recombinant protein polymer-antibody conjugates for applications in nanotechnology and biomedicine. Adv Drug Deliv Rev 2022; 191:114570. [PMID: 36228897 DOI: 10.1016/j.addr.2022.114570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/03/2022] [Accepted: 10/04/2022] [Indexed: 01/24/2023]
Abstract
Currently, there are over 100 antibody-based therapeutics on the market for the treatment of various diseases. The increasing importance of antibody treatment is further highlighted by the recent FDA emergency use authorization of certain antibody therapies for COVID-19 treatment. Protein-based materials have gained momentum for antibody delivery due to their biocompatibility, tunable chemistry, monodispersity, and straightforward synthesis and purification. In this review, we discuss progress in engineering the molecular features of protein-based biomaterials, in particular recombinant protein polymers, for introducing novel functionalities and enhancing the delivery properties of antibodies and related binding protein domains.
Collapse
Affiliation(s)
- Antonio G Goncalves
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States
| | - Emily J Hartzell
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States
| | - Millicent O Sullivan
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States.
| | - Wilfred Chen
- Department of Chemical and Biomolecular Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, United States.
| |
Collapse
|
14
|
Li Y, Champion JA. Self-assembling nanocarriers from engineered proteins: Design, functionalization, and application for drug delivery. Adv Drug Deliv Rev 2022; 189:114462. [PMID: 35934126 DOI: 10.1016/j.addr.2022.114462] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 06/09/2022] [Accepted: 07/15/2022] [Indexed: 01/24/2023]
Abstract
Self-assembling proteins are valuable building blocks for constructing drug nanocarriers due to their self-assembly behavior, monodispersity, biocompatibility, and biodegradability. Genetic and chemical modifications allow for modular design of protein nanocarriers with effective drug encapsulation, targetability, stimuli responsiveness, and in vivo half-life. Protein nanocarriers have been developed to deliver various therapeutic molecules including small molecules, proteins, and nucleic acids with proven in vitro and in vivo efficacy. This article reviews recent advances in protein nanocarriers that are not derived from natural protein nanostructures, such as protein cages or virus like particles. The protein nanocarriers described here are self-assembled from rationally or de novo designed recombinant proteins, as well as recombinant proteins complexed with other biomolecules, presenting properties that are unique from those of natural protein carriers. Design, functionalization, and therapeutic application of protein nanocarriers will be discussed.
Collapse
Affiliation(s)
- Yirui Li
- BioEngineering Program, Georgia Institute of Technology, United States
| | - Julie A Champion
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, 950 Atlantic Drive NW, Atlanta, GA 30332, United States; BioEngineering Program, Georgia Institute of Technology, United States.
| |
Collapse
|
15
|
Bittencourt DMDC, Oliveira P, Michalczechen-Lacerda VA, Rosinha GMS, Jones JA, Rech EL. Bioengineering of spider silks for the production of biomedical materials. Front Bioeng Biotechnol 2022; 10:958486. [PMID: 36017345 PMCID: PMC9397580 DOI: 10.3389/fbioe.2022.958486] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Spider silks are well known for their extraordinary mechanical properties. This characteristic is a result of the interplay of composition, structure and self-assembly of spider silk proteins (spidroins). Advances in synthetic biology have enabled the design and production of spidroins with the aim of biomimicking the structure-property-function relationships of spider silks. Although in nature only fibers are formed from spidroins, in vitro, scientists can explore non-natural morphologies including nanofibrils, particles, capsules, hydrogels, films or foams. The versatility of spidroins, along with their biocompatible and biodegradable nature, also placed them as leading-edge biological macromolecules for improved drug delivery and various biomedical applications. Accordingly, in this review, we highlight the relationship between the molecular structure of spider silk and its mechanical properties and aims to provide a critical summary of recent progress in research employing recombinantly produced bioengineered spidroins for the production of innovative bio-derived structural materials.
