1
|
Tayebi-Khorrami V, Fadaei MR, Fallahianshafiei S, Askari VR. Immune checkpoint blocking in cancer therapy using thermosensitive hydrogels: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04171-2. [PMID: 40314764 DOI: 10.1007/s00210-025-04171-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/09/2025] [Indexed: 05/03/2025]
Abstract
Cancer is a challenging issue requiring new strategies for management and control. Immune checkpoint blockades (ICBs) increase the body's immune response against cancer by targeting specific receptors on T-lymphocytes. The FDA approved different ICBs for cancer treatment: anti-PD-1, PDL-1, and CTLA-4 inhibitors. Many immune checkpoint inhibitors (ICIs) are in clinical trials, highlighting their significance. Challenges like resistance and side effects have led researchers to explore new delivery strategies for ICIs. Thermosensitive hydrogels can change from sol to gel and vice versa due to their structure. They interact with aqueous medium through groups like ethyl, methyl, and propyl, forming hydrogen bonds. These bonds of hydrogen are temperature-sensitive and cause the change of the polymer from sol to gel at a temperature named critical solution temperature (CST). The using temperature-responsive polymers and ICBs showed a promising approach to sustained localized cancer therapy with lowering side effects on normal tissues. In this paper, we first define new investigations on immune therapy in cancer via ICBs. Then, we present recent studies of thermosensitive polymers in cancer therapy and the most used thermosensitive polymers in studies. Eventually, we discuss studies that used thermosensitive polymers in the delivery of ICBs and discuss new investigations in this field.
Collapse
Affiliation(s)
- Vahid Tayebi-Khorrami
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Reza Fadaei
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Vahid Reza Askari
- Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Zhao H, Jiang S, Zhai S, Lv X, Shi C, Chen Y, Zhang R. Recent advances of intelligent polymer gels as active carriers for medical imaging-guided cancer therapy: A review. Int J Biol Macromol 2025; 301:140451. [PMID: 39884629 DOI: 10.1016/j.ijbiomac.2025.140451] [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: 09/12/2024] [Revised: 01/21/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
Cancer is a major global health challenges and various imaging-guided techniques are gaining prominence for its diagnosis and therapy with the advantages of high sensitivity, spatial resolution, and quantitative capability. The effectiveness of these approaches mainly relies on the establishment of a responsive platform. The intelligent polymer gel composites as the active carriers have drawn considerable attention owing to their outstanding versatility, three-layer network structure, mechanical adjustability and so forth, which may be contribute to loading drug, imaging probe, and targeting biomarkers in medical imaging-guided cancer therapy (IGCT). In this review, significant progress has been highlighted in fabrication of intelligent polymer gel composites with multi-functional molecules for the anticancer drugs delivery systems, multimodal imaging-guided management and developing molecular devices in various tumors. In addition, we also explore the current challenges and future development directions of these composites as carriers with the assistance of IGCT for precise anti-cancer applications in clinical practice.
Collapse
Affiliation(s)
- Huifang Zhao
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China; School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China.
| | - Siyi Jiang
- First Clinical Medical College of Shanxi Medical University, Taiyuan 030001, China
| | - Shuyu Zhai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan 030032, China
| | - Xiaoyang Lv
- First Clinical Medical College of Shanxi Medical University, Taiyuan 030001, China
| | - Chaoqun Shi
- School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China
| | - Yi Chen
- School of Basic Medical Science, Shanxi Medical University, Taiyuan 030001, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital, Fifth Hospital of Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
3
|
Szupryczyński K, Czeleń P, Jeliński T, Szefler B. What is the Reason That the Pharmacological Future of Chemotherapeutics in the Treatment of Lung Cancer Could Be Most Closely Related to Nanostructures? Platinum Drugs in Therapy of Non-Small and Small Cell Lung Cancer and Their Unexpected, Possible Interactions. The Review. Int J Nanomedicine 2024; 19:9503-9547. [PMID: 39296940 PMCID: PMC11410046 DOI: 10.2147/ijn.s469217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/19/2024] [Indexed: 09/21/2024] Open
Abstract
Over the course of several decades, anticancer treatment with chemotherapy drugs for lung cancer has not changed significantly. Unfortunately, this treatment prolongs the patient's life only by a few months, causing many side effects in the human body. It has also been proven that drugs such as Cisplatin, Carboplatin, Oxaliplatin and others can react with other substances containing an aromatic ring in which the nitrogen atom has a free electron group in its structure. Thus, such structures may have a competitive effect on the nucleobases of DNA. Therefore, scientists are looking not only for new drugs, but also for new alternative ways of delivering the drug to the cancer site. Nanotechnology seems to be a great hope in this matter. Creating a new nanomedicine would reduce the dose of the drug to an absolute minimum, and thus limit the toxic effect of the drug; it would allow for the exclusion of interactions with competitive compounds with a structure similar to nucleobases; it would also permit using the so-called targeted treatment and bypassing healthy cells; it would allow for the introduction of other treatment options, such as radiotherapy directly to the cancer site; and it would provide diagnostic possibilities. This article is a review that aims to systematize the knowledge regarding the anticancer treatment of lung cancer, but not only. It shows the clear possibility of interactions of chemotherapeutics with compounds competitive to the nitrogenous bases of DNA. It also shows the possibilities of using nanostructures as potential Platinum drug carriers, and proves that nanomedicine can easily become a new medicinal product in personalized medicine.
Collapse
Affiliation(s)
- Kamil Szupryczyński
- Doctoral School of Medical and Health Sciences, Faculty of Pharmacy, Collegium Medicum, Nicolaus, Copernicus University, Bydgoszcz, Poland
| | - Przemysław Czeleń
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Jeliński
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Beata Szefler
- Department of Physical Chemistry, Faculty of Pharmacy, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
4
|
Dhote NS, Patel RD, Kuwar U, Agrawal M, Alexander A, Jain P, Ajazuddin. Application of Thermoresponsive Smart Polymers based in situ Gel as a Novel Carrier for Tumor Targeting. Curr Cancer Drug Targets 2024; 24:375-396. [PMID: 37534485 DOI: 10.2174/1568009623666230803111718] [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: 03/07/2023] [Revised: 05/12/2023] [Accepted: 06/08/2023] [Indexed: 08/04/2023]
Abstract
The temperature-triggered in situ gelling system has been revolutionized by introducing an intelligent polymeric system. Temperature-triggered polymer solutions are initially in a sol state and then undergo a phase transition to form a gel at body temperature due to various parameters like pH, temperature, and so on. These smart polymers offer a number of advantages, including ease of administration, long duration of release of the drug, low administration frequency with good patient compliance, and targeted drug delivery with fewer adverse effects. Polymers such as poly(N-isopropylacrylamide) (PNIPAAm), polyethylene glycol (PEG), poly (N, N'-diethyl acrylamide), and polyoxypropylene (PPO) have been briefly discussed. In addition to various novel Drug Delivery Systems (DDS), the smart temperature-triggered polymeric system has various applications in cancer therapy and many other disease conditions. This review focuses on the principals involved in situ gelling systems using various temperature-triggered polymers for chemotherapeutic purposes, using smart DDS, and their advanced application in cancer therapy, as well as available marketed formulations and recent advances in these thermoresponsive sol-gel transforming systems.
Collapse
Affiliation(s)
- Nidhi Sudhir Dhote
- Department of Pharmaceutics, School of Pharmacy & Technology Management, Narsee Monjee Institute of Management Studies (NMIMS), Shirpur, 425 405, Maharashtra, India
| | - Rajat Dineshbhai Patel
- Department of Pharmaceutics, School of Pharmacy & Technology Management, Narsee Monjee Institute of Management Studies (NMIMS), Shirpur, 425 405, Maharashtra, India
| | - Utkarsha Kuwar
- Department of Pharmaceutics, School of Pharmacy & Technology Management, Narsee Monjee Institute of Management Studies (NMIMS), Shirpur, 425 405, Maharashtra, India
| | - Mukta Agrawal
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Hyderabad, 509 301, Telangana, India
| | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam, 781101, India
| | - Parag Jain
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh, 490024, India
| | - Ajazuddin
- Rungta College of Pharmaceutical Sciences and Research, Kohka-Kurud Road, Bhilai, Chhattisgarh, 490024, India
| |
Collapse
|
5
|
de Dios-Pérez I, González-Garcinuño Á, Tabernero A, Blanco-López M, García-Esteban JA, Moreno-Rodilla V, Curto B, Pérez-Esteban P, Martín Del Valle EM. Development of a thermosensitive hydrogel based on Polaxamer 407 and gellan gum with inclusion complexes (Sulfobutylated-β-cyclodextrin-Farnesol) as a local drug delivery system. Eur J Pharm Sci 2023; 191:106618. [PMID: 37866674 DOI: 10.1016/j.ejps.2023.106618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 09/12/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
This work proposes the development of a thermosensitive local drug release system based on Polaxamer 407, also known as Pluronic® F-127 (PF-127), Gellan Gum (GG) and the inclusion complex Sulfobutylated-β-cyclodextrin (CD) with Farnesol (FOH). Rheological properties of the hydrogels and their degradation were studied. According to the rheological results, a solution of 20% w/v of PF-127 forms a strong gel with a gelling temperature of about 25 °C (storage modulus of 15,000 Pa). The addition of the GG increased the storage modulus (optimal concentration of 0.5 % w/v) twofold without modifying the gelling temperature. Moreover, including 0.5% w/v of GG also increased 6 times the degradation time of the hydrogel. Regarding the inclusion complex, the addition of free CD decreased the viscosity and the gel strength since polymer chains were included in CD cavity without affecting the gelling temperature. Contrarily, the inclusion complex CD-FOH did not significantly modify any property of the formulation because the FOH was hosted in the CD. Furthermore, a mathematical model was developed to adjust the degradation time. This model highlights that the addition of the GG decreases the number of released chains from the polymeric network (which coincides with an increase in the storage modulus) and that the free CD reduces the degradation rate, protecting the polymeric chains. Finally, FOH release was quantified with a specific device, that was designed and printed for this type of system, observing a sustainable drug release (similar to FOH aqueous solubility, 8 μM) dependent on polymer degradation.
