1
|
Gupta J, Sharma G. Nanogel: A versatile drug delivery system for the treatment of various diseases and their future perspective. Drug Deliv Transl Res 2025; 15:455-482. [PMID: 39103593 DOI: 10.1007/s13346-024-01684-w] [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] [Accepted: 07/23/2024] [Indexed: 08/07/2024]
Abstract
Nanogel (NG) drug delivery systems have emerged as promising tools for targeted and controlled drug release, revolutionizing treatment approaches across various diseases. Their unique physicochemical properties, such as nano size, high surface area, biocompatibility, stability, and tunable drug release, make them ideal carriers for a wide range of therapeutic agents. Nanogels (NGs), characterized by their 3D network of crosslinked polymers, offer unique edges like high drug loading capacity, controlled release, and targeted delivery. Additionally, the diverse applications of NGs in medical therapeutics highlight their versatility and potential impact on improving patient outcomes. Their application spans cancer treatment, infectious diseases, and chronic conditions, allowing for precise drug delivery to specific tissues or cells, minimizing side effects, and enhancing therapeutic efficacy. Despite their potential, challenges such as scalability, manufacturing reproducibility, and regulatory hurdles must be addressed. Achieving clinical translation requires overcoming these obstacles to ensure therapeutic payloads' safe and efficient delivery. Strategies such as surface modification and incorporating stimuli-responsive elements enhanced NG performance and addressed specific therapeutic challenges. Advances in nanotechnology, biomaterials, and targeted drug design offer opportunities to improve the performance of NGs and address current limitations. Tailoring NGs for exploring combination therapies and integrating diagnostics for real-time monitoring represent promising avenues for future research. In conclusion, NG drug delivery systems have demonstrated tremendous potential in diverse disease applications. Overcoming challenges and leveraging emerging technologies will pave the way for their widespread clinical implementation, ushering in a new era of precision medicine and improved patient care.
Collapse
Affiliation(s)
- Jitendra Gupta
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India.
| | - Gaurang Sharma
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, Uttar Pradesh, India
| |
Collapse
|
2
|
Peng N, Du Y, Liu J, Li D, Li Y, Deng K, Li L, Jia P, Min J, Lin J. Injectable Polydopamine Nanoparticle-Incorporated Hydrogels for Antiangiogenesis and Stimulating Tumoricidal Immunity to Inhibit Metastasis and Recurrence Postresection. ACS APPLIED MATERIALS & INTERFACES 2024; 16:64447-64462. [PMID: 39540317 DOI: 10.1021/acsami.4c10363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Surgical resection is still the main means for clinical treatments of breast cancer, but the postoperative immunosuppressive microenvironment and neoangiogenesis of the residual tumors easily lead to tumor metastasis and recurrence, which will further endanger patients' lives. The combination of antiangiogenic therapy and immunotherapy may promote the mutually reinforced cycle of immune reprogramming and vascular normalization to avoid tumor metastasis and recurrence. Herein, we prepared polydopamine nanoparticles for improving tissue adhesion and enriching tumor-associated antigens. This nanoregulator together with regorafenib (REG) was further incorporated into a hydrogel developed from grafting adipic acid dihydrazide onto 2,2,6,6-tetramethylpiperidine-1-oxyl radical oxidized chitin and oxidized hyaluronic acid, which was injectable at the cavity after subcutaneous tumor surgery with good mechanical properties and degradability. The system showed long-term release of REG. After combining with anti-PD-L1, the hydrogel applied to the surgical wound exhibited a reduction in tumor metastasis and recurrence. This effect was achieved by suppressing angiogenesis and enhancing antitumor immunity, characterized by increased levels of effector T lymphocytes and activation of dendritic cells within tumors, spleens, and draining lymph nodes. The injectable hydrogel offers a promising strategy for postoperative management aimed at preventing tumor metastasis and recurrence.
Collapse
Affiliation(s)
- Na Peng
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P. R. China
| | - Yijing Du
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P. R. China
| | - Jinhong Liu
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| | - Danyang Li
- College of Textiles and Clothing, Yancheng Institute of Technology, Yancheng 224051, Jiangsu, P. R. China
| | - Yonghao Li
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| | - Kai Deng
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan 430071, P. R. China
| | - Lewei Li
- Key Laboratory of Coal Conversion and New Carbon Materials of Hubei Province, School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, Hubei, P. R. China
| | - Peizhi Jia
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| | - Juan Min
- Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan 430071, Hubei, P. R. China
| | - Jiumao Lin
- Academy of Integrative Medicine, Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, P. R. China
| |
Collapse
|
3
|
Kawasaki R, Miura Y, Kono N, Fujita S, Yamana K, Ikeda A. Boron Agent Delivery Platforms Based on Natural Products for Boron Neutron Capture Therapy. ChemMedChem 2024; 19:e202400323. [PMID: 38830821 DOI: 10.1002/cmdc.202400323] [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: 04/30/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/05/2024]
Abstract
Boron neutron capture therapy (BNCT) is one of the most promising modalities for cancer treatment due to its minimal invasiveness. Although two types of boron agents are clinically used, several issues persist in their delivery, including poor water solubility, instability in aqueous media, selectivity toward cancer cells, accumulation in cancer cells, retention time in tumor tissue, and efficiency in achieving the boron neutron capture reaction. Addressing these challenges, numerous groups have explored various boron agents to enhance the therapeutic benefits of BNCT. This review summarizes delivery platforms based on natural products for BNCT.
Collapse
Affiliation(s)
- Riku Kawasaki
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| | - Yamato Miura
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| | - Nanami Kono
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| | - Seiya Fujita
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| | - Keita Yamana
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| | - Atsushi Ikeda
- Program of Applied Chemistry, Graduate School of Advanced Science and Engineering, Hiroshima University, 1-4-1 Kagamiyama, Higashi-Hiroshima, 739-8527, Japan
| |
Collapse
|
4
|
Kareemi AF, Likhitkar S. Applications and advancements of polysaccharide-based nanostructures for enhanced drug delivery. Colloids Surf B Biointerfaces 2024; 238:113883. [PMID: 38615389 DOI: 10.1016/j.colsurfb.2024.113883] [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: 02/01/2024] [Revised: 03/20/2024] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
Growing demand for highly effective, site-specific delivery of pharmaceuticals and nutraceuticals using nano-sized carriers has prompted increased scrutiny of carrier biocompatibility and biodegradability. To address these concerns, biodegradable natural polymers have emerged as a transformative domain, offering non-toxic, precisely targetable carriers capable of finely modulating cargo pharmacokinetics while generating innocuous decomposition by-products. This comprehensive review illuminates the emergence of polysaccharide-based nanoparticulate drug delivery systems. These systems establish an interactive interface between drug and targeted organs, guided by strategic modifications to polysaccharide backbones, which facilitate the creation of morphologically, constitutionally, and characteristically vibrant nanostructures through various fabrication routes, underpinning their pivotal role in biomedical applications. Advancements crucial to enhancing polysaccharide-based drug delivery, such as surface modifications and bioinspired modifications for enhanced targeting, and stimuli-responsive release, strategies to overcome biological barriers, enhance tumor penetration, and optimize therapeutic outcomes are highlighted. This review also examines some potent challenges, and the contemporary way out of them, and discusses future perspectives in the field.
Collapse
Affiliation(s)
- Asra Fatimah Kareemi
- Department of Chemistry, St. Aloysius College (Autonomous), Jabalpur, Madhya Pradesh 482001, India
| | - Sweta Likhitkar
- Department of Chemistry, St. Aloysius College (Autonomous), Jabalpur, Madhya Pradesh 482001, India.
| |
Collapse
|
5
|
Yamamoto K, Sawada SI, Shindo S, Nakamura S, Kwon YM, Kianinejad N, Vardar S, Hernandez M, Akiyoshi K, Kawai T. Cationic Glucan Dendrimer Gel-Mediated Local Delivery of Anti-OC-STAMP-siRNA for Treatment of Pathogenic Bone Resorption. Gels 2024; 10:377. [PMID: 38920924 PMCID: PMC11202495 DOI: 10.3390/gels10060377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/17/2024] [Accepted: 05/28/2024] [Indexed: 06/27/2024] Open
Abstract
Osteoclast stimulatory transmembrane protein (OC-STAMP) plays a pivotal role in the promotion of cell fusion during osteoclast differentiation (osteoclastogenesis) in the context of pathogenic bone resorption. Thus, it is plausible that the suppression of OC-STAMP through a bioengineering approach could lead to the development of an effective treatment for inflammatory bone resorptive diseases with minimum side effects. Here, we synthesized two types of spermine-bearing (Spe) cationic glucan dendrimer (GD) gels (with or without C12) as carriers of short interfering RNA (siRNA) to silence OC-STAMP. The results showed that amphiphilic C12-GD-Spe gel was more efficient in silencing OC-STAMP than GD-Spe gel and that the mixture of anti-OC-STAMP siRNA/C12-GD-Spe significantly downregulated RANKL-induced osteoclastogenesis. Also, local injection of anti-OC-STAMP-siRNA/C12-GD-Spe could attenuate bone resorption induced in a mouse model of periodontitis. These results suggest that OC-STAMP is a promising target for the development of a novel bone regenerative therapy and that C12-GD-Spe gel provides a new nanocarrier platform of gene therapies for osteolytic disease.
