1
|
Guo Z, Huang T, Lv X, Yin R, Wan P, Li G, Zhang P, Xiao C, Chen X. Tumor microenvironment-activated polypeptide nanoparticles for oncolytic immunotherapy. Biomaterials 2025; 314:122870. [PMID: 39369669 DOI: 10.1016/j.biomaterials.2024.122870] [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: 07/31/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/08/2024]
Abstract
Cationic oncolytic polypeptides have gained increasing attention owing to their ability to directly lyse cancer cells and activate potent antitumor immunity. However, the low tumor cell selectivity and inherent toxicity induced by positive charges of oncolytic polypeptides hinder their systemic application. Herein, a tumor microenvironment-responsive nanoparticle (DNP) is developed by the self-assembly of a cationic oncolytic polypeptide (PLP) with a pH-sensitive anionic polypeptide via electrostatic interactions. After the formation of DNP, the positive charges of PLP are shielded. DNPs can keep stable in physiological conditions (pH 7.4) but respond to acidic tumor microenvironment (pH 6.8) to release oncolytic PLP. As a result, DNPs evoke potent immunogenic cell death by disrupting cell membranes, damaging mitochondria and increasing intracellular levels of reactive oxygen species. In vivo results indicate that DNPs significantly improve the biocompatibility of PLP, and inhibit tumor growth, recurrence and metastasis by direct oncolysis and activation of antitumor immune responses. In summary, these results indicate that pH-sensitive DNPs represent a prospective strategy to improve the tumor selectivity and biosafety of cationic polymers for oncolytic immunotherapy.
Collapse
Affiliation(s)
- Zhihui Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Tianze Huang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Xueli Lv
- College of Chemistry, Jilin University, Changchun, 130012, PR China
| | - Renyong Yin
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| | - Pengqi Wan
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | - Gao Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China.
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China.
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, 230026, PR China
| |
Collapse
|
2
|
Wang J, Yuan Y, Su C, Bao Y, Xu W, Yao Y, Liang L, Zeng Y, Xiong M. pH-Ultrasensitive Membranolytic Polyesters with Alternating Sequence of Ionizable and Hydrophobic Groups for Selective Oncolytic Therapy. J Am Chem Soc 2024. [PMID: 39731565 DOI: 10.1021/jacs.4c14199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2024]
Abstract
Oncolytic therapy, inducing cell death via cell membrane lysis, holds considerable promise in cancer treatment. However, achieving precise control over the structure and function of oncolytic materials for highly selective oncolytic therapy is a key challenge in the context of the subtle differences between tumor and normal tissues/cells. Herein, we report the development of pH-ultrasensitive oncolytic polyesters (pOPs) with an alternating sequence of ionizable and hydrophobic groups. This design enables a refined "OFF" to "ON" switch within 0.2 pH units, ensuring high selectivity in membranolytic activity and cytotoxicity of pOPs between the pH levels of normal tissues and tumor acidity. The top-performing pOP, P(P-AC7), demonstrated a maximum tolerated dose of >100 mg kg-1 after intravenous administration and potent cytotoxicity at pH 6.8. Notably, the molecular weight of P(P-AC7) had a minimal effect on its pH-dependent cytotoxicity once the degree of polymerization was ≥49, ensuring consistency in properties across batches. P(P-AC7) exerts membranolytic activity by interacting with phosphatidylserine at pH 6.8 and shows high antitumor efficacy in various tumor models. Overall, we developed a strategy to develop oncolytic polymers with a precise structure for selective oncolytic therapy.