Collapse
Affiliation(s)
| | - Álvaro González-Garcinuño
- Department of Chemical Engineering, University of Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Spain
| | - Antonio Tabernero
- Department of Chemical Engineering, University of Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Spain
| | | | | | | | - Belén Curto
- Department of Computer Science and Automation, University of Salamanca, Spain
| | - Patricia Pérez-Esteban
- Institute of Translational Medicine, Heritage Building, Mindelsohn Way, Birmingham, B15 2TH, England
| | - Eva M Martín Del Valle
- Department of Chemical Engineering, University of Salamanca, Spain; Institute for Biomedical Research of Salamanca (IBSAL), Spain.
| |
Collapse
|
6
|
Exploration of hemocompatibility and intratumoral accumulation of paclitaxel after loco-regional administration of thermoresponsive hydrogel composed of poloxamer and xanthan gum: An application to dose-dense chemotherapy. Int J Biol Macromol 2023; 226:746-759. [PMID: 36495991 DOI: 10.1016/j.ijbiomac.2022.11.285] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 11/11/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
Although paclitaxel is a front-line chemotherapeutic agent for the treatment of metastatic breast cancer, its intravenous therapy produces deleterious adverse effects. In an attempt to address the issue, the present study aimed to develop a paclitaxel loaded thermosensitive/thermoresponsive hydrogel (PTXNp-TGel) for loco-regional administration to breast tumors to provide dose-dense chemotherapy. Poloxamer and xanthan gum were used to prepare TGel by the cold method. In vitro and in vivo performance of PTXNp-TGel was compared with TGel, pure drug loaded TGel (PTX-TGel) and marketed formulation, Taxol®. The formulated PTXNp-TGel showed acceptable gelation temperature and time (37 °C and 57 s), lower viscosity at room temperature and higher viscosity at body temperature to support sol-gel transition with increasing temperature, and sustained drug release up to 21 days. Additionally, PTXNp-TGel showed negligible hemolytic toxicity as compared to PTX-TGel and Taxol®. Intratumoral administration of PTXNp-TGel produced significantly higher antitumor activity as indicated by lowest relative tumor volume (1.50) and relative antitumor proliferation rate (27.71 %) in comparison with PTX-TGel, Taxol®, and PTXNp (p < 0.05). Finally, insignificant body weight loss during the experimental period, lack of hematotoxicity, nephrotoxicity, and hepatotoxicity imply improved therapeutic performance of the locally administrated dose-dense therapy of PTXNp-TGel as compared to Taxol®.
Collapse
|
7
|
Sharma A, Shambhwani D, Pandey S, Singh J, Lalhlenmawia H, Kumarasamy M, Singh SK, Chellappan DK, Gupta G, Prasher P, Dua K, Kumar D. Advances in Lung Cancer Treatment Using Nanomedicines. ACS OMEGA 2023; 8:10-41. [PMID: 36643475 PMCID: PMC9835549 DOI: 10.1021/acsomega.2c04078] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 10/13/2022] [Indexed: 06/01/2023]
Abstract
Carcinoma of the lungs is among the most menacing forms of malignancy and has a poor prognosis, with a low overall survival rate due to delayed detection and ineffectiveness of conventional therapy. Therefore, drug delivery strategies that may overcome undesired damage to healthy cells, boost therapeutic efficacy, and act as imaging tools are currently gaining much attention. Advances in material science have resulted in unique nanoscale-based theranostic agents, which provide renewed hope for patients suffering from lung cancer. Nanotechnology has vastly modified and upgraded the existing techniques, focusing primarily on increasing bioavailability and stability of anti-cancer drugs. Nanocarrier-based imaging systems as theranostic tools in the treatment of lung carcinoma have proven to possess considerable benefits, such as early detection and targeted therapeutic delivery for effectively treating lung cancer. Several variants of nano-drug delivery agents have been successfully studied for therapeutic applications, such as liposomes, dendrimers, polymeric nanoparticles, nanoemulsions, carbon nanotubes, gold nanoparticles, magnetic nanoparticles, solid lipid nanoparticles, hydrogels, and micelles. In this Review, we present a comprehensive outline on the various types of overexpressed receptors in lung cancer, as well as the various targeting approaches of nanoparticles.
Collapse
Affiliation(s)
- Akshansh Sharma
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
| | | | - Sadanand Pandey
- Department
of Chemistry, College of Natural Sciences, Yeungnam University, Gyeongsan, Gyeongbuk 38541, South Korea
| | - Jay Singh
- Department
of Chemistry, Institute of Science, Banaras
Hindu University, Varanasi 221005, India
| | - Hauzel Lalhlenmawia
- Department
of Pharmacy, Regional Institute of Paramedical
and Nursing Sciences, Zemabawk, Aizawl, Mizoram 796017, India
| | - Murali Kumarasamy
- Department
of Biotechnology, National Institute of
Pharmaceutical Education and Research, Hajipur 844102, India
| | - Sachin Kumar Singh
- School
of Pharmaceutical Sciences, Lovely Professional
University, Phagwara 144411, India
- Faculty
of Health, Australian Research Centre in Complementary and Integrative
Medicine, University of Technology, Sydney, Ultimo-NSW 2007, Australia
| | - Dinesh Kumar Chellappan
- Department
of Life Sciences, School of Pharmacy, International
Medical University, Kuala Lumpur 57000, Malaysia
| | - Gaurav Gupta
- Department
of Pharmacology, School of Pharmacy, Suresh
Gyan Vihar University, Jaipur 302017, India
- Department
of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical
and Technical Sciences, Saveetha University, Chennai 602117, India
- Uttaranchal
Institute of Pharmaceutical Sciences, Uttaranchal
University, Dehradun 248007, India
| | - Parteek Prasher
- Department
of Chemistry, University of Petroleum &
Energy Studies, Dehradun 248007, India
| | - Kamal Dua
- Faculty
of Health, Australian Research Centre 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
| | - Deepak Kumar
- Department
of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Shoolini University, Solan 173229, India
| |
Collapse
|
8
|
Peng W, Yao C, Pan Q, Zhang Z, Ye J, Shen B, Zhou G, Fang Y. Novel considerations on EGFR-based therapy as a contributor to cancer cell death in NSCLC. Front Oncol 2023; 13:1120278. [PMID: 36910653 PMCID: PMC9995697 DOI: 10.3389/fonc.2023.1120278] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/10/2023] [Indexed: 02/25/2023] Open
Abstract
Epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitors (TKIs) represented by gefitinib and erlotinib are widely used in treating non-small cell lung cancer (NSCLC). However, acquired resistance to EGFR-TKI treatment remains a clinical challenge. In recent years, emerging research investigated in EGFR-TKI-based combination therapy regimens, and remarkable achievements have been reported. This article focuses on EGFR-TKI-based regimens, reviews the standard and novel application of EGFR targets, and summarizes the mechanisms of EGFR-TKI combinations including chemotherapy, anti-vascular endothelial growth factor monoclonal antibodies, and immunotherapy in the treatment of NSCLC. Additionally, we summarize clinical trials of EGFR-TKI-based combination therapy expanding indications to EGFR mutation-negative lung malignancies. Moreover, novel strategies are under research to explore new drugs with good biocompatibility. Nanoparticles encapsulating non-coding RNA and chemotherapy of new dosage forms drawn great attention and showed promising prospects in effective delivery and stable release. Overall, as the development of resistance to EGFR-TKIs treatment is inevitable in most of the cases, further research is needed to clarify the underlying mechanism of the resistance, and to evaluate and establish EGFR-TKI combination therapies to diversify the treatment landscape for NSCLC.
Collapse
Affiliation(s)
- Weiwei Peng
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Chengyun Yao
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Qin Pan
- Department of Medical Oncology, Liyang People's Hospital, Liyang, China
| | - Zhi Zhang
- Department of Thoracic Surgery, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Jinjun Ye
- Department of Radiation Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Bo Shen
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Guoren Zhou
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| | - Ying Fang
- Department of Medical Oncology, Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
9
|
Dinakar YH, Karole A, Parvez S, Jain V, Mudavath SL. Organ-restricted delivery through stimuli-responsive nanocarriers for lung cancer therapy. Life Sci 2022; 310:121133. [DOI: 10.1016/j.lfs.2022.121133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/22/2022] [Accepted: 10/24/2022] [Indexed: 11/07/2022]
|
10
|
Zheng S, Cai Y, Hong Y, Gong Y, Gao L, Li Q, Li L, Sun X. Legumain/pH dual-responsive lytic peptide-paclitaxel conjugate for synergistic cancer therapy. Drug Deliv 2022; 29:1764-1775. [PMID: 35638851 PMCID: PMC9176665 DOI: 10.1080/10717544.2022.2081380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
After molecule targeted drug, monoclonal antibody and antibody–drug conjugates (ADCs), peptide–drug conjugates (PDCs) have become the next generation targeted anti-tumor drugs due to its properties of low molecule weight, efficient cell penetration, low immunogenicity, good pharmacokinetic and large-scale synthesis by solid phase synthesis. Herein, we present a lytic peptide PTP7-drug paclitaxel conjugate assembling nanoparticles (named PPP) that can sequentially respond to dual stimuli in the tumor microenvironment, which was designed for passive tumor-targeted delivery and on-demand release of a tumor lytic peptide (PTP-7) as well as a chemotherapeutic agent of paclitaxel (PTX). To achieve this, tumor lytic peptide PTP-7 was connected with polyethylene glycol by a peptide substrate of legumain to serve as hydrophobic segments of nanoparticles to protect the peptide from enzymatic degradation. After that, PTX was connected to the amino group of the polypeptide side chain through an acid-responsive chemical bond (2-propionic-3-methylmaleic anhydride, CDM). Therefore, the nanoparticle (PPP) collapsed when it encountered the weakly acidic tumor microenvironment where PTX molecules fell off, and further triggered the cleavage of the peptide substrate by legumain that is highly expressed in tumor stroma and tumor cell surface. Moreover, PPP presents improved stability, improved drug solubility, prolonged blood circulation and significant inhibition ability on tumor growth, which gives a reasonable strategy to accurately deliver small molecule drugs and active peptides simultaneously to tumor sites.
Collapse
Affiliation(s)
- Shanshan Zheng
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Yue Cai
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Yulu Hong
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Yubei Gong
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Licheng Gao
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Qingyong Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Le Li
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Xuanrong Sun
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals & College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| |
Collapse
|
11
|
Dethe MR, A P, Ahmed H, Agrawal M, Roy U, Alexander A. PCL-PEG copolymer based injectable thermosensitive hydrogels. J Control Release 2022; 343:217-236. [PMID: 35090961 PMCID: PMC9134269 DOI: 10.1016/j.jconrel.2022.01.035] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 01/09/2023]
Abstract
A number of stimuli-responsive-based hydrogels has been widely explored in biomedical applications in the last few decades because of their excellent biodegradability and biocompatibility. The development of synthetic chemistry and materials science leads to the emergence of in situ stimuli-responsive hydrogels. In this regard, several synthetic and natural polymers have been synthesized and utilized to prepare temperature-sensitive in situ forming hydrogels. This could be best used via injections as temperature stimulus could trigger in situ hydrogels gelation and swelling behaviors. There are many smart polymers available for the formulation of the in situ based thermoresponsive injectable hydrogel. Among these, poly (ε-caprolactone) (PCL) polymer has been recognized and approved by the FDA for numerous biomedical applications. More specifically, the PCL is coupled with polyethylene glycol (PEG) to obtain amphiphilic thermosensitive "smart" copolymers (PCL-PEG), to form rapid and reversible physical gelation behavior. However, the chemical structure of the copolymer is a critical aspect in determining water solubility, thermo-gelation behavior, drug release rate, degradation rate, and the possibility to deliver a diverse range of drugs. In this review, we have highlighted the typical PCL-PEG-based thermosensitive injectable hydrogels progress in the last decade for tissue engineering and localized drug delivery applications to treat various diseases. Additionally, the impact of molecular weight of PCL-PEG upon gelling behavior has also been critically highlighted for optimum hydrogels properties for potential pharmaceutical and biomedical applications.