Collapse
Affiliation(s)
- Kenta Yamamoto
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.S.); (S.N.); (T.K.)
| | - Shin-Ichi Sawada
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 605-0981, Japan; (S.-I.S.); (K.A.)
- Synergy Institute for Futuristic Mucosal Vaccine Research and Development, Chiba University, Chiba 260-8670, Japan
| | - Satoru Shindo
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.S.); (S.N.); (T.K.)
| | - Shin Nakamura
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.S.); (S.N.); (T.K.)
| | - Young M. Kwon
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA (N.K.)
| | - Nazanin Kianinejad
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA (N.K.)
| | - Saynur Vardar
- Department of Periodontology, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.V.); (M.H.)
| | - Maria Hernandez
- Department of Periodontology, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.V.); (M.H.)
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 605-0981, Japan; (S.-I.S.); (K.A.)
| | - Toshihisa Kawai
- Department of Oral Science and Translational Research, College of Dental Medicine, Nova Southeastern University, Fort Lauderdale, FL 33328, USA; (S.S.); (S.N.); (T.K.)
| |
Collapse
|
6
|
Prasher P, Sharma M, Agarwal V, Singh SK, Gupta G, Dureja H, Dua K. Cationic cycloamylose based nucleic acid nanocarriers. Chem Biol Interact 2024; 395:111000. [PMID: 38614318 DOI: 10.1016/j.cbi.2024.111000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 04/02/2024] [Accepted: 04/07/2024] [Indexed: 04/15/2024]
Abstract
Nucleic acid delivery by viral and non-viral methods has been a cornerstone for the contemporary gene therapy aimed at correcting the defective genes, replacing of the missing genes, or downregulating the expression of anomalous genes is highly desirable for the management of various diseases. Ostensibly, it becomes paramount for the delivery vectors to intersect the biological barriers for accessing their destined site within the cellular environment. However, the lipophilic nature of biological membranes and their potential to limit the entry of large sized, charged, hydrophilic molecules thus presenting a sizeable challenge for the cellular integration of negatively charged nucleic acids. Furthermore, the susceptibility of nucleic acids towards the degrading enzymes (nucleases) in the lysosomes present in cytoplasm is another matter of concern for their cellular and nuclear delivery. Hence, there is a pressing need for the identification and development of cationic delivery systems which encapsulate the cargo nucleic acids where the charge facilitates their cellular entry by evading the membrane barriers, and the encapsulation shields them from the enzymatic attack in cytoplasm. Cycloamylose bearing a closed loop conformation presents a robust candidature in this regard owing to its remarkable encapsulating tendency towards nucleic acids including siRNA, CpG DNA, and siRNA. The presence of numerous hydroxyl groups on the cycloamylose periphery provides sites for its chemical modification for the introduction of cationic groups, including spermine, (3-Chloro-2 hydroxypropyl) trimethylammonium chloride (Q188), and diethyl aminoethane (DEAE). The resulting cationic cycloamylose possesses a remarkable transfection efficiency and provides stability to cargo oligonucleotides against endonucleases, in addition to modulating the undesirable side effects such as unwanted immune stimulation. Cycloamylose is known to interact with the cell membranes where they release certain membrane components such as phospholipids and cholesterol thereby resulting in membrane destabilization and permeabilization. Furthermore, cycloamylose derivatives also serve as formulation excipients for improving the efficiency of other gene delivery systems. This review delves into the various vector and non-vector-based gene delivery systems, their advantages, and limitations, eventually leading to the identification of cycloamylose as an ideal candidate for nucleic acid delivery. The synthesis of cationic cycloamylose is briefly discussed in each section followed by its application for specific delivery/transfection of a particular nucleic acid.
Collapse
Affiliation(s)
- Parteek Prasher
- Department of Chemistry, University of Petroleum & Energy Studies, Energy Acres, Dehradun, 248007, India.
| | - Mousmee Sharma
- Department of Chemistry, Uttaranchal University, Dehradun, 248007, India
| | - Vipul Agarwal
- Cluster for Advanced Macromolecular Design (CAMD), School of Chemical Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; School of Medical and Life Sciences, Sunway University, 47500 Sunway City, Malaysia
| | - Gaurav Gupta
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak, 124001, India
| | - Kamal Dua
- Faculty of Health, Australian Research Center in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
7
|
Adachi T, Tahara Y, Yamamoto K, Yamamoto T, Kanamura N, Akiyoshi K, Mazda O. Cholesterol-Bearing Polysaccharide-Based Nanogels for Development of Novel Immunotherapy and Regenerative Medicine. Gels 2024; 10:206. [PMID: 38534624 DOI: 10.3390/gels10030206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/26/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024] Open
Abstract
Novel functional biomaterials are expected to bring about breakthroughs in developing immunotherapy and regenerative medicine through their application as drug delivery systems and scaffolds. Nanogels are defined as nanoparticles with a particle size of 100 nm or less and as having a gel structure. Nanogels have a three-dimensional network structure of cross-linked polymer chains, which have a high water content, a volume phase transition much faster than that of a macrogel, and a quick response to external stimuli. As it is possible to transmit substances according to the three-dimensional mesh size of the gel, a major feature is that relatively large substances, such as proteins and nucleic acids, can be taken into the gel. Furthermore, by organizing nanogels as a building block, they can be applied as a scaffold material for tissue regeneration. This review provides a brief overview of the current developments in nanogels in general, especially drug delivery, therapeutic applications, and tissue engineering. In particular, polysaccharide-based nanogels are interesting because they have excellent complexation properties and are highly biocompatible.
Collapse
Affiliation(s)
- Tetsuya Adachi
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Yoshiro Tahara
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara Miyakodani, Kyoto-fu, Kyotanabe-shi 610-0321, Japan
| | - Kenta Yamamoto
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| |
Collapse
|
8
|
Li J, Wu K, Zhang J, Gao H, Xu X. Progress in the treatment of drug-loaded nanomaterials in renal cell carcinoma. Biomed Pharmacother 2023; 167:115444. [PMID: 37716114 DOI: 10.1016/j.biopha.2023.115444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/18/2023] Open
Abstract
Renal cell carcinoma (RCC) is a common urinary tract tumor that arises from the highly heterogeneous epithelium of the renal tubules. The incidence of kidney cancer is second only to the incidence of bladder cancer, and has shown an upward trend over time. Although surgery is the preferred treatment for localized RCC, treatment decisions should be customized to individual patients considering their overall health status and the risk of developing or worsening chronic kidney disease postoperatively. Anticancer drugs are preferred to prevent perioperative and long-term postoperative complications; however, resistance to chemotherapy remains a considerable problem during the treatment process. To overcome this challenge, nanocarriers have emerged as a promising strategy for targeted drug delivery for cancer treatment. Nanocarriers can transport anticancer agents, achieving several-fold higher cytotoxic concentrations in tumors and minimizing toxicity to the remaining parts of the body. This article reviews the use of nanomaterials, such as liposomes, polymeric nanoparticles, nanocomposites, carbon nanomaterials, nanobubbles, nanomicelles, and mesoporous silica nanoparticles, for RCC treatment, and discusses their advantages and disadvantages.
Collapse
Affiliation(s)
- Jianyang Li
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Kunzhe Wu
- Department of Urology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jinmei Zhang
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Huan Gao
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaohua Xu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, China.
| |
Collapse
|
9
|
Afrin H, Geetha Bai R, Kumar R, Ahmad SS, Agarwal SK, Nurunnabi M. Oral delivery of RNAi for cancer therapy. Cancer Metastasis Rev 2023; 42:699-724. [PMID: 36971908 PMCID: PMC10040933 DOI: 10.1007/s10555-023-10099-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 03/14/2023] [Indexed: 03/29/2023]
Abstract
Cancer is a major health concern worldwide and is still in a continuous surge of seeking for effective treatments. Since the discovery of RNAi and their mechanism of action, it has shown promises in targeted therapy for various diseases including cancer. The ability of RNAi to selectively silence the carcinogenic gene makes them ideal as cancer therapeutics. Oral delivery is the ideal route of administration of drug administration because of its patients' compliance and convenience. However, orally administered RNAi, for instance, siRNA, must cross various extracellular and intracellular biological barriers before it reaches the site of action. It is very challenging and important to keep the siRNA stable until they reach to the targeted site. Harsh pH, thick mucus layer, and nuclease enzyme prevent siRNA to diffuse through the intestinal wall and thereby induce a therapeutic effect. After entering the cell, siRNA is subjected to lysosomal degradation. Over the years, various approaches have been taken into consideration to overcome these challenges for oral RNAi delivery. Therefore, understanding the challenges and recent development is crucial to offer a novel and advanced approach for oral RNAi delivery. Herein, we have summarized the delivery strategies for oral delivery RNAi and recent advancement towards the preclinical stages.