Collapse
Affiliation(s)
- Jihong Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yueling Yuan
- Shenshan Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Shanwei 516621, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Chanjuan Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Yan Bao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Medical Research Center, Sun Yat-Sen Memorial Hospital, Foshan 528200, China
| | - Weide Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yandan Yao
- Shenshan Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Shanwei 516621, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Lifang Liang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yuxuan Zeng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Menghua Xiong
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
- National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| |
Collapse
|
3
|
Li Y, Ji W, Wang C, Chang L, Zhang Q, Gao J, Wang T, Wu W. Poly l-Lactic Acid Nanofiber Membrane Effectively Inhibits Liver Cancer Cells Growth and Prevents Postoperative Residual Cancer Recurrence. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39681510 DOI: 10.1021/acsami.4c18625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Electrospun nanocarrier systems, widely employed in the medical field, exhibit the capability to encapsulate multiple drugs and mitigate complications. Doxorubicin hydrochloride (DOX) represents a frequently utilized chemotherapeutic agent for liver cancer patients. Sodium bicarbonate (SB) serves to neutralize the acidic tumor microenvironment, while ibuprofen (IBU) attenuates inflammatory factor production. The combination of these three commonly used drugs facilitates antitumor efficacy and relapse prevention. Composite fibrous membranes were prepared by incorporating the antitumor drug DOX into MSN, which was then codispersed with IBU in a poly l-lactic acid (PLLA) electrospinning solution after acid sensitization using SB. The resulting membrane was characterized using transmission electron microscopy and scanning electron microscopy. The toxic effect of this fibrous membrane and its pro-apoptotic effect on tumor cells were evaluated, along with the expression of cell proliferation-related factors, immune/inflammatory factors, and apoptosis-related factors. Immunohistochemistry and HE staining confirmed its ability to inhibit recurrence of postoperative residual cancer without causing toxicity to vital organs. The PLLA-MSN@DOX-SB-IBU nanofibrous membrane not only mitigates the cardiotoxicity associated with DOX but also inhibits tumor cell proliferation and enhances the tumor microenvironment, demonstrating significant antitumor efficacy. Furthermore, it effectively prevents the recurrence of residual cancer postsurgery while exhibiting excellent biocompatibility. The PLLA-MSN@DOX-SB-IBU nanofibrous membrane demonstrates significant potential in impeding the progression of hepatocellular carcinoma and mitigating the recurrence of residual cancer following surgical intervention for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yanxu Li
- General Surgery Department, Affiliated Hospital of Yang Zhou University, Medical College of Yang Zhou University, Yang Zhou University, Yang Zhou, Jiang Su 225009, China
| | - Weiben Ji
- General Surgery Department, Affiliated Hospital of Yang Zhou University, Medical College of Yang Zhou University, Yang Zhou University, Yang Zhou, Jiang Su 225009, China
| | - Chaoying Wang
- General Surgery Department, Affiliated Hospital of Yang Zhou University, Medical College of Yang Zhou University, Yang Zhou University, Yang Zhou, Jiang Su 225009, China
| | - Lai Chang
- Taixing People's Hospital in Jiangsu Province, Tai Zhou, Jiang Su 225400, China
| | - Quan Zhang
- College of Veterinary Medicine, Yangzhou University, Yang Zhou, Jiang Su 225009, China
| | - Jiefeng Gao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yang Zhou, Jiang Su 225009, China
| | - Tao Wang
- College of Veterinary Medicine, Yangzhou University, Yang Zhou, Jiang Su 225009, China
| | - Wei Wu
- General Surgery Department, Affiliated Hospital of Yang Zhou University, Medical College of Yang Zhou University, Yang Zhou University, Yang Zhou, Jiang Su 225009, China
| |
Collapse
|
4
|
Chen L, He Y, Lan J, Li Z, Gu D, Nie W, Zhang T, Ding Y. Advancements in nano drug delivery system for liver cancer therapy based on mitochondria-targeting. Biomed Pharmacother 2024; 180:117520. [PMID: 39395257 DOI: 10.1016/j.biopha.2024.117520] [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: 07/10/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024] Open
Abstract
Based on poor efficacy and non-specific toxic side effects of conventional drug therapy for liver cancer, nano-based drug delivery system (NDDS) offers the advantage of drug targeting delivery. Subcellular targeting of nanomedicines on this basis enables more precise and effective termination of tumor cells. Mitochondria, as the crucial cell powerhouse, possesses distinctive physical and chemical properties in hepatoma cells different from that in hepatic cells, and controls apoptosis, tumor metastasis, and cellular drug resistance in hepatoma cells through metabolism and dynamics, which serves as a good choice for drug targeting delivery. Thus, mitochondria-targeting NDDS have become a recent research focus, showcasing the design of cationic nanoparticles, metal nanoparticles, mitochondrial peptide modification and so on. Although many studies have shown good results regarding anti-tumor efficacy, it is a long way to go before the successful translation of clinical application. Based on these, we summarized the specificity and importance of mitochondria in hepatoma cells, and reviewed the current mitochondria-targeting NDDS for liver cancer therapy, aiming to provide a better understanding for current development process, strengths and weaknesses of mitochondria-targeting NDDS as well as informing subsequent improvements and developments.