Collapse
Affiliation(s)
- Mithun Rajendra Dethe
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati 781101, India
| | - Prabakaran A
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati 781101, India
| | - Hafiz Ahmed
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati 781101, India
| | - Mukta Agrawal
- School of Pharmacy & Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS), Polepally SEZ, TSIIC Jadcherla, Hyderabad 509301, India
| | - Upal Roy
- Department of Health and Biomedical Sciences, College of Health Affairs, One West University Blvd., Brownsville, TX 78520, United States of America
| | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati 781101, India.
| |
Collapse
|
12
|
Mohammadi M, Karimi M, Malaekeh-Nikouei B, Torkashvand M, Alibolandi M. Hybrid in situ- forming injectable hydrogels for local cancer therapy. Int J Pharm 2022; 616:121534. [PMID: 35124117 DOI: 10.1016/j.ijpharm.2022.121534] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/16/2022] [Accepted: 01/28/2022] [Indexed: 01/17/2023]
Abstract
Injectable in situ forming hydrogels are amongst the efficient local drug delivery systems for cancer therapy. Providing a 3D hydrogel network within the target tissue capable of sustained release of the chemotherapeutics made them attractive candidates for increasing the therapeutic index. Remarkable swelling properties, mechanical strength, biocompatibility, wide composition variety and tunable polymeric moieties have led to preparation of injectable hydrogels which also could be used as cavity adaptive chemotherapeutic-loaded implants to prevent post -surgical cancer recurrence. Implementation of various polymers, nanoparticles, peptide and proteins and different crosslinking chemistry facilitated the fabrication of hybrid hydrogels with favorable characteristics such as stimuli sensitive platforms or multifunctional systems. In the current review, we focused on design and fabrication strategies of injectable in situ forming hydrogels and summarized recent hybrid hydrogels used for local cancer therapy.
Collapse
Affiliation(s)
- Marzieh Mohammadi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Malihe Karimi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bizhan Malaekeh-Nikouei
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Torkashvand
- Fouman Faculty of Engineering, College of Engineering, University of Tehran, Tehran, Iran
| | - Mona Alibolandi
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
13
|
Hani U, M. YB, Wahab S, Siddiqua A, Osmani RAM, Rahamathulla M. A Comprehensive Review of Current Perspectives on Novel Drug Delivery Systems and Approaches for Lung Cancer Management. J Pharm Innov 2021. [DOI: 10.1007/s12247-021-09582-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
14
|
Othman M, Mahmud K, Mohammed R, Mohd Noor SNF, Tuan Din SA, Zabidi MA. Encapsulation of hemoglobin within mPEG- b-PCL micelle for development of artificial oxygen carrier. INT J POLYM MATER PO 2021. [DOI: 10.1080/00914037.2021.1915782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Mislia Othman
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Khadijah Mahmud
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Rafeezul Mohammed
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Siti Noor Fazliah Mohd Noor
- Biomaterial and Craniofacial Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Sharifah Azdiana Tuan Din
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| | - Muhammad Azrul Zabidi
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Malaysia
| |
Collapse
|
15
|
Esmaeili J, Barati A, Ai J, Nooshabadi VT, Mirzaei Z. Employing hydrogels in tissue engineering approaches to boost conventional cancer-based research and therapies. RSC Adv 2021; 11:10646-10669. [PMID: 35423538 PMCID: PMC8695814 DOI: 10.1039/d1ra00855b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 02/22/2021] [Indexed: 12/17/2022] Open
Abstract
Cancer is a complicated disease that involves the efforts of researchers to introduce and investigate novel successful treatments. Traditional cancer therapy approaches, especially chemotherapy, are prone to possible systemic side effects, such as the dysfunction of liver or kidney, neurological side effects and a decrease of bone marrow activity. Hydrogels, along with tissue engineering techniques, provide tremendous potential for scientists to overcome these issues through the release of drugs at the site of tumor. Hydrogels demonstrated competency as potent and stimulus-sensitive drug delivery systems for tumor removal, which is attributed to their unique features, including high water content, biocompatibility, and biodegradability. In addition, hydrogels have gained more attention as 3D models for easier and faster screening of cancer and tumors due to their potential in mimicking the extracellular matrix. Hydrogels as a reservoir can be loaded by an effective dosage of chemotherapeutic agents, and then deliver them to targets. In comparison to conventional procedures, hydrogels considerably decreased the total cost, duration of research, and treatment time. This study provides a general look into the potential role of hydrogels as a powerful tool to augment cancer studies for better analysis of cancerous cell functions, cell survival, angiogenesis, metastasis, and drug screening. Moreover, the upstanding application of drug delivery systems related to the hydrogel in order to sustain the release of desired drugs in the tumor cell-site were explored.
Collapse
Affiliation(s)
- Javad Esmaeili
- Department of Chemical Engineering, Faculty of Engineering, Arak University Arak Iran
- Department of Tissue Engineering, TISSUEHUB CO. Tehran Iran
| | - Abolfazl Barati
- Department of Chemical Engineering, Faculty of Engineering, Arak University Arak Iran
| | - Jafar Ai
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Technologies, Tehran University of Medical Sciences Tehran 14177-55469 Iran
| | - Vajihe Taghdiri Nooshabadi
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Technologies, Tehran University of Medical Sciences Tehran 14177-55469 Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences Semnan Iran
| | - Zeynab Mirzaei
- Faculty of Biomedical Engineering, Amirkabir University of Technology Hafez str. 424 Tehran Iran
- Department of Tissue Engineering, TISSUEHUB CO. Tehran Iran
| |
Collapse
|
16
|
Wei W, Li H, Yin C, Tang F. Research progress in the application of in situ hydrogel system in tumor treatment. Drug Deliv 2020; 27:460-468. [PMID: 32166987 PMCID: PMC7144265 DOI: 10.1080/10717544.2020.1739171] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/01/2020] [Accepted: 03/02/2020] [Indexed: 01/30/2023] Open
Abstract
The in situ hydrogel drug delivery system is a hot research topic in recent years. Combining both properties of hydrogel and solution, in situ hydrogels can provide many advantages for drug delivery application, including easy application, high local drug concentration, prolonged drug retention time, reduced drug dose in vivo, good biocompatibility and improved patient compliance, thus has potential in tumor treatment. In this paper, the related literature reports in recent years were reviewed to summarize and discuss the research progress and development prospects in the application of in situ hydrogels in tumor treatment.
Collapse
Affiliation(s)
- Weipeng Wei
- Department of Clinical Pharmacy, School of Pharmacy, Zunyi Medical University, Zunyi, China
- The Key Laboratory of Clinical Pharmacy in Zunyi City, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Hongfang Li
- Department of Clinical Pharmacy, School of Pharmacy, Zunyi Medical University, Zunyi, China
- The Key Laboratory of Clinical Pharmacy in Zunyi City, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Chengchen Yin
- Department of Clinical Pharmacy, School of Pharmacy, Zunyi Medical University, Zunyi, China
- The Key Laboratory of Clinical Pharmacy in Zunyi City, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Fushan Tang
- Department of Clinical Pharmacy, School of Pharmacy, Zunyi Medical University, Zunyi, China
- The Key Laboratory of Clinical Pharmacy in Zunyi City, Zunyi, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
17
|
Tong CWS, Wu MMX, Yan VW, Cho WCS, To KKW. Repurposing loperamide to overcome gefitinib resistance by triggering apoptosis independent of autophagy induction in KRAS mutant NSCLC cells. Cancer Treat Res Commun 2020; 25:100229. [PMID: 33152554 DOI: 10.1016/j.ctarc.2020.100229] [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: 09/02/2020] [Revised: 10/17/2020] [Accepted: 10/23/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND Gefitinib is an epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI) approved for first-line treatment of non-small cell lung cancer (NSCLC) with sensitizing EGFR mutations. However, NSCLC patients bearing mutant KRAS are inherently unresponsive to gefitinib. Defective autophagy was proposed to mediate resistance to EGFR-TKIs. In this study, the reversal of primary resistance to gefitinib in NSCLC by putative autophagy inducers was investigated. MATERIALS AND METHODS A few putative autophagy inducers were investigated in NSCLC cells harboring KRAS or EGFR mutations. Quantitative real-time PCR and Western blot analysis were used to evaluate expression of autophagy-related genes and proteins. Sulforhodamine B assay was used to evaluate cytotoxicity of drug combinations. Flow cytometric asssays were used to study apoptotic and cell cycle effects. RESULTS The antidiarrheal agent loperamide was identified as an autophagy inducer. Loperamide promoted the formation of autophagosomes and it potentiated the cytotoxic effect of gefitinib specifically in NSCLC cells bearing mutant KRAS and wild-type EGFR. Gefitinib-loperamide combination enhanced apoptosis and G1 cell cycle arrest, both of which could not be reversed by pharmacological autophagy inhibitor (3-methyladenine). Moreover, synergistic anticancer effect of gefitinib-loperamide combination was observed in both autophagy-proficient (Atg5-wild type) and -deficient (Atg5-knockout) mouse embryonic fibroblasts. Loperamide overcome gefitinib resistance in NSCLC harboring mutant KRAS and wild-type EGFR through increased apoptosis but independent of autophagy induction. CONCLUSION Loperamide could be repurposed to overcome primary resistance to gefitinib in KRAS-mutation bearing NSCLC as it also helps relieve the common side effect of diarrhea caused by EGFR-TKIs.