Collapse
Affiliation(s)
- Daniela Matias de C. Bittencourt
- Embrapa Genetic Resources and Biotechnology, National Institute of Science and Technology—Synthetic Biology, Brasília, DF, Brazil
| | - Paula Oliveira
- Department of Biology, Utah State University, Logan, UT, United States
| | | | - Grácia Maria Soares Rosinha
- Embrapa Genetic Resources and Biotechnology, National Institute of Science and Technology—Synthetic Biology, Brasília, DF, Brazil
| | - Justin A. Jones
- Department of Biology, Utah State University, Logan, UT, United States
| | - Elibio L. Rech
- Embrapa Genetic Resources and Biotechnology, National Institute of Science and Technology—Synthetic Biology, Brasília, DF, Brazil
| |
Collapse
|
16
|
Kelly G, Milligan JJ, Mastria EM, Kim S, Zelenetz SR, Dobbins J, Cai LY, Li X, Nair SK, Chilkoti A. Intratumoral delivery of brachytherapy and immunotherapy by a thermally triggered polypeptide depot. J Control Release 2022; 343:267-276. [PMID: 35077742 PMCID: PMC8960370 DOI: 10.1016/j.jconrel.2022.01.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/15/2022] [Accepted: 01/18/2022] [Indexed: 10/19/2022]
Abstract
Biomaterial-based approaches for a combination of radiotherapy and immunotherapy can improve outcomes in metastatic cancer through local delivery of both therapeutic modalities to the primary tumor to control local tumor growth and distant metastases. This study describes an injectable depot for sustained intratumoral (i.t.) delivery of an iodine-131 (131I) radionuclide and a CpG oligodeoxynucleotide immunostimulant, driven by the thermally sensitive phase transition behavior of elastin-like polypeptides (ELPs). We synthesized and characterized an ELP with an oligolysine tail (ELP-K12) that forms an electrostatic complex with CpG for delivery from an ELP depot and evaluated the ability of the complex to enhance local and systemic tumor control as a monotherapy and in combination with 131I-ELP brachytherapy. I.t delivery of CpG from an ELP-K12 depot dramatically prolongs i.t. retention to more than 21 days as compared to soluble CpG that is only retained within the tumor for <24 h. ELP-K12 also enhances CpG delivery by increasing cellular uptake of CpG to generate greater toll-like receptor 9 (TLR9) activation than CpG alone. I.t. treatment with an ELP-K12/CpG depot slows primary tumor growth and reduces lung metastases in a poorly immunogenic 4 T1 syngeneic breast cancer model whereas i.t treatment of CpG alone has no significant effect on primary tumor growth or metastases. Notably, a combination of 131I-ELP brachytherapy and ELP-K12/CpG delivered i.t. inhibited 4 T1 tumor growth and strongly decreased the development of lung metastases, leading to a synergistic improvement in mouse survival. These preclinical results demonstrate that injectable ELP depots may provide a useful approach for the delivery of combination radio- and immuno-therapy to treat metastatic disease.
Collapse
Affiliation(s)
- Garrett Kelly
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Joshua J. Milligan
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Eric M. Mastria
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Sarah Kim
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Stephanie R. Zelenetz
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Jarrett Dobbins
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Leon Y. Cai
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Xinghai Li
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA
| | - Smita K. Nair
- Department of Surgery, Duke University School of Medicine, 2301 Erwin Rd., DUMC Box 370, Durham, NC 27710, USA
| | - Ashutosh Chilkoti
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, 101 Science Dr., Campus Box 90281, Durham, NC 27708, USA.
| |
Collapse
|
17
|
Florczak A, Deptuch T, Kucharczyk K, Dams-Kozlowska H. Systemic and Local Silk-Based Drug Delivery Systems for Cancer Therapy. Cancers (Basel) 2021; 13:5389. [PMID: 34771557 PMCID: PMC8582423 DOI: 10.3390/cancers13215389] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 12/26/2022] Open
Abstract
For years, surgery, radiotherapy, and chemotherapy have been the gold standards to treat cancer, although continuing research has sought a more effective approach. While advances can be seen in the development of anticancer drugs, the tools that can improve their delivery remain a challenge. As anticancer drugs can affect the entire body, the control of their distribution is desirable to prevent systemic toxicity. The application of a suitable drug delivery platform may resolve this problem. Among other materials, silks offer many advantageous properties, including biodegradability, biocompatibility, and the possibility of obtaining a variety of morphological structures. These characteristics allow the exploration of silk for biomedical applications and as a platform for drug delivery. We have reviewed silk structures that can be used for local and systemic drug delivery for use in cancer therapy. After a short description of the most studied silks, we discuss the advantages of using silk for drug delivery. The tables summarize the descriptions of silk structures for the local and systemic transport of anticancer drugs. The most popular techniques for silk particle preparation are presented. Further prospects for using silk as a drug carrier are considered. The application of various silk biomaterials can improve cancer treatment by the controllable delivery of chemotherapeutics, immunotherapeutics, photosensitizers, hormones, nucleotherapeutics, targeted therapeutics (e.g., kinase inhibitors), and inorganic nanoparticles, among others.