Collapse
Affiliation(s)
- Lixia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yitian He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Donghao Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenlong Nie
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
5
|
Harini K, Girigoswami K, Thirumalai A, Girigoswami A. Polymer-Based Antimicrobial Peptide Mimetics for Treating Multi-drug Resistant Infections: Therapy and Toxicity Evaluation. Int J Pept Res Ther 2024; 30:64. [DOI: 10.1007/s10989-024-10648-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2024] [Indexed: 01/03/2025]
|
6
|
Jiang M, Li W, Liang J, Pang M, Li S, Xu G, Zhu M, Liang H, Zhang Z, Yang F. Developing a Palladium(II) Agent to Overcome Multidrug Resistance and Metastasis of Liver Tumor by Targeted Multiacting on Tumor Cell, Inactivating Cancer-Associated Fibroblast and Activating Immune Response. J Med Chem 2024; 67:16296-16310. [PMID: 39238096 DOI: 10.1021/acs.jmedchem.4c01175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2024]
Abstract
To targeted overcome the multidrug resistance (MDR) and metastasis of liver tumors, we proposed to develop a palladium (Pd) agent based on a specific residue of human serum albumin (HSA) for multiacting on tumor cell and other components in the tumor microenvironment. To this end, a series of Pd(II) 2-acetylpyridine thiosemicarbazone compounds were optimized to obtain a Pd(II) compound (5b) with significant cytotoxicity against HepG2/ADM cells. Subsequently, we constructed a HSA-5b complex delivery system and revealed the structural mechanism of HSA delivering 5b. Importantly, 5b/HSA-5b effectively inhibited the growth and metastasis of multidrug resistant liver tumors, and HSA enhanced the targeting ability of 5b and reduced its side effects in vivo. Furthermore, we confirmed the mechanisms of 5b/HSA-5b integrating to overcome MDR and metastasis of liver tumors: multiacting on cancer cell, activating immune response, and inactivating cancer-associated fibroblasts.
Collapse
Affiliation(s)
- Ming Jiang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Wenjuan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Jinzhe Liang
- MOE Key Laboratory of Bioinorganic and Synthetic Chemistry, Guangdong Basic Research Center of Excellence for Functional Molecular Engineering, School of Chemistry, Sun Yat-Sen University, Guangzhou 510006, PR China
| | - Min Pang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Minghui Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, PR China
| |
Collapse
|
7
|
Li Y, Li C, Yan J, Liao Y, Qin C, Wang L, Huang Y, Yang C, Wang J, Ding X, Yang YY, Yuan P. Polymeric micellar nanoparticles for effective CRISPR/Cas9 genome editing in cancer. Biomaterials 2024; 309:122573. [PMID: 38677222 DOI: 10.1016/j.biomaterials.2024.122573] [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: 11/12/2023] [Revised: 03/21/2024] [Accepted: 04/12/2024] [Indexed: 04/29/2024]
Abstract
The clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein 9 (Cas9) gene editing has attracted extensive attentions in various fields, however, its clinical application is hindered by the lack of effective and safe delivery system. Herein, we reported a cationic micelle nanoparticle composed of cholesterol-modified branched small molecular PEI (PEI-CHO) and biodegradable PEG-b-polycarbonate block copolymer (PEG-PC), denoted as PEG-PC/PEI-CHO/pCas9, for the CRISPR/Cas9 delivery to realize genomic editing in cancer. Specifically, PEI-CHO condensed pCas9 into nanocomplexes, which were further encapsulated into PEG-PC nanoparticles (PEG-PC/PEI-CHO/pCas9). PEG-PC/PEI-CHO/pCas9 had a PEG shell, protecting DNA from degradation by nucleases. Enhanced cellular uptake of PEG-PC/PEI-CHO/pCas9 nanoparticles was observed as compared to that mediated by Lipo2k/pCas9 nanoparticles, thus leading to significantly elevated transfection efficiency after escaping from endosomes via the proton sponge effect of PEI. In addition, the presence of PEG shell greatly improved biocompatibility, and significantly enhanced the in vivo tumor retention of pCas9 compared to PEI-CHO/pCas9. Notably, apparent downregulation of GFP expression could be achieved both in vitro and in vivo by using PEG-PC/PEI-CHO/pCas9-sgGFP nanoparticles. Furthermore, PEG-PC/PEI-CHO/pCas9-sgMcl1 induced effective apoptosis and tumor suppression in a HeLa tumor xenograft mouse model by downregulating Mcl1 expression. This work may provide an alternative paradigm for the efficient and safe genome editing in cancer.