Collapse
Affiliation(s)
- Humayra Afrin
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
| | - Renu Geetha Bai
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Chair of Biosystems Engineering, Institute of Forestry and Engineering, Estonian University of Life Sciences, Kreutzwaldi 56/1, 51006, Tartu, Estonia
| | - Raj Kumar
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
| | - Sheikh Shafin Ahmad
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, TX, 79965, USA
| | - Sandeep K Agarwal
- Section of Immunology, Allergy and Rheumatology, Department of Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Md Nurunnabi
- Environmental Science & Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA.
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Texas at El Paso, 1101 N. Campbell St, El Paso, TX, 79902, USA.
- Aerospace Center (cSETR), University of Texas at El Paso, El Paso, TX, 79965, USA.
- Biomedical Engineering, College of Engineering, University of Texas at El Paso, El Paso, TX, 79965, USA.
| |
Collapse
|
10
|
Sankaranarayanan A, Ramprasad A, Shree Ganesh S, Ganesh H, Ramanathan B, Shanmugavadivu A, Selvamurugan N. Nanogels for bone tissue engineering - from synthesis to application. NANOSCALE 2023. [PMID: 37305943 DOI: 10.1039/d3nr01246h] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Nanogels are cross-linked hydrogel nanoparticles with a three-dimensional, tunable porous structure that merges the best features of hydrogels and nanoparticles, including the ability to retain their hydrated nature and to swell and shrink in response to environmental changes. Nanogels have attracted increasing attention for use in bone tissue engineering as scaffolds for growth factor transport and cell adhesion. Their three-dimensional structures allow the encapsulation of a wide range of hydrophobic and hydrophilic drugs, enhance their half-life, and impede their enzymatic breakdown in vivo. Nanogel-based scaffolds are a viable treatment modality for enhanced bone regeneration. They act as carriers for cells and active ingredients capable of controlled release, enhanced mechanical support, and osteogenesis for enhanced bone tissue regeneration. However, the development of such nanogel constructs might involve combinations of several biomaterials to fabricate active ingredients that can control release, enhance mechanical support, and facilitate osteogenesis for more effective bone tissue regeneration. Hence, this review aims to highlight the potential of nanogel-based scaffolds to address the needs of bone tissue engineering.
Collapse
Affiliation(s)
- Aravind Sankaranarayanan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - Anushikaa Ramprasad
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - S Shree Ganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - Harini Ganesh
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - Bharathi Ramanathan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - Abinaya Shanmugavadivu
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur-603203, Tamil Nadu, India.
| |
Collapse
|
11
|
Wu R, Wang K, Gai Y, Li M, Wang J, Wang C, Zhang Y, Xiao Z, Jiang D, Gao Z, Xia X. Nanomedicine for renal cell carcinoma: imaging, treatment and beyond. J Nanobiotechnology 2023; 21:3. [PMID: 36597108 PMCID: PMC9809106 DOI: 10.1186/s12951-022-01761-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 12/26/2022] [Indexed: 01/04/2023] Open
Abstract
The kidney is a vital organ responsible for maintaining homeostasis in the human body. However, renal cell carcinoma (RCC) is a common malignancy of the urinary system and represents a serious threat to human health. Although the overall survival of RCC has improved substantially with the development of cancer diagnosis and management, there are various reasons for treatment failure. Firstly, without any readily available biomarkers, timely diagnosis has been greatly hampered. Secondly, the imaging appearance also varies greatly, and its early detection often remains difficult. Thirdly, chemotherapy has been validated as unavailable for treating renal cancer in the clinic due to its intrinsic drug resistance. Concomitant with the progress of nanotechnological methods in pharmaceuticals, the management of kidney cancer has undergone a transformation in the recent decade. Nanotechnology has shown many advantages over widely used traditional methods, leading to broad biomedical applications ranging from drug delivery, prevention, diagnosis to treatment. This review focuses on nanotechnologies in RCC management and further discusses their biomedical translation with the aim of identifying the most promising nanomedicines for clinical needs. As our understanding of nanotechnologies continues to grow, more opportunities to improve the management of renal cancer are expected to emerge.
Collapse
Affiliation(s)
- Ruolin Wu
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Keshan Wang
- grid.33199.310000 0004 0368 7223Department of Urology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Yongkang Gai
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Mengting Li
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Jingjing Wang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Chenyang Wang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Yajing Zhang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Zhiwei Xiao
- grid.413247.70000 0004 1808 0969Department of Nuclear Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Dawei Jiang
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Zairong Gao
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| | - Xiaotian Xia
- grid.33199.310000 0004 0368 7223Department of Nuclear Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022 Hubei People’s Republic of China ,grid.412839.50000 0004 1771 3250Hubei Province Key Laboratory of Molecular Imaging, Wuhan, China ,grid.419897.a0000 0004 0369 313XKey Laboratory of Biological Targeted Therapy, The Ministry of Education, Wuhan, China
| |
Collapse
|
12
|
Ansari A, Sheriker A, Siddique MUM, Patil M, Goyal SN, Murtuja S, Ahsan MN, Hasnain MS, Nayak AK. Cancer targeting by tailor-made polysaccharides. TAILOR-MADE POLYSACCHARIDES IN DRUG DELIVERY 2023:347-376. [DOI: 10.1016/b978-0-12-821286-8.00011-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
13
|
Narayanan KB, Bhaskar R, Han SS. Recent Advances in the Biomedical Applications of Functionalized Nanogels. Pharmaceutics 2022; 14:2832. [PMID: 36559325 PMCID: PMC9782855 DOI: 10.3390/pharmaceutics14122832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Nanomaterials have been extensively used in several applications in the past few decades related to biomedicine and healthcare. Among them, nanogels (NGs) have emerged as an important nanoplatform with the properties of both hydrogels and nanoparticles for the controlled/sustained delivery of chemo drugs, nucleic acids, or other bioactive molecules for therapeutic or diagnostic purposes. In the recent past, significant research efforts have been invested in synthesizing NGs through various synthetic methodologies such as free radical polymerization, reversible addition-fragmentation chain-transfer method (RAFT) and atom transfer radical polymerization (ATRP), as well as emulsion techniques. With further polymeric functionalizations using activated esters, thiol-ene/yne processes, imines/oximes formation, cycloadditions, nucleophilic addition reactions of isocyanates, ring-opening, and multicomponent reactions were used to obtain functionalized NGs for targeted delivery of drug and other compounds. NGs are particularly intriguing for use in the areas of diagnosis, analytics, and biomedicine due to their nanodimensionality, material characteristics, physiological stability, tunable multi-functionality, and biocompatibility. Numerous NGs with a wide range of functionalities and various external/internal stimuli-responsive modalities have been possible with novel synthetic reliable methodologies. Such continuous development of innovative, intelligent materials with novel characteristics is crucial for nanomedicine for next-generation biomedical applications. This paper reviews the synthesis and various functionalization strategies of NGs with a focus on the recent advances in different biomedical applications of these surface modified/functionalized single-/dual-/multi-responsive NGs, with various active targeting moieties, in the fields of cancer theranostics, immunotherapy, antimicrobial/antiviral, antigen presentation for the vaccine, sensing, wound healing, thrombolysis, tissue engineering, and regenerative medicine.
Collapse
Affiliation(s)
- Kannan Badri Narayanan
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
- Research Institute of Cell Culture, Yeungnam University, 280 Daehak-Ro, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
14
|
Suksiri P, Sansanaphongpricha K, Muangsin N, Krusong K. Development of positively-charged cycloamylose, CAQ as efficient nanodelivery system for siRNA. Biochem Eng J 2022. [DOI: 10.1016/j.bej.2022.108767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
15
|
Gene Regulations upon Hydrogel-Mediated Drug Delivery Systems in Skin Cancers-An Overview. Gels 2022; 8:gels8090560. [PMID: 36135270 PMCID: PMC9498739 DOI: 10.3390/gels8090560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/24/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
The incidence of skin cancer has increased dramatically in recent years, particularly in Caucasian populations. Specifically, the metastatic melanoma is one of the most aggressive cancers and is responsible for more than 80% of skin cancer deaths around the globe. Though there are many treatment techniques, and drugs have been used to cure this belligerent skin cancer, the side effects and reduced bioavailability of drug in the targeted area makes it difficult to eradicate. In addition, cellular metabolic pathways are controlled by the skin cancer driver genes, and mutations in these genes promote tumor progression. Consequently, the MAPK (RAS-RAF-MEK-ERK pathway), WNT and PI3K signaling pathways are found to be important molecular regulators in melanoma development. Even though hydrogels have turned out to be a promising drug delivery system in skin cancer treatment, the regulations at the molecular level have not been reported. Thus, we aimed to decipher the molecular pathways of hydrogel drug delivery systems for skin cancer in this review. Special attention has been paid to the hydrogel systems that deliver drugs to regulate MAPK, PI3K-AKT-mTOR, JAK-STAT and cGAS-STING pathways. These signaling pathways can be molecular drivers of skin cancers and possible potential targets for the further research on treatment of skin cancers.