Collapse
Affiliation(s)
- Anna Florczak
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; (A.F.); (T.D.); (K.K.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Tomasz Deptuch
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; (A.F.); (T.D.); (K.K.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Kamil Kucharczyk
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; (A.F.); (T.D.); (K.K.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Hanna Dams-Kozlowska
- Department of Cancer Immunology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; (A.F.); (T.D.); (K.K.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| |
Collapse
|
18
|
Abstract
RNA-based therapeutics have shown great promise in treating a broad spectrum of diseases through various mechanisms including knockdown of pathological genes, expression of therapeutic proteins, and programmed gene editing. Due to the inherent instability and negative-charges of RNA molecules, RNA-based therapeutics can make the most use of delivery systems to overcome biological barriers and to release the RNA payload into the cytosol. Among different types of delivery systems, lipid-based RNA delivery systems, particularly lipid nanoparticles (LNPs), have been extensively studied due to their unique properties, such as simple chemical synthesis of lipid components, scalable manufacturing processes of LNPs, and wide packaging capability. LNPs represent the most widely used delivery systems for RNA-based therapeutics, as evidenced by the clinical approvals of three LNP-RNA formulations, patisiran, BNT162b2, and mRNA-1273. This review covers recent advances of lipids, lipid derivatives, and lipid-derived macromolecules used in RNA delivery over the past several decades. We focus mainly on their chemical structures, synthetic routes, characterization, formulation methods, and structure-activity relationships. We also briefly describe the current status of representative preclinical studies and clinical trials and highlight future opportunities and challenges.
Collapse
Affiliation(s)
- Yuebao Zhang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Changzhen Sun
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Chang Wang
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Katarina E Jankovic
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
| | - Yizhou Dong
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Biomedical Engineering, The Center for Clinical and Translational Science, The Comprehensive Cancer Center, Dorothy M. Davis Heart & Lung Research Institute, Department of Radiation Oncology, The Ohio State University, Columbus, Ohio 43210, United States
| |
Collapse
|
19
|
Liu Y, Huang W, Meng M, Chen M, Cao C. Progress in the application of spider silk protein in medicine. J Biomater Appl 2021; 36:859-871. [PMID: 33853426 DOI: 10.1177/08853282211003850] [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: 11/17/2022]
Abstract
Spider silk protein has attracted much attention on account of its excellent mechanical properties, biodegradability, and biocompatibility. As the main protein component of spider silk, spidroin plays important role in spider spinning under natural circumstances and biomaterial application in medicine as well. Compare to the native spidroin which has a large molecular weight (>300 kDa) with highly repeat glycine and polyalanine regions, the recombinant spidroin was maintained the core amino motifs and much easier to collect. Here, we reviewed the application of recombinant spider silk protein eADF4(C16), major ampullate spidroin (MaSp), minor ampullate spidroin (MiSp), and the derivatives of recombinant spider silk protein in drug delivery system. Moreover, we also reviewed the application of spider silk protein in the field of alternative materials, repairing materials, wound dressing, surgical sutures along with advances in recombinant spider silk protein.
Collapse
Affiliation(s)
- Yi Liu
- 1Key Laboratories of Fine Chemicals and Surfactants in Sichuan Provincial Universities, School of Chemical Engineering, Sichuan University of Science & Engineering, Zigong, China
| | - Wei Huang
- 1Key Laboratories of Fine Chemicals and Surfactants in Sichuan Provincial Universities, School of Chemical Engineering, Sichuan University of Science & Engineering, Zigong, China
| | - Minsi Meng
- 1Key Laboratories of Fine Chemicals and Surfactants in Sichuan Provincial Universities, School of Chemical Engineering, Sichuan University of Science & Engineering, Zigong, China
| | - Minhui Chen
- 2Department of Neurosurgery, Zigong Fourth People's Hospital, Zigong, China
| | - Chengjian Cao
- 3Department of Clinical Laboratory, Zigong First People's Hospital, Zigong, China
| |
Collapse
|
20
|
Kucharczyk K, Kaczmarek K, Jozefczak A, Slachcinski M, Mackiewicz A, Dams-Kozlowska H. Hyperthermia treatment of cancer cells by the application of targeted silk/iron oxide composite spheres. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 120:111654. [PMID: 33545822 DOI: 10.1016/j.msec.2020.111654] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/21/2020] [Accepted: 10/14/2020] [Indexed: 10/23/2022]
Abstract
Magnetic iron oxide nanoparticles (IONPs) are one of the most extensively studied materials for theranostic applications. IONPs can be used for magnetic resonance imaging (MRI), delivery of therapeutics, and hyperthermia treatment. Silk is a biocompatible material and can be used for biomedical applications. Previously, we produced spheres made of H2.1MS1 bioengineered silk that specifically carried a drug to the Her2-overexpressing cancer cells. To confer biocompatibility and targeting properties to IONPs, we blended these particles with bioengineered spider silks. Three bioengineered silks (MS1Fe1, MS1Fe2, and MS1Fe1Fe2) functionalized with the adhesion peptides F1 and F2, were constructed and investigated to form the composite spheres with IONPs carrying a positive or negative charge. Due to its highest IONP content, MS1Fe1 silk was used to produce spheres from the H2.1MS1:MS1Fe silk blend to obtain a carrier with cell-targeting properties. Composite H2.1MS1:MS1Fe1/IONP spheres made of silks blended at different ratios were obtained. Although the increased content of MS1Fe1 silk in particles resulted in an increased affinity of the spheres to IONPs, it decreased the binding of the composite particles to cancer cells. The H2.1MS1:MS1Fe1 particles prepared at a ratio of 8:2 and loaded with IONPs exhibited the ability to bind to the targeted cancer cells similar to the control spheres without IONPs. Moreover, when exposed to the alternating magnetic field, these particles generated 2.5 times higher heat. They caused an almost three times higher percentage of apoptosis in cancer cells than the control particles. The blending of silks enabled the generation of cancer-targeting spheres with a high affinity for iron oxide nanoparticles, which can be used for anti-cancer hyperthermia therapy.