Collapse
Affiliation(s)
- Yuzhen Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Chun Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Jiachang Yan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Ying Liao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Chengyuan Qin
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Lingyin Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yi Huang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Chuan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Republic of Singapore
| | - Jianwei Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China.
| | - Xin Ding
- School of Medicine, Sun Yat-sen University, Shenzhen, 518107, China.
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 20 Biopolis Way, Centros #06-01, Singapore, 138668, Republic of Singapore.
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China; School of Pharmaceutical Sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
8
|
Li Z, Wang S, Zhao L, Feng S, Che H. Synthesis and Characterization of Guanidinylated CO-Releasing Micelles Based on Biodegradable Polycarbonate. Biomacromolecules 2024; 25:5149-5159. [PMID: 39045816 DOI: 10.1021/acs.biomac.4c00542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
As one of the gaseous signals in living cells, carbon monoxide (CO) not only participates in many biological activities but also serves as a therapeutic agent for the treatment of diseases. However, the limited applicability of CO in gas therapy emerges from the inconvenience of direct administration of CO. Here we reported the construction of guanidinylated CO-releasing micelles, which are composed of poly(trimethylene carbonate) (PTMC)-based CO donors. The in vitro studies demonstrated that micelles in the presence of light irradiation can induce cancer death, whereas no obvious toxicity to normal cells was observed. Moreover, the functionalization of guanidine groups imparts improved cellular uptake efficiency to micelles owing to the specific interactions with the surface of cells, which synergistically increase the anticancer capacity of the system. The guanidine-functionalized CO-releasing micelles provide a new strategy for the construction of CO-releasing nanocarriers, which are expected to find applications in gas therapeutics.
Collapse
Affiliation(s)
- Zhezhe Li
- Department of Chemical Engineering, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Suzhen Wang
- Department of Chemical Engineering, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Lili Zhao
- Department of Chemical Engineering, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Shaofeng Feng
- Department of Chemical Engineering, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| | - Hailong Che
- Department of Chemical Engineering, School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
| |
Collapse
|
9
|
Chen P, Huang X, Li W, Wen W, Cao Y, Li J, Huang Y, Hu Y. Myeloid-derived growth factor in diseases: structure, function and mechanisms. Mol Med 2024; 30:103. [PMID: 39030488 PMCID: PMC11264862 DOI: 10.1186/s10020-024-00874-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024] Open
Abstract
Myeloid-derived growth factor (MYDGF) is a novel secreted protein with potent antiapoptotic and tissue-repairing properties that is present in nearly 140 human tissues and cell lines, with the highest abundance in the oral epithelium and skin. Initially, MYDGF was found in bone marrow-derived monocytes and macrophages for cardioprotection and repair after myocardial infarction. Subsequent studies have shown that MYDGF plays an important role in other cardiovascular diseases (e.g., atherosclerosis and heart failure), metabolic disorders, renal disease, autoimmune/inflammatory disorders, and cancers. Although the underlying mechanisms have not been fully explored, the role of MYDGF in health and disease may involve cell apoptosis and proliferation, tissue repair and regeneration, anti-inflammation, and glycolipid metabolism regulation. In this review, we summarize the current progress in understanding the role of MYDGF in health and disease, focusing on its structure, function and mechanisms. The graphical abstract shows the current role of MYDGF in different organs and diseases (Fig. 1).