Collapse
|
16
|
Liu Y, Geng Y, Yue B, Lo PC, Huang J, Jin H. Injectable Hydrogel as a Unique Platform for Antitumor Therapy Targeting Immunosuppressive Tumor Microenvironment. Front Immunol 2022; 12:832942. [PMID: 35111169 PMCID: PMC8801935 DOI: 10.3389/fimmu.2021.832942] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 12/27/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy can boost the immune response of patients to eliminate tumor cells and suppress tumor metastasis and recurrence. However, immunotherapy resistance and the occurrence of severe immune-related adverse effects are clinical challenges that remain to be addressed. The tumor microenvironment plays a crucial role in the therapeutic efficacy of cancer immunotherapy. Injectable hydrogels have emerged as powerful drug delivery platforms offering good biocompatibility and biodegradability, minimal invasion, convenient synthesis, versatility, high drug-loading capacity, controlled drug release, and low toxicity. In this review, we summarize the application of injectable hydrogels as a unique platform for targeting the immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Yushuai Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuanyuan Geng
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Beilei Yue
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Pui-Chi Lo
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, SAR China
| | - Jing Huang
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Honglin Jin
- College of Biomedicine and Health and College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
17
|
Krusong K, Ismail A, Wangpaiboon K, Pongsawasdi P. Production of Large-Ring Cyclodextrins by Amylomaltases. Molecules 2022; 27:molecules27041446. [PMID: 35209232 PMCID: PMC8875642 DOI: 10.3390/molecules27041446] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/10/2022] [Accepted: 02/18/2022] [Indexed: 02/01/2023] Open
Abstract
Amylomaltase is a well-known glucan transferase that can produce large ring cyclodextrins (LR-CDs) or so-called cycloamyloses via cyclization reaction. Amylomaltases have been found in several microorganisms and their optimum temperatures are generally around 60–70 °C for thermostable amylomaltases and 30–45 °C for the enzymes from mesophilic bacteria and plants. The optimum pHs for mesophilic amylomaltases are around pH 6.0–7.0, while the thermostable amylomaltases are generally active at more acidic conditions. Size of LR-CDs depends on the source of amylomaltases and the reaction conditions including pH, temperature, incubation time, and substrate. For example, in the case of amylomaltase from Corynebacterium glutamicum, LR-CD productions at alkaline pH or at a long incubation time favored products with a low degree of polymerization. In this review, we explore the synthesis of LR-CDs by amylomaltases, structural information of amylomaltases, as well as current applications of LR-CDs and amylomaltases.
Collapse
Affiliation(s)
- Kuakarun Krusong
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phyathai Rd., Patumwan, Bangkok 10330, Thailand; (A.I.); (K.W.)
- Correspondence: ; Tel.: + 66-(0)2-218-5413
| | - Abbas Ismail
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phyathai Rd., Patumwan, Bangkok 10330, Thailand; (A.I.); (K.W.)
| | - Karan Wangpaiboon
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phyathai Rd., Patumwan, Bangkok 10330, Thailand; (A.I.); (K.W.)
| | - Piamsook Pongsawasdi
- Starch and Cyclodextrin Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Phyathai Rd., Patumwan, Bangkok 10330, Thailand;
| |
Collapse
|
18
|
Gupta N, Malviya R. Role of Polysaccharides Mimetic Components in Targeted Cancer Treatment. Curr Drug Targets 2022; 23:856-868. [PMID: 35156570 DOI: 10.2174/1389450123666220214121505] [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: 09/14/2021] [Revised: 11/09/2021] [Accepted: 12/02/2021] [Indexed: 11/22/2022]
Abstract
Organic or inorganic compounds are synthesized or formulated in a manner that they completely show their therapeutic actions like as a natural polysaccharide in the body. Polysaccharides, the major type of natural polymers, are efficiently biologically active, non-toxic, hydrophilic, and biodegradable and show various properties. In this manuscript, the main focus is on delivering anticancer drugs with the help of mimetic components of polysaccharides. All data collected for this manuscript was from PubMed, Elsevier, Taylor, and Francis Bentham science journals. Most chemotherapeutics are therapeutically toxin to the human body, have a narrow therapeutic index, sluggish pharmaceutical delivery mechanisms, and are poorly soluble in water. The use of mimetic components of polysaccharides leads to the enhancement of the solubility of drugs in the biological environment. The manuscript summarizes the use of mimetic components of polysaccharides along with anticancer agents which are capable to inhibit the growth of cancerous cells in the body which shows lesser adverse effects in the biological system compared to other therapies.
Collapse
Affiliation(s)
- Nandan Gupta
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University Greater Noida, Uttar Pradesh, India
| |
Collapse
|
19
|
|
20
|
Suksiri P, Ismail A, Sirirattanachatchawan C, Wangpaiboon K, Muangsin N, Tananuwong K, Krusong K. Enhancement of large ring cyclodextrin production using pretreated starch by glycogen debranching enzyme from Corynebacterium glutamicum. Int J Biol Macromol 2021; 193:81-87. [PMID: 34678383 DOI: 10.1016/j.ijbiomac.2021.10.082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 11/19/2022]
Abstract
Synthesis of large-ring cyclodextrins (LR-CDs) in any significant amount has been challenging. This study enhanced the LR-CDs production by Thermus filiformis amylomaltase (TfAM) enzyme by starch pretreatment using glycogen debranching enzyme from Corynebacterium glutamicum (CgGDE). CgGDE pretreated tapioca starch gave LR-CD conversion of 31.2 ± 2.2%, compared with LR-CDs produced from non-treated tapioca starch (16.0 ± 2.4%). CgGDE pretreatment enhanced amylose content by approximately 30%. Notably, a shorter incubation time of 1 h is sufficient for CgGDE starch pretreatment to produce high LR-CD yield, compared with 6 h required for the commercial isoamylase. High-Performance Anion Exchange Chromatography coupled with Pulsed Amperometric Detection (HPAEC-PAD) and Gel Permeable Chromatography (GPC) revealed that CgGDE is more efficient than the commercial isoamylase in debranching tapioca starch and gave lower molecular weight products. In addition, lower amount of by-products (linear oligosaccharides) were detected in cyclization reaction when using CgGDE-pretreated starch. In conclusion, CgGDE is a highly effective enzyme to promote LR-CD synthesis from starch with a shorter incubation time than the commercial isoamylase.
Collapse
Affiliation(s)
- Pornchanok Suksiri
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; Program of Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Abbas Ismail
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Chay Sirirattanachatchawan
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand; Program of Biotechnology, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Karan Wangpaiboon
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Nongnuj Muangsin
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kanitha Tananuwong
- Department of Food Technology, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kuakarun Krusong
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
21
|
Leoni C, Gattulli BAR, Pesole G, Ceci LR, Volpicella M. Amylomaltases in Extremophilic Microorganisms. Biomolecules 2021; 11:biom11091335. [PMID: 34572549 PMCID: PMC8465469 DOI: 10.3390/biom11091335] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
Amylomaltases (4-α-glucanotransferases, E.C. 2.4.1.25) are enzymes which can perform a double-step catalytic process, resulting in a transglycosylation reaction. They hydrolyse glucosidic bonds of α-1,4'-d-glucans and transfer the glucan portion with the newly available anomeric carbon to the 4'-position of an α-1,4'-d-glucan acceptor. The intramolecular reaction produces a cyclic α-1,4'-glucan. Amylomaltases can be found only in prokaryotes, where they are involved in glycogen degradation and maltose metabolism. These enzymes are being studied for possible biotechnological applications, such as the production of (i) sugar substitutes; (ii) cycloamyloses (molecules larger than cyclodextrins), which could potentially be useful as carriers and encapsulating agents for hydrophobic molecules and also as effective protein chaperons; and (iii) thermoreversible starch gels, which could be used as non-animal gelatin substitutes. Extremophilic prokaryotes have been investigated for the identification of amylomaltases to be used in the starch modifying processes, which require high temperatures or extreme conditions. The aim of this article is to present an updated overview of studies on amylomaltases from extremophilic Bacteria and Archaea, including data about their distribution, activity, potential industrial application and structure.
Collapse
Affiliation(s)
- Claudia Leoni
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Via Amendola, 70126 Bari, Italy; (C.L.); (B.A.R.G.); (G.P.)
| | - Bruno A. R. Gattulli
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Via Amendola, 70126 Bari, Italy; (C.L.); (B.A.R.G.); (G.P.)
| | - Graziano Pesole
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Via Amendola, 70126 Bari, Italy; (C.L.); (B.A.R.G.); (G.P.)
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Amendola 165/A, 70126 Bari, Italy
| | - Luigi R. Ceci
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Via Amendola, 70126 Bari, Italy; (C.L.); (B.A.R.G.); (G.P.)