Collapse
Affiliation(s)
- Kamil Kucharczyk
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| | - Katarzyna Kaczmarek
- Chair of Acoustics, Faculty of Physics, Adam Mickiewicz University, Poznan, Poland
| | - Arkadiusz Jozefczak
- Chair of Acoustics, Faculty of Physics, Adam Mickiewicz University, Poznan, Poland
| | - Mariusz Slachcinski
- Faculty of Chemical Technology, Institute of Chemistry and Technical Electrochemistry, Poznan University of Technology, Poznan, Poland
| | - Andrzej Mackiewicz
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland
| | - Hanna Dams-Kozlowska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poland; Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan, Poland.
| |
Collapse
|
21
|
Florczak A, Grzechowiak I, Deptuch T, Kucharczyk K, Kaminska A, Dams-Kozlowska H. Silk Particles as Carriers of Therapeutic Molecules for Cancer Treatment. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E4946. [PMID: 33158060 PMCID: PMC7663281 DOI: 10.3390/ma13214946] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/30/2020] [Accepted: 10/31/2020] [Indexed: 12/15/2022]
Abstract
Although progress is observed in cancer treatment, this disease continues to be the second leading cause of death worldwide. The current understanding of cancer indicates that treating cancer should not be limited to killing cancer cells alone, but that the target is the complex tumor microenvironment (TME). The application of nanoparticle-based drug delivery systems (DDS) can not only target cancer cells and TME, but also simultaneously resolve the severe side effects of various cancer treatment approaches, leading to more effective, precise, and less invasive therapy. Nanoparticles based on proteins derived from silkworms' cocoons (like silk fibroin and sericins) and silk proteins from spiders (spidroins) are intensively explored not only in the oncology field. This natural-derived material offer biocompatibility, biodegradability, and simplicity of preparation methods. The protein-based material can be tailored for size, stability, drug loading/release kinetics, and functionalized with targeting ligands. This review summarizes the current status of drug delivery systems' development based on proteins derived from silk fibroin, sericins, and spidroins, which application is focused on systemic cancer treatment. The nanoparticles that deliver chemotherapeutics, nucleic acid-based therapeutics, natural-derived agents, therapeutic proteins or peptides, inorganic compounds, as well as photosensitive molecules, are introduced.
Collapse
Affiliation(s)
- Anna Florczak
- Department of Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; (A.F.); (I.G.); (T.D.); (K.K.); (A.K.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Inga Grzechowiak
- Department of Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; (A.F.); (I.G.); (T.D.); (K.K.); (A.K.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Tomasz Deptuch
- Department of Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; (A.F.); (I.G.); (T.D.); (K.K.); (A.K.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Kamil Kucharczyk
- Department of Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; (A.F.); (I.G.); (T.D.); (K.K.); (A.K.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| | - Alicja Kaminska
- Department of Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; (A.F.); (I.G.); (T.D.); (K.K.); (A.K.)
| | - Hanna Dams-Kozlowska
- Department of Medical Biotechnology, Poznan University of Medical Sciences, 61-866 Poznan, Poland; (A.F.); (I.G.); (T.D.); (K.K.); (A.K.)
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-866 Poznan, Poland
| |
Collapse
|
22
|
Chambre L, Martín-Moldes Z, Parker RN, Kaplan DL. Bioengineered elastin- and silk-biomaterials for drug and gene delivery. Adv Drug Deliv Rev 2020; 160:186-198. [PMID: 33080258 PMCID: PMC7736173 DOI: 10.1016/j.addr.2020.10.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/30/2020] [Accepted: 10/16/2020] [Indexed: 12/11/2022]
Abstract
Advances in medical science have led to diverse new therapeutic modalities, as well as enhanced understanding of the progression of various disease states. These findings facilitate the design and development of more customized and exquisite drug delivery systems that aim to improve therapeutic indices of drugs to treat a variety of conditions. Synthetic polymer-based drug carriers have often been the focus of such research. However, these structures suffer from challenges with heterogeneity of the starting material, limited chemical features, complex functionalization methods, and in some cases a lack of biocompatibility. Consequently, protein-based polymers have garnered much attention in recent years due to their monodisperse features, ease of production and functionalization, and biocompatibility. Genetic engineering techniques enable the advancement of protein-based drug delivery systems with finely tuned physicochemical properties, and thus an expanded level of customization unavailable with synthetic polymers. Of these genetically engineered proteins, elastin-like proteins (ELP), silk-like proteins (SLP), and silk-elastin-like proteins (SELP) provide a unique set of alternatives for designing drug delivery systems due to their inherent chemical and physical properties and ease of engineering afforded by recombinant DNA technologies. In this review we examine the advantages of genetically engineered drug delivery systems with emphasis on ELP and SLP constructions. Methods for fabrication and relevant biomedical applications will also be discussed.