Collapse
Affiliation(s)
- Christy W S Tong
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mia M X Wu
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Vivi W Yan
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - William C S Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong SAR, China
| | - Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
18
|
Rossi SM, Ryan BK, Kelly HM. Evaluation of the activity of a chemo-ablative, thermoresponsive hydrogel in a murine xenograft model of lung cancer. Br J Cancer 2020; 123:369-377. [PMID: 32457364 PMCID: PMC7403591 DOI: 10.1038/s41416-020-0904-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/30/2019] [Accepted: 02/05/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Minimally invasive intratumoural administration of thermoresponsive hydrogels, that transition from liquid to gel in response to temperature, has been proposed as a potential treatment modality for solid tumours. The aim of this study was to assess the inherent cytotoxicity of a poloxamer-based thermoresponsive hydrogel in a murine xenograft model of lung cancer. METHODS In vitro viability assessment was carried out in a lung cancer (A549) and non-cancerous (Balb/c 3T3 clone A31) cell line. Following intratumoural administration of saline or the thermoresponsive hydrogel to an A549 xenograft model in female Athymic Nude-Foxn1nu mice (n = 6/group), localisation was confirmed using IVIS imaging. Tumour volume was assessed using callipers measurements over 14 days. Blood serum was analysed for liver and kidney damage and ex vivo tissue samples were histologically assessed. RESULTS The thermoresponsive hydrogel demonstrated a dose-dependent cancer cell-specific toxicity in vitro and was retained in situ for at least 14 days in the xenograft model. Tumour volume increase was statistically significantly lower than saline treated control at day 14 (n = 6, p = 0.0001), with no associated damage of hepatic or renal tissue observed. CONCLUSIONS Presented is a poloxamer-based thermoresponsive hydrogel, suitable for intratumoural administration and retention, which has demonstrated preliminary evidence of local tumour control, with minimal off-site toxicity.
Collapse
Affiliation(s)
- Seóna M Rossi
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen's Green, Dublin 2, Ireland
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen's Green, Dublin 2, Ireland
| | - Benedict K Ryan
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen's Green, Dublin 2, Ireland
| | - Helena M Kelly
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland (RCSI), 123 St. Stephen's Green, Dublin 2, Ireland.
- Tissue Engineering Research Group, Department of Anatomy, Royal College of Surgeons in Ireland (RCSI), 123 St Stephen's Green, Dublin 2, Ireland.
| |
Collapse
|
19
|
Otroj M, Taymouri S, Varshosaz J, Mirian M. Preparation and characterization of dry powder containing sunitinib loaded PHBV nanoparticles for enhanced pulmonary delivery. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101570] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
20
|
Agrawal NK, Allen P, Song YH, Wachs RA, Du Y, Ellington AD, Schmidt CE. Oligonucleotide-functionalized hydrogels for sustained release of small molecule (aptamer) therapeutics. Acta Biomater 2020; 102:315-325. [PMID: 31760222 DOI: 10.1016/j.actbio.2019.11.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 01/10/2023]
Abstract
Natural and synthetic hydrogels have been widely investigated as biomaterial scaffolds to promote tissue repair and regeneration. Nevertheless, the scaffold alone is often insufficient to drive new tissue growth, instead requiring continuous delivery of therapeutics, such as proteins or other biomolecules that work in concert with structural support provided by the scaffold. However, because of the high-water content, hydrogels tend to be permeable and cause rapid release of the encapsulated drug, which could lead to serious complications from local overdose and may result in the significant waste of encapsulated therapeutic(s). To this end, we designed an oligonucleotide-functionalized hydrogel that can provide sustained and controlled delivery of therapeutics for up to 4 weeks. To prove this concept, we successfully achieved sustained release (for over 28 days) of model anti-Nogo receptor (anti-NgR) RNA aptamer from oligonucleotide-functionalized hyaluronic acid-based hydrogel by changing the complementarity between the short antisense sequences and the aptamer. Furthermore, the released aptamer successfully blocked neuro-inhibitory effects of myelin-derived inhibitors and promoted neurite outgrowth from rat dorsal root ganglia in vitro. Because antisense sequences can be designed to bind to proteins, peptides, and aptamer, our oligonucleotide-functionalized hydrogel offers a promising therapeutic delivery system to obtain controlled release (both bolus and sustained) of various therapeutics for the treatment of complex diseases and injury models, such as spinal cord injury. STATEMENT OF SIGNIFICANCE: Producing a therapeutic effect often requires the administration of multiple injections with high dosages. This regimen causes discomfort to the patient and raises cost of treatment. Additionally, systemic delivery of therapeutics often results in adverse effects; therefore, local delivery at the site of injury is desirable. Therefore, in this study, we designed an oligonucleotide-functionalized biomaterial platform using ssDNA oligonucleotides (immobile species) as antisense sequences to increase residence time and fine-tune the release of anti-nogo receptor aptamer (mobile species) for spinal cord injury application. Because antisense sequences can be designed to bind proteins, peptides, and aptamer, our hydrogel offers a promising delivery system to obtain controlled release of various therapeutics for the treatment of complex diseases and injury models.
Collapse
|
21
|
Liu Z, Wang J. Biological Influence of Nonswelling Microgels on Cartilage Induction of Mouse Adipose-Derived Stem Cells. BIOMED RESEARCH INTERNATIONAL 2019; 2019:6508094. [PMID: 31737672 PMCID: PMC6815524 DOI: 10.1155/2019/6508094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 06/09/2019] [Accepted: 07/24/2019] [Indexed: 11/17/2022]
Abstract
In cartilage tissue engineering, the target cells' functional performance depends on the biomaterials. However, it is difficult to develop an appropriate scaffold to differentiate mouse adipose-derived stem cells (mADSCs) into chondrocyte despite an increasing number of studies on biological scaffold materials. The purpose of this study was to create a novel scaffold for mADSC culture and chondrogenic differentiation with a new series of microgels based on polyethyleneimine (PEI), polyethylene glycol (PEG), and poly (L-lactic acid) (PLLA) and able to resist swelling with changes in temperature, pH, and polymer concentration. The biocompatibility and ability of the nonswelling microgels were then examined and served as scaffolds for cell culture and for cartilage differentiation. The results show that the new microgels are a novel biomaterial that both retains its nonswelling properties under various conditions and facilitates important scaffold functions such as cell adhesion, proliferation, and cartilage induction.
Collapse
Affiliation(s)
- Zheng Liu
- Department of Spine Surgery, Xiangya Hospital, Central South University, 87, Xiangya Road, Changsha 410008, Hunan, China
- Hunan Engineering Laboratory of Advanced Artificial Osteo-Materials, Xiangya Hospital, Central South University, 87, Xiangya Road, Changsha 410008, Hunan, China
| | - Jun Wang
- Department of Spine Surgery, Xiangya Hospital, Central South University, 87, Xiangya Road, Changsha 410008, Hunan, China
- Hunan Engineering Laboratory of Advanced Artificial Osteo-Materials, Xiangya Hospital, Central South University, 87, Xiangya Road, Changsha 410008, Hunan, China
| |
Collapse
|
22
|
Biocompatibility and paclitaxel/cisplatin dual-loading of nanotubes prepared from poly(ethylene glycol)-polylactide-poly(ethylene glycol) triblock copolymers for combination cancer therapy. Saudi Pharm J 2019; 27:1025-1035. [PMID: 31997910 PMCID: PMC6978636 DOI: 10.1016/j.jsps.2019.08.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Accepted: 08/30/2019] [Indexed: 12/26/2022] Open
Abstract
Nanotubes were prepared by self-assembly of the copolymer using co-solvent evaporation method. The biocompatibility of the nanotubes was assessed in comparison with spherical micelles and filomicelles prepared from poly(ethylene glycol)-poly(L-lactide-co-glycolide) (PEG-PLGA) and poly(ethylene glycol)-poly(L-lactide) (PEG-PLA), respectively. Several aspects of biocompatibility of the aggregates were considered, including agar diffusion and MTT assay, release of cytokines, hemolysis, protein adsorption, dynamic clotting in vitro, and Zebrafish embryonic compatibility in vivo. The nanotubes present good cell compatibility and blood compatibility in vitro, and almost no toxicity towards Zebrafish embryos development in vivo. Furthermore, dual-loading of hydrophilic cisplatin and hydrophobic paclitaxel was achieved in the nanotubes with high loading content and loading efficiency. The release of both drugs was slower from dual-loaded nanotubes than from single-loaded ones, but the total amount of released drugs in higher for dual-loaded nanotubes than from single-loaded ones. Cellular uptake and inhibition tests showed that the nanotubes were successfully taken up by tumor cells and effectively inhibited cell growth. It is thus concluded that PEG-PLA-PEG nanotubes with outstanding biocompatibility could be promising for co-delivery of hydrophilic and hydrophobic agents in combination cancer therapy.
Collapse
|
23
|
Simultaneous controlled release of 5-FU, DOX and PTX from chitosan/PLA/5-FU/g-C3N4-DOX/g-C3N4-PTX triaxial nanofibers for breast cancer treatment in vitro. Colloids Surf B Biointerfaces 2019; 179:495-504. [DOI: 10.1016/j.colsurfb.2019.04.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 03/15/2019] [Accepted: 04/11/2019] [Indexed: 11/19/2022]
|
24
|
Wang HH, Fu ZG, Li W, Li YX, Zhao LS, Wen L, Zhang JJ, Wen N. The synthesis and application of nano doxorubicin- indocyanine green matrix metalloproteinase-responsive hydrogel in chemophototherapy for head and neck squamous cell carcinoma. Int J Nanomedicine 2019; 14:623-638. [PMID: 30697046 PMCID: PMC6339648 DOI: 10.2147/ijn.s191069] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Head and neck squamous cell carcinoma (HNSCC) is one of the most common malignancies, with high rates of mortality and morbidity worldwide. Owing to the special anatomical location of this tumor, an effective, minimally invasive treatment with low systemic toxicity is highly desirable. Hydrogels have shown great potential for tumor-targeting therapy, with excellent performance. However, there have been few reports on co-loading photosensitizers and chemotherapeutic drugs into hydrogels. In this study, we synthesized a nano doxorubicin-indocyanine green matrix metalloproteinase (MMP)-responsive hydrogel (denoted as NDIMH), combining chemotherapy and phototherapy, to achieve superior antitumor efficacy. Methods First, NDIMH was synthesized and characterized by scanning electron microscopy and drug-release assays. Second, the photosensitivity properties and antitumor efficiency of this drug delivery system were studied in vivo and in vitro. Last, the imaging and biodistribution of NDIMH were monitored using the Maestro EX in vivo imaging system. Results The nanodrugs loaded into the smart hydrogel exhibited uniform size distribution, excellent size stability, and a sustained release in the presence of MMP-2. NDIMH showed ideal photosensitivity characteristics under light. NDIMH with 808 nm near-infrared (NIR) irradiation effectively inhibited the viability, invasion, and metastasis of SCC-15 in vitro. After intratumoral injection of NDIMH with 808 nm NIR illumination, the hydrogels exhibited favorable synergistic antitumor efficacy and acceptable biosafety. Additionally, fluorescence imaging showed that NDIMH could significantly improve the retention of nanodrugs at the tumor site. Conclusion The intratumoral injection of NDIMH with 808 nm NIR irradiation could be a promising chemophototherapy alternative for HNSCC.