- Correspondence: (L.R.C.); (M.V.); Tel.: +39-080-544-3311 (L.R.C. & M.V.)
| | - Mariateresa Volpicella
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, Consiglio Nazionale delle Ricerche, Via Amendola, 70126 Bari, Italy; (C.L.); (B.A.R.G.); (G.P.)
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Via Amendola 165/A, 70126 Bari, Italy
- Correspondence: (L.R.C.); (M.V.); Tel.: +39-080-544-3311 (L.R.C. & M.V.)
| |
Collapse
|
22
|
Jeong HM, Lee Y, Shin YJ, Woo SH, Kim JS, Jeong DW, Shin S, Jeon SH, Shim JH. Development of an enzymatic encapsulation process for a cycloamylose inclusion complex with resveratrol. Food Chem 2020; 345:128777. [PMID: 33321347 DOI: 10.1016/j.foodchem.2020.128777] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/29/2022]
Abstract
Cyclodextrin glucanotransferase (CGTase; EC 2.4.1.19) produces cycloamyloses (CAs), which are large cyclic glucans, and subsequently transforms them to α-, β-, and γ-cyclodextrins. We developed a novel encapsulation process based on the cyclization activity of CGTase and applied it to the formation of CA inclusion complexes with resveratrol (RVT), which has limited bioavailability due to its low water solubility. The encapsulated RVT (CA-RVT) was purified using preparative high-performance liquid chromatography. The water solubility of CA-RVT was 6,000-fold higher than that of RVT. CA-RVT in water demonstrated 98% stability for 1 week at 4 °C. According to radical scavenging activity and anti-inflammatory assays, CA-RVT in aqueous solution exhibited similar activities as an equal amount of RVT in dimethyl sulfoxide, suggesting the limited solubility of RVT can be overcome through CA encapsulation by CGTase, thus enhancing its nutraceutical value as a functional ingredient in the food industry.
Collapse
Affiliation(s)
- Hyun-Mo Jeong
- Department of Food Science and Nutrition, and the Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, South Korea.
| | - Yeontaek Lee
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gangwon-do 24252, South Korea
| | - Yu-Jeong Shin
- Department of Food Science and Nutrition, and the Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, South Korea.
| | - Seung-Hye Woo
- Department of Food Science and Nutrition, and the Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, South Korea.
| | - Ji-Soo Kim
- Department of Food Science and Nutrition, and the Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, South Korea.
| | - Da-Woon Jeong
- Department of Food Science and Nutrition, and the Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, South Korea.
| | - Sooyong Shin
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gangwon-do 24252, South Korea.
| | - Sung Ho Jeon
- Department of Life Science and Multidisciplinary Genome Institute, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gangwon-do 24252, South Korea.
| | - Jae-Hoon Shim
- Department of Food Science and Nutrition, and the Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, South Korea.
| |
Collapse
|
23
|
Misiak P, Markiewicz KH, Szymczuk D, Wilczewska AZ. Polymeric Drug Delivery Systems Bearing Cholesterol Moieties: A Review. Polymers (Basel) 2020; 12:E2620. [PMID: 33172152 PMCID: PMC7694753 DOI: 10.3390/polym12112620] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 12/21/2022] Open
Abstract
This review aims to provide an overview of polymers comprising cholesterol moiety/ies designed to be used in drug delivery. Over the last two decades, there have been many papers published in this field, which are summarized in this review. The primary focus of this article is on the methods of synthesis of polymers bearing cholesterol in the main chain or as side chains. The data related to the composition, molecular weight, and molecular weight distribution of polymers are presented. Moreover, other aspects, such as forms of carriers, types of encapsulated drugs, encapsulation efficiency and capacity, are also included.
Collapse
Affiliation(s)
- Paweł Misiak
- Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1k, 15-245 Bialystok, Poland; (K.H.M.); (D.S.)
| | | | | | - Agnieszka Z. Wilczewska
- Faculty of Chemistry, University of Bialystok, Ciolkowskiego 1k, 15-245 Bialystok, Poland; (K.H.M.); (D.S.)
| |
Collapse
|
24
|
Pinelli F, Ortolà ÓF, Makvandi P, Perale G, Rossi F. In vivo drug delivery applications of nanogels: a review. Nanomedicine (Lond) 2020; 15:2707-2727. [PMID: 33103960 DOI: 10.2217/nnm-2020-0274] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
In recent years, nanogels have emerged as promising drug delivery vehicles; their ability in holding active molecules, macromolecules and drugs, together with the capability to respond to external stimuli, makes them a suitable tool for a wide range of applications. These features allow nanogels to be exploited against many challenges of nanomedicine associated with different kinds of pathologies which require the use of specific drug delivery systems. In this review our aim is to give the reader an overview of the diseases that can be treated with nanogels as drug delivery systems, such as cancer, CNS disorders, cardiovascular diseases, wound healing and other diseases of human body. For all of these pathologies, biological in vivo assays can be found in the literature and in this work. We focus on the peculiarities of these nanogels, highlighting their features and their advantages in respect to conventional treatments.
Collapse
Affiliation(s)
- Filippo Pinelli
- Department of Chemistry, Materials & Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, 20131, Milan, Italy
| | - Óscar Fullana Ortolà
- Department of Chemistry, Materials & Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, 20131, Milan, Italy
| | - Pooyan Makvandi
- Institute for Polymers, Composites & Biomaterials, National Research Council, Via Campi Flegrei, 34 - 80078 Pozzuoli (NA), Italy.,Istituto Italiano di Tecnologia, Centre for Micro-BioRobotics, Viale Rinaldo Piaggio 34, 56025 Pontedera, Pisa, Italy.,Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Giuseppe Perale
- Faculty of Biomedical Sciences, University of Southern Switzerland (USI), Via Buffi 13, 6900 Lugano, Switzerland
| | - Filippo Rossi
- Department of Chemistry, Materials & Chemical Engineering "Giulio Natta", Politecnico di Milano, via Mancinelli 7, 20131, Milan, Italy
| |
Collapse
|
25
|
Imamura K, Matsuura T, Nakagawa A, Kitamura S, Kusunoki M, Takaha T, Unno H. Structural analysis and reaction mechanism of the disproportionating enzyme (D-enzyme) from potato. Protein Sci 2020; 29:2085-2100. [PMID: 32808707 PMCID: PMC7513719 DOI: 10.1002/pro.3932] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/14/2020] [Accepted: 08/14/2020] [Indexed: 11/06/2022]
Abstract
Starch produced by plants is a stored form of energy and is an important dietary source of calories for humans and domestic animals. Disproportionating enzyme (D-enzyme) catalyzes intramolecular and intermolecular transglycosylation reactions of α-1, 4-glucan. D-enzyme is essential in starch metabolism in the potato. We present the crystal structures of potato D-enzyme, including two different types of complex structures: a primary Michaelis complex (substrate binding mode) for 26-meric cycloamylose (CA26) and a covalent intermediate for acarbose. Our study revealed that the acarbose and CA26 reactions catalyzed by potato D-enzyme involve the formation of a covalent intermediate with the donor substrate. HPAEC of reaction substrates and products revealed the activity of the potato D-enzyme on acarbose and CA26 as donor substrates. The structural and chromatography analyses provide insight into the mechanism of the coupling reaction of CA and glucose catalyzed by the potato D-enzyme. The enzymatic reaction mechanism does not involve residual hydrolysis. This could be particularly useful in preventing unnecessary starch degradation leading to reduced crop productivity. Optimization of this mechanism would be important for improvements of starch storage and productivity in crops.
Collapse
Affiliation(s)
- Kayo Imamura
- Laboratory of Enzyme Chemistry, Graduate School of Agriculture and Biological ScienceOsaka Prefecture UniversityOsakaJapan
| | | | | | - Shinichi Kitamura
- Laboratory of Biophysical Chemistry, Graduate School of Agriculture and Biological ScienceOsaka Prefecture UniversityOsakaJapan
- Present address:
Laboratory of Advanced Food Process EngineeringOsaka Prefecture University, 1‐2, Gakuen‐cho, Nakaku, Osaka, Sakai 599‐8570Japan
| | | | - Takeshi Takaha
- Biochemical Research LaboratoriesEzaki Glico Co., LtdOsakaJapan
- Present address:
Sanawa Starch Co., Ltd. 594 Unate, Kashihara, Nara 634‐8585Japan
| | - Hideaki Unno
- Graduate School of EngineeringNagasaki UniversityNagasakiJapan
- Organization for Marine Science and TechnologyNagasaki UniversityNagasakiJapan
| |
Collapse
|
26
|
Adachi T, Boschetto F, Miyamoto N, Yamamoto T, Marin E, Zhu W, Kanamura N, Tahara Y, Akiyoshi K, Mazda O, Nishimura I, Pezzotti G. In Vivo Regeneration of Large Bone Defects by Cross-Linked Porous Hydrogel: A Pilot Study in Mice Combining Micro Tomography, Histological Analyses, Raman Spectroscopy and Synchrotron Infrared Imaging. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E4275. [PMID: 32992758 PMCID: PMC7579234 DOI: 10.3390/ma13194275] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/23/2020] [Accepted: 09/23/2020] [Indexed: 01/25/2023]
Abstract
The transplantation of engineered three-dimensional (3D) bone graft substitutes is a viable approach to the regeneration of severe bone defects. For large bone defects, an appropriate 3D scaffold may be necessary to support and stimulate bone regeneration, even when a sufficient number of cells and cell cytokines are available. In this study, we evaluated the in vivo performance of a nanogel tectonic 3D scaffold specifically developed for bone tissue engineering, referred to as nanogel cross-linked porous-freeze-dry (NanoCliP-FD) gel. Samples were characterized by a combination of micro-computed tomography scanning, Raman spectroscopy, histological analyses, and synchrotron radiation-based Fourier transform infrared spectroscopy. NanoCliP-FD gel is a modified version of a previously developed nanogel cross-linked porous (NanoCliP) gel and was designed to achieve highly improved functionality in bone mineralization. Spectroscopic imaging of the bone tissue grown in vivo upon application of NanoCliP-FD gel enables an evaluation of bone quality and can be employed to judge the feasibility of NanoCliP-FD gel scaffolding as a therapeutic modality for bone diseases associated with large bone defects.