Collapse
Affiliation(s)
- Laura Chambre
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Zaira Martín-Moldes
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - Rachael N Parker
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St, Medford, MA 02155, USA.
| |
Collapse
|
23
|
Abstract
Spider web proteins are unique materials created by nature that, considering the combination of their properties, do not have analogues among natural or human-created materials. Obtaining significant amounts of these proteins from natural sources is not feasible. Biotechnological manufacturing in heterological systems is complicated by the very high molecular weight of spidroins and their specific amino acid composition. Obtaining recombinant analogues of spidroins in heterological systems, mainly in bacteria and yeast, has become a compromise solution. Because they can self-assemble, these proteins can form various materials, such as fibers, films, 3D-foams, hydrogels, tubes, and microcapsules. The effectiveness of spidroin hydrogels in deep wound healing, as 3D scaffolds for bone tissue regeneration and as oriented fibers for axon growth and nerve tissue regeneration, was demonstrated in animal models. The possibility to use spidroin micro- and nanoparticles for drug delivery was demonstrated, including the use of modified spidroins for virus-free DNA delivery into animal cell nuclei. In the past few years, significant interest has arisen concerning the use of these materials as biocompatible and biodegradable soft optics to construct photonic crystal super lenses and fiber optics and as soft electronics to use in triboelectric nanogenerators. This review summarizes the latest achievements in the field of spidroin production, the creation of materials based on them, the study of these materials as a scaffold for the growth, proliferation, and differentiation of various types of cells, and the prospects for using these materials for medical applications (e.g., tissue engineering, drug delivery, coating medical devices), soft optics, and electronics. Accumulated data suggest the use of recombinant spidroins in medical practice in the near future.
Collapse
Affiliation(s)
- Vladimir G Debabov
- State Research Institute for Genetics and Selection of Industrial Microorganisms of National Research Center "Kurchatov Institute" (NRC "Kurchatov Institute"-GOSNIIGENETIKA), Moscow 117545, Russia
| | - Vladimir G Bogush
- State Research Institute for Genetics and Selection of Industrial Microorganisms of National Research Center "Kurchatov Institute" (NRC "Kurchatov Institute"-GOSNIIGENETIKA), Moscow 117545, Russia
| |
Collapse
|
24
|
Kucharczyk K, Florczak A, Deptuch T, Penderecka K, Jastrzebska K, Mackiewicz A, Dams-Kozlowska H. Drug affinity and targeted delivery: double functionalization of silk spheres for controlled doxorubicin delivery into Her2-positive cancer cells. J Nanobiotechnology 2020; 18:56. [PMID: 32228620 PMCID: PMC7106823 DOI: 10.1186/s12951-020-00609-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/16/2020] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND The optimal drug delivery system should be biocompatible, biodegradable, and allow the sustained release of the drug only after it reaches the target cells. Silk, as a natural polymer, is a great candidate for building drug carriers. Genetically engineered silks offer the possibility of functionalization. Previously, we characterized bioengineered silk spheres that were functionalized with H2.1 peptide that selectively delivered a drug to Her2-positive cancer cells. However, drug leakage from the silk spheres showed the need for improved control. RESULTS To control the drug loading and release, we designed and produced functional silk (DOXMS2) that contains a DOX peptide with an affinity for doxorubicin. The DOXMS2 spheres showed the decreased release of doxorubicin compared with MS2 particles. Next, the DOXMS2 silk was blended with the H2.1MS1 polymer to improve the control of doxorubicin binding and release into Her2-positive cancer cells. The H2.1MS1:DOXMS2 particles showed the highest doxorubicin-loading capacity and binding per cell, which resulted in the highest cytotoxic effect compared with that of other sphere variants. Since drug release at a pH of 7.4 from the blended H2.1MS1:DOXMS2 particles was significantly lower than from blended spheres without DOXMS2 silk, this indicated that such particles could control the release of the drug into the circulatory system before the carrier reached the tumor site. CONCLUSIONS This strategy, which is based on the blending of silks, allows for the generation of particles that deliver drugs in a controlled manner.