Collapse
Affiliation(s)
- Huan-Huan Wang
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China,
| | - Zhi-Guang Fu
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China,
| | - Wei Li
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China,
| | - Yun-Xia Li
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China,
| | - Li-Sheng Zhao
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China,
| | - Li Wen
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China,
| | - Jian-Jun Zhang
- College of Chemical Engineering, Beijing University of Chemical Technology, Beijing 100029, China,
| | - Ning Wen
- Department of Stomatology, Chinese PLA General Hospital, Beijing 100853, China,
| |
Collapse
|
25
|
Wu M, Liu J, Hu C, Li D, Yang J, Wu Z, Yang L, Chen Y, Fu S, Wu J. Olaparib nanoparticles potentiated radiosensitization effects on lung cancer. Int J Nanomedicine 2018; 13:8461-8472. [PMID: 30587971 PMCID: PMC6294076 DOI: 10.2147/ijn.s181546] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Poly (ADP-ribose) polymerase (PARP) is a key enzyme in the repair process of DNA strand breaks (DSBs). Olaparib (Ola) is a PARP inhibitor that is involved in arresting PARP release from radiotherapy (RT)-induced damaged DNA to potentiate the effect of RT. Although the underlying mechanisms for the radiosensitization effects of Ola are well understood in vitro, the radiosensitization effects in vivo are still unclear. Moreover, poor water solubility and severe toxicity are two major impediments for the clinical success of Ola. MATERIALS AND METHODS Here, we developed olaparib nanoparticles (Ola-NPs) and investigated their radiosensitization mechanisms and toxicity using human non-small-cell lung cancer xenograft models in mice. RESULTS The prepared Ola-NPs showed a mean size of 31.96±1.54 nm and a lower polydispersity index of about 0.126±0.014. In addition, the sensitization enhancement ratio of Ola-NPs (3.81) was much higher than that of free Ola (1.66). The combination of Ola-NPs and RT (Ola-NPs + RT) significantly inhibited tumor growth and prolonged survival in mice. The mechanism of enhanced antitumor efficacy might be related to the inhibition of DSB repair and the promotion of cell apoptosis in vivo. No additional toxicity caused by Ola-NPs was observed. CONCLUSION This study demonstrated the principle of using Ola-NPs as a potent radiosensitizer to improve the therapeutic effect of RT relative to free Ola (P<0.05 in all cases).
Collapse
Affiliation(s)
- Min Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China, ;
| | - Jing Liu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China, ;
| | - ChuanFei Hu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China, ;
| | - Dong Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China, ;
| | - Juan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China, ;
| | - ZhouXue Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China, ;
| | - LingLin Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China, ;
| | - Yue Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - ShaoZhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China, ;
| | - JingBo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China, ;
| |
Collapse
|
26
|
Yu S, He C, Chen X. Injectable Hydrogels as Unique Platforms for Local Chemotherapeutics-Based Combination Antitumor Therapy. Macromol Biosci 2018; 18:e1800240. [PMID: 30303620 DOI: 10.1002/mabi.201800240] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/15/2018] [Indexed: 01/06/2025]
Abstract
Different strategies of chemotherapeutics-based combination cancer therapy have presented enhanced antitumor efficiency and are widely used in clinical cancer treatments. However, several drawbacks of the systems for systemic administration, including low drug accumulation at tumor sites and significant systemic side effects limit their efficacy and application in the clinic. Local drug co-delivery systems based on injectable hydrogels may have considerable advantages, such as a facile drug-delivery procedure, targeted delivery of antitumor agents to tumor sites in a sustained manner, and markedly reduced systemic toxicities. Thus, in recent years, these systems have received increasing attention and consequently various injectable hydrogels have been tested as platforms for local chemotherapeutics-based combination antitumor therapy. In this review, the focus is on recent advances in injectable hydrogel-based drug co-delivery systems for local combination antitumor therapy, including multiple chemotherapeutics combination therapy, chemo-immunotherapy, chemo-radiotherapy, and hyperthermia-chemotherapy. Moreover, the rationale and preparation of local co-delivery systems are summarized and discussed.
Collapse
Affiliation(s)
- Shuangjiang Yu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Chaoliang He
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| |
Collapse
|
27
|
Chang CE, Hsieh CM, Chen LC, Su CY, Liu DZ, Jhan HJ, Ho HO, Sheu MT. Novel application of pluronic lecithin organogels (PLOs) for local delivery of synergistic combination of docetaxel and cisplatin to improve therapeutic efficacy against ovarian cancer. Drug Deliv 2018; 25:632-643. [PMID: 29463123 PMCID: PMC6058476 DOI: 10.1080/10717544.2018.1440444] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The synergistic combination of docetaxel (DTX) and cisplatin (CIS) by local drug delivery with a pluronic lecithin organogel (PLO) to facilitate high drug concentrations at tumor sites and less nonspecific distribution to normal organs is thought to be beneficial in chemotherapy. In this study, using Capryol-90 (C90) with the addition of lecithin as the oil phase was developed to carry DTX, which was then incorporated into a PLO-containing CIS to formulate a dual-drug injectable PLO for local delivery. An optimal PLO composite, P13L0.15O1.5, composed of PF127:lecithin:C90 at a 13:0.15:1.5 weight ratio was obtained. The sol-gel transition temperature of P13L0.15O1.5 was found to be 33 °C. Tumor inhibition studies illustrated that DTX/CIS-loaded P13L0.15O1.5 could efficiently suppress tumor growth by both intratumoral and peritumoral injections in SKOV-3 xenograft mouse model. Pharmacokinetic studies showed that subcutaneous administration of P13L0.15O1.5 was able to sustain the release of DTX and CIS leading to their slow absorption into the systemic circulation resulting in lower area under the plasma concentration curve at 0-72 h (AUC0-72) and maximum concentration (Cmax) values but longer half-life (T1/2) and mean residence time (MRT) values. An in vivo biodistribution study showed lower DTX and CIS concentrations in organs compared to other treatment groups after IT administration of the dual drug-loaded P13L0.15O1.5. It was concluded that the local co-delivery of DTX and CIS by PLOs may be a promising and effective platform for local anticancer drug delivery with minimal systemic toxicities.
Collapse
Affiliation(s)
- Chia-En Chang
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Chien-Ming Hsieh
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Ling-Chun Chen
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC.,b Department of Biotechnology and Pharmaceutical Technology , Yuanpei University of Medical Technology , Hsinchu , Taiwan, ROC
| | - Chia-Yu Su
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Der-Zen Liu
- c Graduate Institute of Biomedical Materials and Engineering , Taipei Medical University , Taipei , Taiwan, ROC
| | - Hua-Jing Jhan
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Hsiu-O Ho
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC
| | - Ming-Thau Sheu
- a School of Pharmacy, College of Pharmacy , Taipei Medical University , Taipei , Taiwan, ROC.,d Clinical Research Center and Traditional Herbal Medicine Research Center , Taipei Medical University Hospital , Taipei , Taiwan, ROC
| |
Collapse
|
28
|
Pugliese E, Coentro JQ, Zeugolis DI. Advancements and Challenges in Multidomain Multicargo Delivery Vehicles. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2018; 30:e1704324. [PMID: 29446161 DOI: 10.1002/adma.201704324] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 11/05/2017] [Indexed: 06/08/2023]
Abstract
Reparative and regenerative processes are well-orchestrated temporal and spatial events that are governed by multiple cells, molecules, signaling pathways, and interactions thereof. Yet again, currently available implantable devices fail largely to recapitulate nature's complexity and sophistication in this regard. Herein, success stories and challenges in the field of layer-by-layer, composite, self-assembly, and core-shell technologies are discussed for the development of multidomain/multicargo delivery vehicles.
Collapse
Affiliation(s)
- Eugenia Pugliese
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| | - João Q Coentro
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
- Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Ireland
| |
Collapse
|
29
|
Pardo J, Peng Z, Leblanc RM. Cancer Targeting and Drug Delivery Using Carbon-Based Quantum Dots and Nanotubes. Molecules 2018; 23:E378. [PMID: 29439409 PMCID: PMC6017112 DOI: 10.3390/molecules23020378] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/07/2018] [Accepted: 02/09/2018] [Indexed: 12/14/2022] Open
Abstract
Currently cancer treatment is in large part non-specific with respect to treatment. Medication is often harsh on patients, whereby they suffer several undesired side effects as a result. Carbon-based nanoparticles have attracted attention in recent years due to their ability to act as a platform for the attachment of several drugs and/or ligands. Relatively simple models are often used in cancer research, wherein carbon nanoparticles are conjugated to a ligand that is specific to an overexpressed receptor for imaging and drug delivery in cancer treatment. These carbon nanoparticles confer unique properties to the imaging or delivery vehicle due to their nontoxic nature and their high fluorescence qualities. Chief among the ongoing research within carbon-based nanoparticles emerge carbon dots (C-dots) and carbon nanotubes (CNTs). In this review, the aforementioned carbon nanoparticles will be discussed in their use within doxorubicin and gemcitabine based drug delivery vehicles, as well as the ligand-mediated receptor specific targeted therapy. Further directions of research in current field are also discussed.
Collapse
Affiliation(s)
- Joel Pardo
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL 33146, USA.
| | - Zhili Peng
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL 33146, USA.