Collapse
Affiliation(s)
- Tetsuya Adachi
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (F.B.); (N.M.); (T.Y.); (E.M.); (N.K.)
| | - Francesco Boschetto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (F.B.); (N.M.); (T.Y.); (E.M.); (N.K.)
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan; (W.Z.); (G.P.)
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Nao Miyamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (F.B.); (N.M.); (T.Y.); (E.M.); (N.K.)
- Department of Infectious Diseases, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Toshiro Yamamoto
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (F.B.); (N.M.); (T.Y.); (E.M.); (N.K.)
| | - Elia Marin
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (F.B.); (N.M.); (T.Y.); (E.M.); (N.K.)
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan; (W.Z.); (G.P.)
| | - Wenliang Zhu
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan; (W.Z.); (G.P.)
| | - Narisato Kanamura
- Department of Dental Medicine, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan; (F.B.); (N.M.); (T.Y.); (E.M.); (N.K.)
| | - Yoshiro Tahara
- Department of Chemical Engineering and Materials Science, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe-shi, Kyoto-fu 610-0394, Japan;
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo-ku, Kyoto 615-8510, Japan;
| | - Osam Mazda
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| | - Ichiro Nishimura
- Division of Oral Biology and Medicine, The Jane and Jerry Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095, USA;
- Division of Advanced Prosthodontics, The Jane and Jerry Weintraub Center for Re-constructive Biotechnology, UCLA School of Dentistry, Los Angeles, CA 90095, USA
| | - Giuseppe Pezzotti
- Ceramic Physics Laboratory, Kyoto Institute of Technology, Sakyo-ku, Matsugasaki, Kyoto 606-8585, Japan; (W.Z.); (G.P.)
- Department of Immunology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kamigyo-ku, 465 Kajii-cho, Kyoto 602-8566, Japan
| |
Collapse
|
27
|
Hager S, Fittler FJ, Wagner E, Bros M. Nucleic Acid-Based Approaches for Tumor Therapy. Cells 2020; 9:E2061. [PMID: 32917034 PMCID: PMC7564019 DOI: 10.3390/cells9092061] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/06/2020] [Accepted: 09/07/2020] [Indexed: 12/24/2022] Open
Abstract
Within the last decade, the introduction of checkpoint inhibitors proposed to boost the patients' anti-tumor immune response has proven the efficacy of immunotherapeutic approaches for tumor therapy. Furthermore, especially in the context of the development of biocompatible, cell type targeting nano-carriers, nucleic acid-based drugs aimed to initiate and to enhance anti-tumor responses have come of age. This review intends to provide a comprehensive overview of the current state of the therapeutic use of nucleic acids for cancer treatment on various levels, comprising (i) mRNA and DNA-based vaccines to be expressed by antigen presenting cells evoking sustained anti-tumor T cell responses, (ii) molecular adjuvants, (iii) strategies to inhibit/reprogram tumor-induced regulatory immune cells e.g., by RNA interference (RNAi), (iv) genetically tailored T cells and natural killer cells to directly recognize tumor antigens, and (v) killing of tumor cells, and reprograming of constituents of the tumor microenvironment by gene transfer and RNAi. Aside from further improvements of individual nucleic acid-based drugs, the major perspective for successful cancer therapy will be combination treatments employing conventional regimens as well as immunotherapeutics like checkpoint inhibitors and nucleic acid-based drugs, each acting on several levels to adequately counter-act tumor immune evasion.
Collapse
Affiliation(s)
- Simone Hager
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University (LMU), 81377 Munich, Germany;
| | | | - Ernst Wagner
- Department of Chemistry and Pharmacy, Ludwig-Maximilians-University (LMU), 81377 Munich, Germany;
| | - Matthias Bros
- Department of Dermatology, University Medical Center, 55131 Mainz, Germany;
| |
Collapse
|
28
|
Barclay TG, Day CM, Petrovsky N, Garg S. Review of polysaccharide particle-based functional drug delivery. Carbohydr Polym 2019; 221:94-112. [PMID: 31227171 PMCID: PMC6626612 DOI: 10.1016/j.carbpol.2019.05.067] [Citation(s) in RCA: 211] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 04/26/2019] [Accepted: 05/22/2019] [Indexed: 01/06/2023]
Abstract
This review investigates the significant role polysaccharide particles play in functional drug delivery. The importance of these systems is due to the wide variety of polysaccharides and their natural source meaning that they can provide biocompatible and biodegradable systems with a range of both biological and chemical functionality valuable for drug delivery. This functionality includes protection and presentation of working therapeutics through avoidance of the reticuloendothelial system, stabilization of biomacromolecules and increasing the bioavailability of incorporated small molecule drugs. Transport of the therapeutic is also key to the utility of polysaccharide particles, moving drugs from the site of administration through mucosal binding and transport and using chemistry, size and receptor mediated drug targeting to specific tissues. This review also scrutinizes the methods of synthesizing and constructing functional polysaccharide particle drug delivery systems that maintain and extend the functionality of the natural polysaccharides.
Collapse
Affiliation(s)
- Thomas G Barclay
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia.
| | - Candace Minhthu Day
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia.
| | - Nikolai Petrovsky
- Vaxine Pty Ltd, 1 Flinders Drive, Bedford Park, SA 5042, Australia; Department of Endocrinology, Flinders Medical Centre/Flinders University, Bedford Park, SA 5042, Australia.
| | - Sanjay Garg
- School of Pharmacy and Medical Science, University of South Australia, Adelaide, SA 5000, Australia.
| |
Collapse
|
29
|
Abstract
With its nearly unrestricted possibilities, gene therapy attracts more and more significance in modern-day research. The only issue still seeming to hold back its clinical success is the actual effective delivery of genetic material. Nucleic acids are in general challenging to administer to their intracellular targets due to their unfavorable pharmaceutical characteristics. Polymeric nanogels present a promising delivery platform for oligonucleotide-based therapies, as the growing number of reports deliberated in this review represents. Within the scope of this article, recent progress in the employment of nanogels as gene delivery vectors is summarized and different examples of modified, stimuli-responsive, targeted and co-delivering nanogels are discussed in detail. Furthermore, major aspects of successful gene delivery are addressed and critically debated in regards to nanogels, giving insights into what progress has been made and which key issues still need to be further approached.
Collapse
Affiliation(s)
- Rima Kandil
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, Butenandtstraße 5-13, 81337, Munich, Germany
| | - Olivia M. Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmacy, Ludwig-Maximilians-University, Butenandtstraße 5-13, 81337, Munich, Germany
| |
Collapse
|
30
|
Nanogel tectonic porous 3D scaffold for direct reprogramming fibroblasts into osteoblasts and bone regeneration. Sci Rep 2018; 8:15824. [PMID: 30361649 PMCID: PMC6202359 DOI: 10.1038/s41598-018-33892-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 10/08/2018] [Indexed: 11/20/2022] Open
Abstract
Transplantation of engineered three-dimensional (3D) bone tissue may provide therapeutic benefits to patients with various bone diseases. To achieve this goal, appropriate 3D scaffolds and cells are required. In the present study, we devised a novel nanogel tectonic material for artificial 3D scaffold, namely the nanogel-cross-linked porous (NanoCliP)-freeze-dried (FD) gel, and estimated its potential as a 3D scaffold for bone tissue engineering. As the osteoblasts, directly converted osteoblasts (dOBs) were used, because a large number of highly functional osteoblasts could be induced from fibroblasts that can be collected from patients with a minimally invasive procedure. The NanoCliP-FD gel was highly porous, and fibronectin coating of the gel allowed efficient adhesion of the dOBs, so that the cells occupied the almost entire surface of the walls of the pores after culturing for 7 days. The dOBs massively produced calcified bone matrix, and the culture could be continued for at least 28 days. The NanoCliP-FD gel with dOBs remarkably promoted bone regeneration in vivo after having been grafted to bone defect lesions that were artificially created in mice. The present findings suggest that the combination of the NanoCliP-FD gel and dOBs may provide a feasible therapeutic modality for bone diseases.