Collapse
Affiliation(s)
- Kamil Kucharczyk
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-688 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-688 Poznan, Poland
| | - Anna Florczak
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-688 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-688 Poznan, Poland
| | - Tomasz Deptuch
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-688 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-688 Poznan, Poland
| | - Karolina Penderecka
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-688 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-688 Poznan, Poland
| | - Katarzyna Jastrzebska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-688 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-688 Poznan, Poland
| | - Andrzej Mackiewicz
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-688 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-688 Poznan, Poland
| | - Hanna Dams-Kozlowska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-688 Poznan, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 15 Garbary St, 61-688 Poznan, Poland
| |
Collapse
|
25
|
Saric M, Scheibel T. Engineering of silk proteins for materials applications. Curr Opin Biotechnol 2019; 60:213-220. [DOI: 10.1016/j.copbio.2019.05.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 05/07/2019] [Indexed: 11/26/2022]
|
26
|
Tomeh MA, Hadianamrei R, Zhao X. Silk Fibroin as a Functional Biomaterial for Drug and Gene Delivery. Pharmaceutics 2019; 11:E494. [PMID: 31561578 PMCID: PMC6835371 DOI: 10.3390/pharmaceutics11100494] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2019] [Revised: 09/24/2019] [Accepted: 09/24/2019] [Indexed: 01/31/2023] Open
Abstract
Silk is a natural polymer with unique physicochemical and mechanical properties which makes it a desirable biomaterial for biomedical and pharmaceutical applications. Silk fibroin (SF) has been widely used for preparation of drug delivery systems due to its biocompatibility, controllable degradability and tunable drug release properties. SF-based drug delivery systems can encapsulate and stabilize various small molecule drugs as well as large biological drugs such as proteins and DNA to enhance their shelf lives and control the release to enhance their circulation time in the blood and thus the duration of action. Understanding the properties of SF and the potential ways of manipulating its structure to modify its physicochemical and mechanical properties allows for preparation of modulated drug delivery systems with desirable efficacies. This review will discuss the properties of SF material and summarize the recent advances of SF-based drug and gene delivery systems. Furthermore, conjugation of the SF to other biomolecules or polymers for tissue-specific drug delivery will also be discussed.
Collapse
Affiliation(s)
- Mhd Anas Tomeh
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK.
| | - Roja Hadianamrei
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK.
| | - Xiubo Zhao
- Department of Chemical and Biological Engineering, University of Sheffield, Sheffield S1 3JD, UK.
- School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
27
|
Ulkoski D, Bak A, Wilson JT, Krishnamurthy VR. Recent advances in polymeric materials for the delivery of RNA therapeutics. Expert Opin Drug Deliv 2019; 16:1149-1167. [PMID: 31498013 DOI: 10.1080/17425247.2019.1663822] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: The delivery of nucleic acid therapeutics through non-viral carriers face multiple biological barriers that reduce their therapeutic efficiency. Despite great progress, there remains a significant technological gap that continues to limit clinical translation of these nanocarriers. A number of polymeric materials are being exploited to efficiently deliver nucleic acids and achieve therapeutic effects. Areas covered: We discuss the recent advances in the polymeric materials for the delivery of nucleic acid therapeutics. We examine the use of common polymer architectures and highlight the challenges that exist for their development from bench side to clinic. We also provide an overview of the most notable improvements made to circumvent such challenges, including structural modification and stimuli-responsive approaches, for safe and effective nucleic acid delivery. Expert opinion: It has become apparent that a universal carrier that follows 'one-size' fits all model cannot be expected for delivery of all nucleic acid therapeutics. Carriers need to be designed to exhibit sensitivity and specificity toward individual targets diseases/indications, and relevant subcellular compartments, each of which possess their own unique challenges. The ability to devise synthetic methods that control the molecular architecture enables the future development that allow for the construction of 'intelligent' designs.