- College of Pharmacy and Chemistry, Dali University, Dali 671000, Yunnan, China.
| | - Roger M Leblanc
- Department of Chemistry, University of Miami, 1301 Memorial Drive, Coral Gables, FL 33146, USA.
| |
Collapse
|
30
|
Ferreira N, Ferreira L, Cardoso V, Boni F, Souza A, Gremião M. Recent advances in smart hydrogels for biomedical applications: From self-assembly to functional approaches. Eur Polym J 2018. [DOI: 10.1016/j.eurpolymj.2017.12.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
31
|
Wang Q, Zhang H, Huang J, Xia N, Li T, Xia Q. Self-double-emulsifying drug delivery system incorporated in natural hydrogels: a new way for topical application of vitamin C. J Microencapsul 2018; 35:90-101. [DOI: 10.1080/02652048.2018.1425752] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Qiang Wang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing, China
- Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu, China
| | - Hong Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu, China
| | - Juan Huang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu, China
| | - Nan Xia
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu, China
| | - Tong Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu, China
| | - Qiang Xia
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- National Demonstration Center for Experimental Biomedical Engineering Education, Southeast University, Nanjing, China
- Collaborative Innovation Center of Suzhou Nano Science and Technology, Suzhou, Jiangsu, China
| |
Collapse
|
32
|
Efficient inhibition of cervical cancer by dual drugs loaded in biodegradable thermosensitive hydrogel composites. Oncotarget 2017; 9:282-292. [PMID: 29416613 PMCID: PMC5787464 DOI: 10.18632/oncotarget.22965] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/09/2017] [Indexed: 01/02/2023] Open
Abstract
Background and Purpose We aimed to explore the anti-tumor effect and mechanism of the combination of cisplatin (DDP)-containing thermosensitive hydrogel (PEG-PCL-PEG, or PECE) and paclitaxel (PTX)-loaded MPEG-PCL polymeric micelles called PDMP on human cervical carcinoma (HeLa) cell. In our previous studies, we found that PDMP in situ treatment of lung cancer will be liable to have potential in Lung cancer patients. Results Compared with other treatments, PDMP was most effective in prolonging survival time (P < 0.05), inhibiting tumor growth (P < 0.05), decreasing expression of CD133 (P < 0.05), CD31 (P < 0.05), and aldehyde dehydrogenase 1 (ALDH1) (P > 0.05), inducing G1 phase arrest (P < 0.05), increasing the apoptosis rate (P < 0.05), and in expressing ATM and γ-H2AX (P < 0.05). Conclusions PDMP is regarded as a promising anti-tumor reactant, when it comes to the treatment of cervical carcinoma. Methods we used a xenograft cervical cancer model to verify the anti-tumor activity of PDMP and to explore its mechanism of action. Mice were intratumorally administered with NS, PECE, PTX+DDP or PDMP. After two days of treatment, three mice per group were sacrificed and tumor tissue was harvested. Levels of histone H2AX phosphorylation (γ-H2AX) were determined by immunohistochemistry and ataxia telangiectasia mutated(ATM) protein levels were measured by western blot analysis. In addition, it would sacrifice each of group of three mice through 10 days’ treatment, what’s more, it would harvest tumor by virtue of flow cytometry and immunohistochemical analysis. It would like to use there maining mice to analyze tumor growth and survival. The remaining mice were analyzed for tumor growth and survival.
Collapse
|
33
|
Shen W, Chen X, Luan J, Wang D, Yu L, Ding J. Sustained Codelivery of Cisplatin and Paclitaxel via an Injectable Prodrug Hydrogel for Ovarian Cancer Treatment. ACS APPLIED MATERIALS & INTERFACES 2017; 9:40031-40046. [PMID: 29131563 DOI: 10.1021/acsami.7b11998] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The sustained release of both the hydrophilic drug and hydrophobic drug from one delivery system remains challenging in pharmaceutics and biomaterials science. The combination of hydrophilic cisplatin and hydrophobic paclitaxel (PTX) exhibits a clinical survival advantage compared with the individual drug therapy against various tumors such as ovarian cancer. In this study, a localized, long-term codelivery system of cisplatin and PTX was developed using an injectable and thermosensitive polymer-platinum(IV) conjugate hydrogel as the carrier. The thermosensitive Bi(mPEG-PLGA)-Pt(IV) (PtGel) conjugate was synthesized via covalently linking two mPEG-PLGA copolymers onto a Pt(IV) prodrug, and its concentrated aqueous solution exhibited a reversible sol-gel transition upon heating. Meanwhile, the core-corona micelles formed by the amphiphilic conjugates in water could serve as a reservoir for the solubilization of PTX, and thus an injectable binary drug-loaded hydrogel formulation was obtained. We also found that the introduction of PTX into the conjugate hydrogel decreased its sol-gel transition temperature and improved its gel strength. In vitro release experiments showed that both of the loaded drugs were released in a sustained manner for as long as 2.5 months, which was the longest combination delivery of these two drugs ever reported. In vitro cellular assays revealed that the dual-drug system exhibited a synergistic anticancer effect against ovarian cancer cells. Finally, using the SKOV-3 ovarian cancer xenograft mouse model, we demonstrated that a single injection of the PTX-loaded conjugate hydrogel system resulted in enhanced anticancer efficacy and significantly reduced the side effects, when compared with the multiple injections of the free drug combination.
Collapse
Affiliation(s)
- Wenjia Shen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai 200433, China
| | - Xiaobin Chen
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai 200433, China
| | - Jiabin Luan
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai 200433, China
| | - Danni Wang
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai 200433, China
| | - Lin Yu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai 200433, China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, Fudan University , Shanghai 200433, China
| |
Collapse
|
34
|
Taurin S, Almomen AA, Pollak T, Kim SJ, Maxwell J, Peterson CM, Owen SC, Janát-Amsbury MM. Thermosensitive hydrogels a versatile concept adapted to vaginal drug delivery. J Drug Target 2017; 26:533-550. [PMID: 29096548 DOI: 10.1080/1061186x.2017.1400551] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Vaginal drug delivery represents an attractive strategy for local and systemic delivery of drugs otherwise poorly absorbed after oral administration. The rather dense vascular network, mucus permeability and the physiological phenomenon of the uterine first-pass effect can all be exploited for therapeutic benefit. However, several physiological factors such as an acidic pH, constant secretion, and turnover of mucus as well as varying thickness of the vaginal epithelium can impact sustained drug delivery. In recent years, polymers have been designed to tackle challenges mentioned above. In particular, thermosensitive hydrogels hold great promise due to their stability, biocompatibility, adhesion properties and adjustable drug release kinetics. Here, we discuss the physiological and anatomical uniqueness of the vaginal environment and how it impacts the safe and efficient vaginal delivery and also reviewed several thermosensitive hydrogels deemed suitable for vaginal drug delivery by addressing specific characteristics, which are essential to engage the vaginal environment successfully.
Collapse
Affiliation(s)
- Sebastien Taurin
- a Department of Obstetrics and Gynecology, Division of Gynecologic Oncology , University of Utah Health Sciences , Salt Lake City , UT , USA
| | - Aliyah A Almomen
- a Department of Obstetrics and Gynecology, Division of Gynecologic Oncology , University of Utah Health Sciences , Salt Lake City , UT , USA.,b Department of Pharmaceutics and Pharmaceutical Chemistry , University of Utah , Salt Lake City , UT , USA
| | - Tatianna Pollak
- a Department of Obstetrics and Gynecology, Division of Gynecologic Oncology , University of Utah Health Sciences , Salt Lake City , UT , USA
| | - Sun Jin Kim
- b Department of Pharmaceutics and Pharmaceutical Chemistry , University of Utah , Salt Lake City , UT , USA
| | - John Maxwell
- a Department of Obstetrics and Gynecology, Division of Gynecologic Oncology , University of Utah Health Sciences , Salt Lake City , UT , USA
| | - C Matthew Peterson
- c Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology , University of Utah Health Science Center , Salt Lake City , UT , USA
| | - Shawn C Owen
- b Department of Pharmaceutics and Pharmaceutical Chemistry , University of Utah , Salt Lake City , UT , USA.,d Department of Bioengineering , University of Utah , Salt Lake City , UT , USA
| | - Margit M Janát-Amsbury
- a Department of Obstetrics and Gynecology, Division of Gynecologic Oncology , University of Utah Health Sciences , Salt Lake City , UT , USA.,b Department of Pharmaceutics and Pharmaceutical Chemistry , University of Utah , Salt Lake City , UT , USA.,c Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology , University of Utah Health Science Center , Salt Lake City , UT , USA.,d Department of Bioengineering , University of Utah , Salt Lake City , UT , USA
| |
Collapse
|
35
|
Xu S, Tang YY, Yu YX, Yun Q, Yang JP, Zhang H, Peng Q, Sun X, Yang LL, Fu S, Wu JB. Novel composite drug delivery system as a novel radio sensitizer for the local treatment of cervical carcinoma. Drug Deliv 2017; 24:1139-1147. [PMID: 28797171 PMCID: PMC8241059 DOI: 10.1080/10717544.2017.1362676] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 07/28/2017] [Accepted: 07/29/2017] [Indexed: 01/07/2023] Open
Abstract
In this study, we investigated in vivo radiosensitizing effects of a gel-based dual drug delivery system (DDS) (PECE/DDP + mPEG-PCL/PTX, or PDMP) in a cervical cancer model, and determined its possible mechanisms of action. A xenograft cervical cancer model was used to investigate the radio sensitization effect of PDMP. Mice underwent paclitaxel (PTX) + cisplatin (DDP), PECE, or PDMP treatment followed by single radiation doses ranging from 0 Gy to 20 Gy. Radio sensitization was analyzed by tumor regrowth delay (TGD). The sensitization enhancement ratio (SER) was calculated by the doses needed to yield TGD when using radiation treatment alone and when using radiation plus drug treatment. The impact of irradiation and drugs on TGD was determined, and an optimum radiation dose was chosen for further evaluation of radio sensitizing effects. The data showed that PDMP yielded the highest radio sensitization (SER was 1.3) and a radiation dose of 12 Gy was chosen for further investigation. PDMP + radiotherapy treatment was most effective in inhibiting tumor growth, prolonging survival time, decreasing expression of CD31, CD133, and aldehyde dehydrogenase 1 (ALDH1), inducing G2/M phase arrest, apoptosis, and expression of Ataxia telangiectasia mutated (ATM) and histone H2AX phosphorylation (γ-H2AX). Thus, our data indicated that PDMP is a promising anti-tumor and radio sensitization reagent for the treatment of cervical carcinoma.
Collapse
Affiliation(s)
- Shan Xu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
- Department of Oncology, MianYang Central Hospital, Mianyang, Sichuan Province, China
| | - Yu Ying Tang
- MianYang Central Hospital, Mianyang, Sichuan Province, China
| | - Yan Xin Yu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Qin Yun
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jing Pin Yang
- Department of Oncology, The First People's Hospital of Guangyuan, Guangyuan, Sichuan Province, China
| | - Heng Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Qiuxia Peng
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Xiaoyang Sun
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Ling Lin Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - ShaoZhi Fu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| | - Jing Bo Wu
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
36
|
Rhodamine-loaded surface modified mesoporous silica particles embedded into a thermoresponsive composite hydrogel for prolonged release. Eur Polym J 2017. [DOI: 10.1016/j.eurpolymj.2017.08.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
37
|
Pitorre M, Gondé H, Haury C, Messous M, Poilane J, Boudaud D, Kanber E, Rossemond Ndombina GA, Benoit JP, Bastiat G. Recent advances in nanocarrier-loaded gels: Which drug delivery technologies against which diseases? J Control Release 2017; 266:140-155. [PMID: 28951319 DOI: 10.1016/j.jconrel.2017.09.031] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/21/2017] [Accepted: 09/22/2017] [Indexed: 01/02/2023]
Abstract
The combination of pharmaceutical technologies can be a wise choice for developing innovative therapeutic strategies. The association of nanocarriers and gels provides new therapeutic possibilities due to the combined properties of the two technologies. Gels support the nanocarriers, localize their administration to the target tissue, and sustain their release. In addition to the properties afforded by the gel, nanocarriers can provide additional drug sustained release or different pharmacokinetic and biodistribution profiles than those from nanocarriers administered by the conventional route to improve the drug therapeutic index. This review focuses on recent (over the last ten years) in vivo data showing the advances and advantages of using nanocarrier-loaded gels. Liposomes, micelles, liquid and solid lipid nanocapsules, polymeric nanoparticles, dendrimers, and fullerenes are all nanotechnologies which have been recently assessed for medical applications, such as cancer therapy, the treatment of cutaneous and infectious diseases, anesthesia, the administration of antidepressants, and the treatment of unexpected diseases, such as alopecia.