Collapse
|
31
|
Tada H, Kishida T, Fujiwara H, Kosuga T, Konishi H, Komatsu S, Shiozaki A, Ichikawa D, Okamoto K, Otsuji E, Mazda O. Reprogrammed chondrocytes engineered to produce IL-12 provide novel ex vivo immune-gene therapy for cancer. Immunotherapy 2017; 9:239-248. [PMID: 28231722 DOI: 10.2217/imt-2016-0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM The somatic cell reprogramming technology was applied to a novel and promising ex vivo immune-gene therapy strategy for cancer. To establish a novel ex vivo cytokine gene therapy of cancer using the somatic cell reprogramming procedures. METHODS Mouse fibroblasts were converted into chondrocytes and subsequently transduced with IL-12 gene. The resultant IL-12 induced chondrogenic cells were irradiated with x-ray and inoculated into mice bearing CT26 colon cancer. RESULTS The irradiation at 20 Gy or higher totally eliminated the proliferative potential of the cells, while less significantly influencing the IL-12 production from the cells. An inoculation of the irradiated IL-12 induced chondrogenic cells significantly suppressed tumor by inducing tumor-specific cytotoxic T lymphocytes, enhancing natural killer tumoricidal activity and inhibiting tumor neoangiogenesis in the mice. CONCLUSION The somatic cell reprogramming procedures may provide a novel and effective means to treat malignancies.
Collapse
Affiliation(s)
- Hiroyuki Tada
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan.,Division of Digestive Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Tsunao Kishida
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Hitoshi Fujiwara
- Division of Digestive Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Toshiyuki Kosuga
- Division of Digestive Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Hirotaka Konishi
- Division of Digestive Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Shuhei Komatsu
- Division of Digestive Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Atsushi Shiozaki
- Division of Digestive Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Daisuke Ichikawa
- Division of Digestive Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Kazuma Okamoto
- Division of Digestive Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Eigo Otsuji
- Division of Digestive Surgery, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| | - Osam Mazda
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto 602-8566, Japan
| |
Collapse
|
32
|
Nishimura T, Akiyoshi K. Amylose engineering: phosphorylase-catalyzed polymerization of functional saccharide primers for glycobiomaterials. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2017; 9:e1423. [PMID: 27506150 PMCID: PMC5333464 DOI: 10.1002/wnan.1423] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 06/26/2016] [Accepted: 07/05/2016] [Indexed: 12/31/2022]
Abstract
Interest in amylose and its hybrids has grown over many decades, and a great deal of work has been devoted to developing methods for designing functional amylose hybrids. In this context, phosphorylase-catalyzed polymerization shows considerable promise as a tool for preparing diverse amylose hybrids. Recently, advances have been made in the chemoenzymatic synthesis and characterization of amylose-block-polymers, amylose-graft-polymers, amylose-modified surfaces, hetero-oligosaccharides, and cellodextrin hybrids. Many of these saccharides provide clear opportunities for advances in biomaterials because of their biocompatibility and biodegradability. Important developments in bioapplications of amylose hybrids have also been made, and such newly developed amylose hybrids will help promote the development of new generations of glyco materials. WIREs Nanomed Nanobiotechnol 2017, 9:e1423. doi: 10.1002/wnan.1423 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Tomoki Nishimura
- Department of Polymer Chemistry, Graduate School of EngineeringKyoto UniversityKyotoJapan
- JST-ERATO Akiyoshi Bionanotransporter ProjectKyotoJapan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of EngineeringKyoto UniversityKyotoJapan
- JST-ERATO Akiyoshi Bionanotransporter ProjectKyotoJapan
| |
Collapse
|
33
|
Liu Y, Lin L, Song J, Zhao Y, Chao Z, Li H. Preparation, characterization and anticancer activities of resveratrol loaded redox-sensitive F127-SS-TOC micelles. RSC Adv 2017. [DOI: 10.1039/c7ra08593a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Resveratrol (RES) loaded redox-sensitive F127-SS-TOC micelles (F127-SS-TOC/RES) hold the potential in improving the treatment of breast cancer, which compared to free RES or non-redox-sensitive micelles (F127-TOC/RES).
Collapse
Affiliation(s)
- Yuling Liu
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Sciences
- Beijing
- China
| | - Longfei Lin
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Sciences
- Beijing
- China
| | - Jizheng Song
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Sciences
- Beijing
- China
| | - Ye Zhao
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Sciences
- Beijing
- China
| | - Zhimao Chao
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Sciences
- Beijing
- China
| | - Hui Li
- Institute of Chinese Materia Medica
- China Academy of Chinese Medical Sciences
- Beijing
- China
| |
Collapse
|
34
|
Debele TA, Mekuria SL, Tsai HC. Polysaccharide based nanogels in the drug delivery system: Application as the carrier of pharmaceutical agents. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 68:964-981. [PMID: 27524098 DOI: 10.1016/j.msec.2016.05.121] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 05/23/2016] [Accepted: 05/27/2016] [Indexed: 11/08/2022]
|
35
|
Soni KS, Desale SS, Bronich TK. Nanogels: An overview of properties, biomedical applications and obstacles to clinical translation. J Control Release 2016; 240:109-126. [PMID: 26571000 PMCID: PMC4862943 DOI: 10.1016/j.jconrel.2015.11.009] [Citation(s) in RCA: 331] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Revised: 11/01/2015] [Accepted: 11/09/2015] [Indexed: 01/09/2023]
Abstract
Nanogels have emerged as a versatile hydrophilic platform for encapsulation of guest molecules with a capability to respond to external stimuli that can be used for a multitude of applications. These are soft materials capable of holding small molecular therapeutics, biomacromolecules, and inorganic nanoparticles within their crosslinked networks, which allows them to find applications for therapy as well as imaging of a variety of disease conditions. Their stimuli-responsive behavior can be easily controlled by selection of constituent polymer and crosslinker components to achieve a desired response at the site of action, which imparts nanogels the ability to participate actively in the intended function of the carrier system rather than being passive carriers of their cargo. These properties not only enhance the functionality of the carrier system but also help in overcoming many of the challenges associated with the delivery of cargo molecules, and this review aims to highlight the distinct and unique capabilities of nanogels as carrier systems for the delivery of an array of cargo molecules over other nanomaterials. Despite their obvious usefulness, nanogels are still not a commonplace occurrence in clinical practice. We have also made an attempt to highlight some of the major challenges that need to be overcome to advance nanogels further in the field of biomedical applications.
Collapse
Affiliation(s)
- Kruti S Soni
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Swapnil S Desale
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA
| | - Tatiana K Bronich
- Department of Pharmaceutical Sciences and Center for Drug Delivery and Nanomedicine, College of Pharmacy, University of Nebraska Medical Center, 985830 Nebraska Medical Center, Omaha, NE 68198-5830, USA.
| |
Collapse
|
36
|
Dheer D, Arora D, Jaglan S, Rawal RK, Shankar R. Polysaccharides based nanomaterials for targeted anti-cancer drug delivery. J Drug Target 2016; 25:1-16. [PMID: 27030377 DOI: 10.3109/1061186x.2016.1172589] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
37
|
Vongpichayapaiboon T, Pongsawasdi P, Krusong K. Optimization of large-ring cyclodextrin production from starch by amylomaltase from Corynebacterium glutamicum
and effect of organic solvent on product size. J Appl Microbiol 2016; 120:912-20. [DOI: 10.1111/jam.13087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2015] [Revised: 01/27/2016] [Accepted: 01/30/2016] [Indexed: 01/05/2023]
Affiliation(s)
- T. Vongpichayapaiboon
- Starch and Cyclodextrin Research Unit; Department of Biochemistry; Faculty of Science; Chulalongkorn University; Bangkok Thailand
| | - P. Pongsawasdi
- Starch and Cyclodextrin Research Unit; Department of Biochemistry; Faculty of Science; Chulalongkorn University; Bangkok Thailand
| | - K. Krusong
- Starch and Cyclodextrin Research Unit; Department of Biochemistry; Faculty of Science; Chulalongkorn University; Bangkok Thailand
| |
Collapse
|
38
|
Tahara Y, Akiyoshi K. Current advances in self-assembled nanogel delivery systems for immunotherapy. Adv Drug Deliv Rev 2015; 95:65-76. [PMID: 26482187 DOI: 10.1016/j.addr.2015.10.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/17/2015] [Accepted: 10/09/2015] [Indexed: 10/24/2022]
Abstract
Since nanogels (nanometer-sized gels) were developed two decades ago, they were utilized as carriers of innovative drug delivery systems. In particular, immunological drug delivery via self-assembled nanogels (self-nanogels) owing to their nanometer size and molecular chaperon-like ability to encapsulate large biomolecules is one of the most well studied and successful applications of nanogels. In the present review, we focus on self-nanogel applications as immunological drug delivery systems for cancer vaccines, cytokine delivery, nasal vaccines, and nucleic acid delivery, including several clinical trials. Cancer vaccines were the first practical application of self-nanogels as vehicles for drug delivery. After successful pre-clinical studies, phase I clinical trials were conducted, and it was found that vaccines consisting of self-nanogels could be administered repeatedly to humans without serious adverse effects, and self-nanogel vaccines induced antigen-specific cellular and humoral immunity. Cytokine delivery via self-nanogels led to the sustained release of IL-12, suppressed tumor growth, and increased Th1-type immune responses. Cationic self-nanogels were effective in penetrating the nasal mucosa and resulted in successful nasal vaccines in mice and nonhuman primates. Cationic self-nanogels were also used for the intracellular delivery of proteins and nucleic acids, and were successfully used to knockdown tumor growth factor expression using short interfering RNA with the immunological effect. These studies suggest that self-nanogels are currently one of the most unique and attractive immunological drug delivery systems and are edging closer to practical use.