Collapse
Affiliation(s)
- David Ulkoski
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca , Boston , USA
| | - Annette Bak
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca , Gothenburg , Sweden
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University , Nashville , TN , USA
| | | |
Collapse
|
28
|
Florczak A, Mackiewicz A, Dams-Kozlowska H. Cellular uptake, intracellular distribution and degradation of Her2-targeting silk nanospheres. Int J Nanomedicine 2019; 14:6855-6865. [PMID: 32021156 PMCID: PMC6716583 DOI: 10.2147/ijn.s217854] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 07/27/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The development of nanocarrier technology has attracted great interest in the last decade. Biodegradable spheres made of functionalized silk have considerable potential to be used as drug delivery systems for cancer treatment. A targeting ligand displayed at the surface of a carrier, with a specific affinity towards a particular receptor, can further enhance the accumulation and uptake of nanoparticles at the site of a tumor. MATERIALS AND METHODS The hybrid constructs were obtained by adding a Her2-binding peptide (H2.1) to MS1 and MS2 bioengineered silks based on the MaSp1 and MaSp2 proteins from N. clavipes, respectively. The H2.1MS1 and H2.1MS2 proteins were blended at a weight ratio of 8:2. Stable silk particles were formed by mixing a soluble protein with potassium phosphate using a micromixing technique. We used specific inhibitors of endocytosis to determine the cellular uptake pathway of the silk nanoparticles in human Her2-positive breast cancer cells. The subcellular distribution of silk particles was investigated by evaluating the signal colocalization with organelle-specific tracker. Moreover, lysosomal and exosomal inhibitors were implemented to evaluate their impact on the silk spheres behavior and degradation. RESULTS The functionalized spheres were specifically taken up by Her2-positive cancer cells. Silk particles facilitated the entry into cells through both the clathrin- and caveola-dependent pathways of endocytosis. Upon entering the cells, the particles accumulated in the lysosomes, where intracellular degradation occurred. CONCLUSIONS The present study demonstrated directly that the lysosomal function was essential for silk-based carrier elimination. The degradation of the carrier is of great importance to develop an optimal drug delivery system.
Collapse
Affiliation(s)
- Anna Florczak
- Department of Medical Biotechnology, Poznan University of Medical Sciences, Poznan60-806, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan61-866, Poland
| | - Andrzej Mackiewicz
- Department of Medical Biotechnology, Poznan University of Medical Sciences, Poznan60-806, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan61-866, Poland
| | - Hanna Dams-Kozlowska
- Department of Medical Biotechnology, Poznan University of Medical Sciences, Poznan60-806, Poland
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, Poznan61-866, Poland
| |
Collapse
|
29
|
Kolenda T, Przybyła W, Kapałczyńska M, Teresiak A, Zajączkowska M, Bliźniak R, Lamperska KM. Tumor microenvironment - Unknown niche with powerful therapeutic potential. Rep Pract Oncol Radiother 2018; 23:143-153. [PMID: 29760589 PMCID: PMC5948324 DOI: 10.1016/j.rpor.2018.01.004] [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: 03/20/2017] [Revised: 11/20/2017] [Accepted: 01/20/2018] [Indexed: 12/25/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCC) are in a group of cancers that are the most resistant to treatment. The survival rate of HNSCC patients has been still very low since last 20 years. The existence of relationship between oncogenic and surrounding cells is probably the reason for a poor response to treatment. Fibroblasts are an important element of tumor stroma which increases tumor cells ability to proliferate. Another highly resistance, tumorigenic and metastatic cell population in tumor microenvironment are cancer initiating cells (CICs). The population of cancer initiating cells can be found regardless of differentiation status of cancer and they seem to be crucial for HNSCC development. In this review, we describe the current state of knowledge about HNSCC biological and physiological tumor microenvironment.
Collapse
Affiliation(s)
- Tomasz Kolenda
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Poland
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Weronika Przybyła
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Department of Pediatric Research, Division of Pediatric and Adolescent Medicine, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Marta Kapałczyńska
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Department of Gastroenterology and Hepatology, Charite University Medicine Berlin, Berlin, Germany
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Anna Teresiak
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
| | - Maria Zajączkowska
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, Poznan, Poland
| | - Renata Bliźniak
- Laboratory of Cancer Genetic, Greater Poland Cancer Centre, Poznan, Poland
| | | |
Collapse
|
30
|
Jastrzebska K, Florczak A, Kucharczyk K, Lin Y, Wang Q, Mackiewicz A, Kaplan DL, Dams-Kozlowska H. Delivery of chemotherapeutics using spheres made of bioengineered spider silks derived from MaSp1 and MaSp2 proteins. Nanomedicine (Lond) 2018; 13:439-454. [PMID: 29338625 PMCID: PMC5810845 DOI: 10.2217/nnm-2017-0276] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 12/11/2017] [Indexed: 01/16/2023] Open
Abstract
AIM Analysis of the properties and chemotherapeutics delivery potential of spheres made of bioengineered spider silks MS1 and MS2. MATERIALS & METHODS MS1 and MS2 derived from Nephila clavipes dragline silks - MaSp1 and MaSp2, respectively - formed spheres that were compared in terms of physicochemical properties, cytotoxicity and loading/release of chemotherapeutics. RESULTS MS2 spheres were more dispersed, smaller, of solid core, of higher beta-sheet structure content, and of opposite (negative) charge than MS1 spheres. Preloaded MS2 showed greater applicability for mitoxantrone, while postloaded for etoposide delivery compared with MS1 spheres. However, MS1 spheres were a better choice for doxorubicin delivery than MS2. CONCLUSION Bioengineered silks can be tailored to develop a system with optimal drug loading and release properties.