Collapse
Affiliation(s)
- Marion Pitorre
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France; Master 2 Nanomédecines et R&D Pharmaceutique, Pharmacy Department, UFR Santé, Université Bretagne Loire, Angers, France
| | - Henri Gondé
- Master 2 Nanomédecines et R&D Pharmaceutique, Pharmacy Department, UFR Santé, Université Bretagne Loire, Angers, France
| | - Clotilde Haury
- Master 2 Nanomédecines et R&D Pharmaceutique, Pharmacy Department, UFR Santé, Université Bretagne Loire, Angers, France
| | - Marwa Messous
- Master 2 Nanomédecines et R&D Pharmaceutique, Pharmacy Department, UFR Santé, Université Bretagne Loire, Angers, France
| | - Jérémie Poilane
- Master 2 Nanomédecines et R&D Pharmaceutique, Pharmacy Department, UFR Santé, Université Bretagne Loire, Angers, France
| | - David Boudaud
- Master 2 Nanomédecines et R&D Pharmaceutique, Pharmacy Department, UFR Santé, Université Bretagne Loire, Angers, France
| | - Erdem Kanber
- Master 2 Nanomédecines et R&D Pharmaceutique, Pharmacy Department, UFR Santé, Université Bretagne Loire, Angers, France
| | | | - Jean-Pierre Benoit
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France; Master 2 Nanomédecines et R&D Pharmaceutique, Pharmacy Department, UFR Santé, Université Bretagne Loire, Angers, France
| | - Guillaume Bastiat
- MINT, UNIV Angers, INSERM 1066, CNRS 6021, Université Bretagne Loire, Angers, France; Master 2 Nanomédecines et R&D Pharmaceutique, Pharmacy Department, UFR Santé, Université Bretagne Loire, Angers, France.
| |
Collapse
|
38
|
Manaia EB, Abuçafy MP, Chiari-Andréo BG, Silva BL, Oshiro Junior JA, Chiavacci LA. Physicochemical characterization of drug nanocarriers. Int J Nanomedicine 2017; 12:4991-5011. [PMID: 28761340 PMCID: PMC5516877 DOI: 10.2147/ijn.s133832] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pharmaceutical design has enabled important advances in the prevention, treatment, and diagnosis of diseases. The use of nanotechnology to optimize the delivery of drugs and diagnostic molecules is increasingly receiving attention due to the enhanced efficiency provided by these systems. Understanding the structures of nanocarriers is crucial in elucidating their physical and chemical properties, which greatly influence their behavior in the body at both the molecular and systemic levels. This review was conducted to describe the principles and characteristics of techniques commonly used to elucidate the structures of nanocarriers, with consideration of their size, morphology, surface charge, porosity, crystalline arrangement, and phase. These techniques include X-ray diffraction, small-angle X-ray scattering, dynamic light scattering, zeta potential, polarized light microscopy, transmission electron microscopy, scanning electron microcopy, and porosimetry. Moreover, we describe some of the commonly used nanocarriers (liquid crystals, metal-organic frameworks, silica nanospheres, liposomes, solid lipid nanoparticles, and micelles) and the main aspects of their structures.
Collapse
Affiliation(s)
- Eloísa Berbel Manaia
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Marina Paiva Abuçafy
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Bruna Galdorfini Chiari-Andréo
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
- Department of Biological and Health Sciences, Centro Universitário de Araraquara, UNIARA, Araraquara, SP, Brazil
| | - Bruna Lallo Silva
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - João Augusto Oshiro Junior
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| | - Leila Aparecida Chiavacci
- Department of Drugs and Medicines, School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, SP, Brazil
| |
Collapse
|
39
|
Bini RA, Silva MF, Varanda LC, da Silva MA, Dreiss CA. Soft nanocomposites of gelatin and poly(3-hydroxybutyrate) nanoparticles for dual drug release. Colloids Surf B Biointerfaces 2017; 157:191-198. [PMID: 28595135 DOI: 10.1016/j.colsurfb.2017.05.051] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 04/28/2017] [Accepted: 05/20/2017] [Indexed: 02/07/2023]
Abstract
We developed a nanocomposite gel composed of gelatin and poly(3-hydroxybutyrate) polymeric nanoparticles (PNP) to be used as an injectable gel for the contemporaneous, dual sustained release of bioactive molecules. The hydrogel matrix was formed by a very simple process, using either the physical gelation of gelatin or the natural enzyme transglutaminase to covalently cross-link the gelatin chains in the presence of embedded PNP. Oscillatory rheological measurements showed that the addition of the PNP induced an increase in the storage modulus compared to pure gelatin gels, for both physical and chemical gels. Micrographs from scanning electron microscopy revealed that the presence of PNP disrupted the native structure of the gelatin chains in the hydrogel matrix. Dual drug encapsulation was achieved with curcumin (CM) in the PNP and naproxen sodium(NS) in the gelatin matrix. In vitro release studies showed that the hydrogel matrix acts both as a physical and chemical barrier, delaying the diffusion of the drugs. An initial burst release was observed in the first hours of the measurement, and around 90% was released on the third day for naproxen sodium. In free PNP, 82% of curcumin was relased after four days, while when PNP were embedded in the gelatin matrix only 40% was released over the same time period. Overall, these simple, sustainable soft nanocomposites show potential as an injectable co-sustained drug release system.
Collapse
Affiliation(s)
- Rafael A Bini
- Federal University of Technology - Paraná - UTFPR, Biotechnology and Bioprocess Engineering, Campus Toledo Rua Cristo Rei, 19. 85902-490, Toledo, Brazil.
| | - Mônica F Silva
- University of São Paulo, Colloidal Materials Group, Chemistry Institute of São Carlos, São Carlos, 13566-590, Brazil
| | - Laudemir C Varanda
- University of São Paulo, Colloidal Materials Group, Chemistry Institute of São Carlos, São Carlos, 13566-590, Brazil
| | - Marcelo A da Silva
- King's College London, Institute of Pharmaceutical Science, 150 Stamford, Street, London SE1 9NH, UK
| | - Cécile A Dreiss
- King's College London, Institute of Pharmaceutical Science, 150 Stamford, Street, London SE1 9NH, UK
| |
Collapse
|
40
|
Feng X, Wang G, Neumann K, Yao W, Ding L, Li S, Sheng Y, Jiang Y, Bradley M, Zhang R. Synthesis and characterization of biodegradable poly(ether-ester) urethane acrylates for controlled drug release. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 74:270-278. [DOI: 10.1016/j.msec.2016.12.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Revised: 10/30/2016] [Accepted: 12/04/2016] [Indexed: 10/20/2022]
|
41
|
Yu Y, Xu S, You H, Zhang Y, Yang B, Sun X, Yang L, Chen Y, Fu S, Wu J. In vivo synergistic anti-tumor effect of paclitaxel nanoparticles combined with radiotherapy on human cervical carcinoma. Drug Deliv 2017; 24:75-82. [PMID: 28155566 PMCID: PMC8241140 DOI: 10.1080/10717544.2016.1230902] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
In this study, our purpose was to explore the synergistic anti-tumor effect and mechanism of paclitaxel nanoparticles (PTX-NPs) combined with radiotherapy (RT) on human cervical carcinoma (HeLa). PTX-NPs were prepared by a solid dispersion method using methoxy poly(ethylene glycol)–poly(ɛ-caprolactone) (MPEG–PCL), which combined with RT exerted a potent and high efficient effect against cervical cancer. In vivo antitumor activity of PTX-NPs combined with RT, was estimated using nude mice carrying Hela cell xenograft tumor. The results were evaluated using microfluorine-18-deoxyglucose PET/computed tomography (18F-FDG PET/CT) and immunohistochemistry. The results showed that PTX-NPs possessed a more efficient effect than PTX when combined with RT (p < 0.05). PTX-NPs in combination with RT might inhibit cell proliferation through its action on Ki-67, and decreased micro-vessel density (MVD) associated with CD31 and vascular endothelial growth factor (VEGF). These results suggested that PTX-NPs possessed a synergistic anti-tumor effect against cervical cancer when combined with RT.
Collapse
Affiliation(s)
| | - Shan Xu
- a Department of Oncology and
| | | | | | - Bo Yang
- a Department of Oncology and
| | | | | | - Yue Chen
- b Department of Nuclear Medicine , the Affiliated Hospital of Southwest Medical University , Luzhou , China
| | | | | |
Collapse
|
42
|
Ma Y, Ge Y, Li L. Advancement of multifunctional hybrid nanogel systems: Construction and application in drug co-delivery and imaging technique. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 71:1281-1292. [DOI: 10.1016/j.msec.2016.11.031] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 10/21/2016] [Accepted: 11/08/2016] [Indexed: 12/31/2022]
|
43
|
Patel M, Kaneko T, Matsumura K. Switchable release nano-reservoirs for co-delivery of drugs via a facile micelle–hydrogel composite. J Mater Chem B 2017; 5:3488-3497. [DOI: 10.1039/c7tb00701a] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
44
|
Huang LM, Li LD, Shang L, Zhou QH, Lin J. Preparation of pH-sensitive micelles from miktoarm star block copolymers by ATRP and their application as drug nanocarriers. REACT FUNCT POLYM 2016. [DOI: 10.1016/j.reactfunctpolym.2016.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
45
|
Sharifzadeh G, Wahit MU, Soheilmoghaddam M, Whye WT, Pasbakhsh P. Kappa-carrageenan/halloysite nanocomposite hydrogels as potential drug delivery systems. J Taiwan Inst Chem Eng 2016. [DOI: 10.1016/j.jtice.2016.07.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
46
|
Norouzi M, Nazari B, Miller DW. Injectable hydrogel-based drug delivery systems for local cancer therapy. Drug Discov Today 2016; 21:1835-1849. [PMID: 27423369 DOI: 10.1016/j.drudis.2016.07.006] [Citation(s) in RCA: 336] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 07/05/2016] [Accepted: 07/07/2016] [Indexed: 01/17/2023]
Abstract
Common chemotherapy is often associated with adverse effects in normal cells and tissues. As an alternative approach, localized chemotherapy can diminish the toxicity of systemic chemotherapy while providing a sustained release of the chemotherapeutics at the target tumor site. Therefore, injectable biodegradable hydrogels as drug delivery systems for chemotherapeutics have become a matter of importance. Here, we review the application of a variety of injectable hydrogel-based drug delivery systems, including thermosensitive, pH-sensitive, photosensitive, dual-sensitive, as well as active targeting hydrogels, for the treatment of different types of cancer. Generally, injectable hydrogel-based drug delivery systems are found to be more efficacious than the conventional systemic chemotherapy in terms of cancer treatment.