Collapse
|
39
|
O'Neill EC, Stevenson CEM, Tantanarat K, Latousakis D, Donaldson MI, Rejzek M, Nepogodiev SA, Limpaseni T, Field RA, Lawson DM. Structural Dissection of the Maltodextrin Disproportionation Cycle of the Arabidopsis Plastidial Disproportionating Enzyme 1 (DPE1). J Biol Chem 2015; 290:29834-53. [PMID: 26504082 PMCID: PMC4705983 DOI: 10.1074/jbc.m115.682245] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Indexed: 11/06/2022] Open
Abstract
The degradation of transitory starch in the chloroplast to provide fuel for the plant during the night requires a suite of enzymes that generate a series of short chain linear glucans. However, glucans of less than four glucose units are no longer substrates for these enzymes, whereas export from the plastid is only possible in the form of either maltose or glucose. In order to make use of maltotriose, which would otherwise accumulate, disproportionating enzyme 1 (DPE1; a 4-α-glucanotransferase) converts two molecules of maltotriose to a molecule of maltopentaose, which can now be acted on by the degradative enzymes, and one molecule of glucose that can be exported. We have determined the structure of the Arabidopsis plastidial DPE1 (AtDPE1), and, through ligand soaking experiments, we have trapped the enzyme in a variety of conformational states. AtDPE1 forms a homodimer with a deep, long, and open-ended active site canyon contained within each subunit. The canyon is divided into donor and acceptor sites with the catalytic residues at their junction; a number of loops around the active site adopt different conformations dependent on the occupancy of these sites. The "gate" is the most dynamic loop and appears to play a role in substrate capture, in particular in the binding of the acceptor molecule. Subtle changes in the configuration of the active site residues may prevent undesirable reactions or abortive hydrolysis of the covalently bound enzyme-substrate intermediate. Together, these observations allow us to delineate the complete AtDPE1 disproportionation cycle in structural terms.
Collapse
Affiliation(s)
- Ellis C O'Neill
- From the Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom and
| | - Clare E M Stevenson
- From the Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom and
| | - Krit Tantanarat
- the Starch and Cyclodextrin Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Dimitrios Latousakis
- From the Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom and
| | - Matthew I Donaldson
- From the Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom and
| | - Martin Rejzek
- From the Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom and
| | - Sergey A Nepogodiev
- From the Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom and
| | - Tipaporn Limpaseni
- the Starch and Cyclodextrin Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| | - Robert A Field
- From the Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom and
| | - David M Lawson
- From the Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom and
| |
Collapse
|
40
|
Lin J, Pang H, Guo X, Ding Y, Geng J, Zhang J, Min J. Lentivirus-Mediated RNAi Silencing of VEGF Inhibits Angiogenesis and Growth of Renal Cell Carcinoma in a Nude Mouse Xenograft Model. DNA Cell Biol 2015; 34:717-27. [PMID: 26465082 DOI: 10.1089/dna.2015.2918] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
To construct and screen short hairpin RNA (shRNA) targeting vascular endothelial growth factor (VEGF), and investigate potential values of VEGF-shRNA on angiogenesis and growth in renal cell carcinoma (RCC) in a xenograft tumor model. VEGF-shRNA fragment was designed to connect plasmid vector, and RCC cells were transfected with shRNA. Real-time fluorescent quantitative polymerase chain reaction (RTFQ-PCR) was used to detect interference efficiency of VEGF gene. The xenograft tumor model was established in nude mice, and mice were randomly divided into blank control (BC) group, negative control (NC) group, and experimental group. RNA interference (RNAi) effect was detected by immunohistochemistry, and tumor volume changes were observed. Tumor-bearing nude mice model was established and mice were randomly divided into BC group, NC group, and treatment group. The tumor volume changes and tumor inhibition rate were recorded, and angiogenesis status was observed. The apoptosis of tumor cells and genetic toxicity of VEGF-shRNA were detected. VEGF-shRNA can inhibit VEGF mRNA expression with an inhibition ratio of 72.3%. Compared with NC group and BC group, experimental group presents smaller tumor volume, weight, and poor growth (all p < 0.05). Positive VEGF rate in experimental group is significantly lower than that in NC group and BC group (all p < 0.05). Significantly lower tumor volume, less microvessel density (MVD) value, and higher apoptotic index (AI) are found in treatment group compared with BC group and NC group (all p < 0.05). There was no significant difference in AI between treatment group and BC group regarding adjacent normal tissues (p > 0.05). VEGF plays an important role in the occurrence and development of RCC, chemical synthesis of VEGF small interfering RNA (siRNA) can specifically inhibit VEGF expression, angiogenesis and growth in RCC, and can promote cell apoptosis without genetic toxicity to normal tissues.
Collapse
Affiliation(s)
- Jiahua Lin
- 1 Cadet Brigade, The Fourth Military Medical University , Xi'an, People's Republic of China
| | - Hailin Pang
- 2 Department of Oncology, Tangdu Hospital, The Fourth Military Medical University , Xi'an, People's Republic of China
| | - Xiaojian Guo
- 1 Cadet Brigade, The Fourth Military Medical University , Xi'an, People's Republic of China
| | - Yunfei Ding
- 1 Cadet Brigade, The Fourth Military Medical University , Xi'an, People's Republic of China
| | - Jiaxu Geng
- 1 Cadet Brigade, The Fourth Military Medical University , Xi'an, People's Republic of China
| | - Jingmeng Zhang
- 1 Cadet Brigade, The Fourth Military Medical University , Xi'an, People's Republic of China
| | - Jie Min
- 2 Department of Oncology, Tangdu Hospital, The Fourth Military Medical University , Xi'an, People's Republic of China
| |
Collapse
|
41
|
Nishimura T, Umezaki K, Mukai SA, Sawada SI, Akiyoshi K. Amylose-Based Cationic Star Polymers for siRNA Delivery. BIOMED RESEARCH INTERNATIONAL 2015; 2015:962941. [PMID: 26539548 PMCID: PMC4619918 DOI: 10.1155/2015/962941] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/05/2015] [Accepted: 08/11/2015] [Indexed: 11/18/2022]
Abstract
A new siRNA delivery system using a cationic glyco-star polymer is described. Spermine-modified 8-arm amylose star polymer (with a degree of polymerization of approximately 60 per arm) was synthesized by chemoenzymatic methods. The cationic star polymer effectively bound to siRNA and formed spherical complexes with an average hydrodynamic diameter of 230 nm. The cationic 8-arm star polymer complexes showed superior cellular uptake characteristics and higher gene silencing effects than a cationic 1-arm polymer. These results suggest that amylose-based star polymers are a promising nanoplatform for glycobiomaterials.
Collapse
Affiliation(s)
- Tomoki Nishimura
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo, Kyoto 615-8150, Japan
- ERATO Akiyoshi Bio-Nanotransporter Project, JST, Katsura, Nishikyo, Kyoto 615-8150, Japan
| | - Kaori Umezaki
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo, Kyoto 615-8150, Japan
- ERATO Akiyoshi Bio-Nanotransporter Project, JST, Katsura, Nishikyo, Kyoto 615-8150, Japan
| | - Sada-atsu Mukai
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo, Kyoto 615-8150, Japan
- ERATO Akiyoshi Bio-Nanotransporter Project, JST, Katsura, Nishikyo, Kyoto 615-8150, Japan
| | - Shin-ichi Sawada
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo, Kyoto 615-8150, Japan
- ERATO Akiyoshi Bio-Nanotransporter Project, JST, Katsura, Nishikyo, Kyoto 615-8150, Japan
| | - Kazunari Akiyoshi
- Department of Polymer Chemistry, Graduate School of Engineering, Kyoto University, Katsura, Nishikyo, Kyoto 615-8150, Japan
- ERATO Akiyoshi Bio-Nanotransporter Project, JST, Katsura, Nishikyo, Kyoto 615-8150, Japan
| |
Collapse
|