Collapse
Affiliation(s)
- Katarzyna Jastrzebska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61–688 Poznan, Poland
- NanoBioMedical Centre, Adam Mickiewicz University, 61–614 Poznan, Poland
| | - Anna Florczak
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61–688 Poznan, Poland
- NanoBioMedical Centre, Adam Mickiewicz University, 61–614 Poznan, Poland
| | - Kamil Kucharczyk
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61–688 Poznan, Poland
| | - Yinnan Lin
- Biomedical Engineering Department, Tufts University, Medford, MA 02155, USA
| | - Qin Wang
- Biomedical Engineering Department, Tufts University, Medford, MA 02155, USA
| | - Andrzej Mackiewicz
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61–688 Poznan, Poland
- BioContract Sp. z o.o., 61-051 Poznan, Poland
- Department of Diagnostics & Cancer Immunology, Greater Poland Cancer Centre, 61–688 Poznan, Poland
| | - David L Kaplan
- Biomedical Engineering Department, Tufts University, Medford, MA 02155, USA
| | - Hanna Dams-Kozlowska
- Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61–688 Poznan, Poland
- Department of Diagnostics & Cancer Immunology, Greater Poland Cancer Centre, 61–688 Poznan, Poland
| |
Collapse
|
31
|
Xia Y, Wang C, Xu T, Li Y, Guo M, Lin Z, Zhao M, Zhu B. Targeted delivery of HES5-siRNA with novel polypeptide-modified nanoparticles for hepatocellular carcinoma therapy. RSC Adv 2018; 8:1917-1926. [PMID: 35542585 PMCID: PMC9077277 DOI: 10.1039/c7ra12461a] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/15/2017] [Indexed: 12/18/2022] Open
Abstract
For actively targeted delivery of small interfering RNA (siRNA) to solid tumors, we fabricated functionalized selenium nanoparticles (SeNPs) decorated with the polypeptide RGDfC. Herein, RGDfC was used as tumor-targeted moiety and installed onto the surface of SeNPs to enhance the cellular uptake. RGDfC-SeNPs@siRNA were internalized into the HepG2 cell mainly through clathrin-mediated endocytosis. The active efficacy of the RGDfC-SeNPs@siRNA was confirmed via gene silencing assay, MTT assay and flow cytometry analysis. Owing to the tumor-targeting effect of RGDfC, RGDfC-SeNPs@siRNA achieved an obvious improvement in gene silencing ability, which led to significant growth inhibition of HepG2 cells. Furthermore, treatment with RGDfC-SeNPs@siRNA resulted in greater antitumor efficacy than lipofectamine 2000@siRNA in vitro and in vivo. In addition, the RGDfC-SeNPs@siRNA was almost non-toxic to the key organs of mice. In sum, these findings provide an alternative therapeutic route for targeted cancer treatments. A novel polypeptide RGDfC-modified selenium nanoparticle was fabricated to selectively deliver HES5-siRNA to tumors for hepatocellular carcinoma therapy.![]()
Collapse
Affiliation(s)
- Yu Xia
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Changbing Wang
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Tiantian Xu
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Yinghua Li
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Min Guo
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Zhengfang Lin
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Mingqi Zhao
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| | - Bing Zhu
- Guangzhou Women and children's Medical center
- Guangzhou
- P. R. China
| |
Collapse
|
32
|
Deptuch T, Dams-Kozlowska H. Silk Materials Functionalized via Genetic Engineering for Biomedical Applications. MATERIALS (BASEL, SWITZERLAND) 2017; 10:E1417. [PMID: 29231863 PMCID: PMC5744352 DOI: 10.3390/ma10121417] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/21/2022]
Abstract
The great mechanical properties, biocompatibility and biodegradability of silk-based materials make them applicable to the biomedical field. Genetic engineering enables the construction of synthetic equivalents of natural silks. Knowledge about the relationship between the structure and function of silk proteins enables the design of bioengineered silks that can serve as the foundation of new biomaterials. Furthermore, in order to better address the needs of modern biomedicine, genetic engineering can be used to obtain silk-based materials with new functionalities. Sequences encoding new peptides or domains can be added to the sequences encoding the silk proteins. The expression of one cDNA fragment indicates that each silk molecule is related to a functional fragment. This review summarizes the proposed genetic functionalization of silk-based materials that can be potentially useful for biomedical applications.
Collapse
Affiliation(s)
- Tomasz Deptuch
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-688 Poznan, Poland.
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-688 Poznan, Poland.
| | - Hanna Dams-Kozlowska
- Department of Cancer Immunology, Chair of Medical Biotechnology, Poznan University of Medical Sciences, 61-688 Poznan, Poland.
- Department of Diagnostics and Cancer Immunology, Greater Poland Cancer Centre, 61-688 Poznan, Poland.
| |
Collapse
|