Collapse
Affiliation(s)
- Mohammad Norouzi
- Graduate Program of Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada; Department of Nanotechnology and Tissue Engineering, Stem Cell Technology Research Center, Tehran, Iran.
| | - Bahareh Nazari
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Donald W Miller
- Graduate Program of Biomedical Engineering, University of Manitoba, Winnipeg, MB, Canada; Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
47
|
Silveira CP, Apolinário LM, Fávaro WJ, Paula AJ, Durán N. Doxorubicin-Functionalized Silica Nanoparticles Incorporated into a Thermoreversible Hydrogel and Intraperitoneally Administered Result in High Prostate Antitumor Activity and Reduced Cardiotoxicity of Doxorubicin. ACS Biomater Sci Eng 2016; 2:1190-1199. [PMID: 33465877 DOI: 10.1021/acsbiomaterials.6b00241] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Described here is an anticancer material based on colloidal mesoporous silica nanoparticles (MSNs) functionalized with doxorubicin (DOX), and incorporated into Pluronic F127 hydrogels for prolonged release, with a potential therapeutic application for prostate cancer treatment. The MSNs have spherical morphology, size of about 60 nm, surface area of 970 cm2 g-1 and average pore width of 2.0 nm. A high colloidal stability for the MSNs in the physiological medium used for in vivo administration (NaCl 0.9% w/v) could be attained in the presence of PF127 (from 5 to 18 wt %), where depletion repulsion forces prevent MSN agglomeration. By conjugating DOX, MSN and PF127 (18 wt %) in NaCl 0.9%, the hybrid system has a gelation temperature of 21 °C, which allowed its in vivo administration in the liquid form and further in situ gelation, generating a drug depot system inside the animals after peritoneal injection. The systems were tested in rats with chemically induced prostate cancer and, after this treatment, histopathological analyses confirmed (i) a reduction in the frequency of aggressive tumors; (ii) that the antitumor effect was dependent on MSN concentration; and most importantly (iii) the reduction of DOX intrinsic cardiotoxicity, indicating that the MSNs play a cardioprotective effect.
Collapse
Affiliation(s)
| | | | | | - Amauri J Paula
- Solid-Biological Interface Group (SolBIN), Department of Physics, Universidade Federal do Ceará (UFC), Campus do Pici, 60440-900 Fortaleza, Ceara, Brazil
| | - Nelson Durán
- Farmabrasilis R&D Division, Campinas São Paulo, Brazil
| |
Collapse
|
48
|
Hofferberth SC, Grinstaff MW, Colson YL. Nanotechnology applications in thoracic surgery. Eur J Cardiothorac Surg 2016; 50:6-16. [PMID: 26843431 DOI: 10.1093/ejcts/ezw002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/16/2015] [Indexed: 01/16/2023] Open
Abstract
Nanotechnology is an emerging, rapidly evolving field with the potential to significantly impact care across the full spectrum of cancer therapy. Of note, several recent nanotechnological advances show particular promise to improve outcomes for thoracic surgical patients. A variety of nanotechnologies are described that offer possible solutions to existing challenges encountered in the detection, diagnosis and treatment of lung cancer. Nanotechnology-based imaging platforms have the ability to improve the surgical care of patients with thoracic malignancies through technological advances in intraoperative tumour localization, lymph node mapping and accuracy of tumour resection. Moreover, nanotechnology is poised to revolutionize adjuvant lung cancer therapy. Common chemotherapeutic drugs, such as paclitaxel, docetaxel and doxorubicin, are being formulated using various nanotechnologies to improve drug delivery, whereas nanoparticle (NP)-based imaging technologies can monitor the tumour microenvironment and facilitate molecularly targeted lung cancer therapy. Although early nanotechnology-based delivery systems show promise, the next frontier in lung cancer therapy is the development of 'theranostic' multifunctional NPs capable of integrating diagnosis, drug monitoring, tumour targeting and controlled drug release into various unifying platforms. This article provides an overview of key existing and emerging nanotechnology platforms that may find clinical application in thoracic surgery in the near future.
Collapse
Affiliation(s)
- Sophie C Hofferberth
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Mark W Grinstaff
- Departments of Biomedical Engineering, Chemistry, and Medicine, Boston University, Boston, MA, USA
| | - Yolonda L Colson
- Division of Thoracic Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
49
|
Memic A, Alhadrami HA, Hussain MA, Aldhahri M, Al Nowaiser F, Al-Hazmi F, Oklu R, Khademhosseini A. Hydrogels 2.0: improved properties with nanomaterial composites for biomedical applications. ACTA ACUST UNITED AC 2015; 11:014104. [PMID: 26694229 DOI: 10.1088/1748-6041/11/1/014104] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The incorporation of nanomaterials in hydrogels (hydrated networks of crosslinked polymers) has emerged as a useful method for generating biomaterials with tailored functionality. With the available engineering approaches it is becoming much easier to fabricate nanocomposite hydrogels that display improved performance across an array of electrical, mechanical, and biological properties. In this review, we discuss the fundamental aspects of these materials as well as recent developments that have enabled their application. Specifically, we highlight synthesis and fabrication, and the choice of nanomaterials for multifunctionality as ways to overcome current material property limitations. In addition, we review the use of nanocomposite hydrogels within the framework of biomedical and pharmaceutical disciplines.
Collapse
Affiliation(s)
- Adnan Memic
- Center of Nanotechnology, King Abdulaziz University, Jeddah, Saudi Arabia. Department of Medicine, Center for Biomedical Engineering, Brigham and Women's Hospital, Harvard Medical School, Cambridge, MA 02138, USA
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Wang Z, Xia J, Cai F, Zhang F, Yang M, Bi S, Gui R, Li Y, Xia Y. Aptamer-functionalized hydrogel as effective anti-cancer drugs delivery agents. Colloids Surf B Biointerfaces 2015; 134:40-6. [PMID: 26142627 DOI: 10.1016/j.colsurfb.2015.06.031] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/02/2015] [Accepted: 06/12/2015] [Indexed: 10/23/2022]
Abstract
An aptamer-functionalized hydrogel has been developed, which can be regulated by the AS1411 aptamer with the sol-gel conversion. Also the hydrogel can be further utilized for the controlled encapsulation and release of the cancer drugs. Specially, the AS1411 initiates the hybridization of acrydite-modified oligonucleotides to form the hydrogels and the presence of the target protein nucleolin leads the gel to dissolve as a result of reducing the cross-linking density by competitive target-aptamer binding. Based on the rheology of hydrogels, it is possible to utilize this material for storing and releasing molecules. In this research, the cancer drug doxorubicin is encapsulated inside the gel during the formation of the hydrogel and then released in the presence of nucleolin. Further experiments are carried out to prove the specific recognition of target matter. In vitro researches confirm that the aptamer-functionalized hydrogels can be used as drug carriers in targeted therapy and other biotechnological applications.
Collapse
Affiliation(s)
- Zonghua Wang
- Collaborative Innovation Center for Marine Biomass Fiber Materials and Textiles, Laboratory of Fiber Materials and Modern Textile, The Growing Base for State Key Laboratory, College of Chemical Science and Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, Ningxia Road 308, Qingdao 266071, Shandong, China.
| | - Jianfei Xia
- Collaborative Innovation Center for Marine Biomass Fiber Materials and Textiles, Laboratory of Fiber Materials and Modern Textile, The Growing Base for State Key Laboratory, College of Chemical Science and Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, Ningxia Road 308, Qingdao 266071, Shandong, China
| | - Feng Cai
- Collaborative Innovation Center for Marine Biomass Fiber Materials and Textiles, Laboratory of Fiber Materials and Modern Textile, The Growing Base for State Key Laboratory, College of Chemical Science and Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, Ningxia Road 308, Qingdao 266071, Shandong, China
| | - Feifei Zhang
- Collaborative Innovation Center for Marine Biomass Fiber Materials and Textiles, Laboratory of Fiber Materials and Modern Textile, The Growing Base for State Key Laboratory, College of Chemical Science and Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, Ningxia Road 308, Qingdao 266071, Shandong, China
| | - Min Yang
- Collaborative Innovation Center for Marine Biomass Fiber Materials and Textiles, Laboratory of Fiber Materials and Modern Textile, The Growing Base for State Key Laboratory, College of Chemical Science and Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, Ningxia Road 308, Qingdao 266071, Shandong, China
| | - Sai Bi
- Collaborative Innovation Center for Marine Biomass Fiber Materials and Textiles, Laboratory of Fiber Materials and Modern Textile, The Growing Base for State Key Laboratory, College of Chemical Science and Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, Ningxia Road 308, Qingdao 266071, Shandong, China
| | - Rijun Gui
- Collaborative Innovation Center for Marine Biomass Fiber Materials and Textiles, Laboratory of Fiber Materials and Modern Textile, The Growing Base for State Key Laboratory, College of Chemical Science and Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, Ningxia Road 308, Qingdao 266071, Shandong, China
| | - Yanhui Li
- Collaborative Innovation Center for Marine Biomass Fiber Materials and Textiles, Laboratory of Fiber Materials and Modern Textile, The Growing Base for State Key Laboratory, College of Chemical Science and Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, Ningxia Road 308, Qingdao 266071, Shandong, China
| | - Yanzhi Xia
- Collaborative Innovation Center for Marine Biomass Fiber Materials and Textiles, Laboratory of Fiber Materials and Modern Textile, The Growing Base for State Key Laboratory, College of Chemical Science and Engineering, Shandong Sino-Japanese Center for Collaborative Research of Carbon Nanomaterials, Qingdao University, Ningxia Road 308, Qingdao 266071, Shandong, China
| |
Collapse
|