Collapse
Affiliation(s)
- Peng Chen
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Xiaohui Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Weiwen Li
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Weixing Wen
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Yue Cao
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Jiahuan Li
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China
| | - Yuli Huang
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China.
- The George Institute for Global Health, Faculty of Medicine, University of New South Wales, Sydney, NSW2006, Australia.
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation Research, Guangzhou, 510000, China.
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China.
| | - Yunzhao Hu
- Department of Cardiology, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China.
- Medical Research Center, Shunde Hospital, Southern Medical University, The First People's Hospital of Shunde, NO. 1 Jiazi Road, Lunjiao, Shunde District, Foshan City, Guangdong, 528308, China.
| |
Collapse
|
10
|
Yin R, Wan P, Guo Z, Yi X, Zhang P, Shen W, Chen L, Xiao C, Chen X. Enzyme-responsive oncolytic polypeptide for tumor therapy. Acta Biomater 2024; 181:415-424. [PMID: 38704115 DOI: 10.1016/j.actbio.2024.04.044] [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: 01/12/2024] [Revised: 04/10/2024] [Accepted: 04/26/2024] [Indexed: 05/06/2024]
Abstract
Host defense peptide-mimicking cationic oncolytic polymers have attracted increasing attention for cancer treatment in recent years. However, polymers with large amounts of positive charge may cause rapid clearance and severe off-target toxicity. To facilitate in vivo application, an alkaline phosphatase (ALP)-responsive oncolytic polypeptide precursor (C12-PLL/PA) has been reported in this work. C12-PLL/PA could be hydrolyzed into the active form of the oncolytic polypeptide (C12-PLL) by the extracellular alkaline phosphatase within solid tumors, thereby resulting in the conversion of the negative charge to positive charge and restoring its membrane-lytic activity. Detailed mechanistic studies showed that C12-PLL/PA could effectively destroy cancer cell membranes and subsequently result in rapid necrosis of cancer cells. More importantly, C12-PLL/PA significantly inhibited the tumor growth in the 4T1 orthotopic breast tumor model with negligible side effects. In summary, these findings demonstrated that the shielding of the amino groups with phosphate groups represents a secure and effective strategy to develop cationic oncolytic polypeptide, which represents a valuable reference for the design of enzyme-activated oncolytic polymers. STATEMENT OF SIGNIFICANCE: Recently, there has been a growing interest in fabricating host defense peptide-mimicking cationic oncolytic polymers for cancer therapy. However, there remain concerns about the tumor selectivity and off-target toxicity of these cationic polymers. In this study, an alkaline phosphatase-responsive oncolytic polypeptide precursor (C12-PLL/PA) has been developed to selectively target cancer cells while sparing normal cells. Mechanistic investigations demonstrated that C12-PLL/PA effectively disrupted cancer cell membranes, leading to rapid necrosis. Both in vitro and in vivo experiments showed promising anticancer activity and reliable safety of C12-PLL/PA. The findings suggest that this synthetic enzyme-responsive polypeptide holds potential as a tumor-specific oncolytic polymer, paving the way for future applications in cancer therapy.
Collapse
Affiliation(s)
- Renyong Yin
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Penqi Wan
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Zhihui Guo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China
| | - Xuan Yi
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, PR China.
| | - Li Chen
- Department of Chemistry, Northeast Normal University, Changchun 130024, PR China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China.
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China; School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, PR China; Jilin Biomedical Polymers Engineering Laboratory, Changchun 130022, PR China.
| |
Collapse
|
11
|
Kumar N, Nakaji-Hirabayashi T, Kato M, Matsumura K, Rajan R. Design of Highly Selective Zn-Coordinated Polyampholyte for Cancer Treatment and Inhibition of Tumor Metastasis. Biomacromolecules 2024; 25:1481-1490. [PMID: 38343080 DOI: 10.1021/acs.biomac.3c01044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Developing anticancer agents with negligible cytotoxicity against normal cells while mitigating multidrug resistance and metastasis is challenging. Previously reported cationic polymers have effectively eradicated cancers but are clinically unsuitable due to their limited selectivity. Herein, a series of poly(l-lysine)- and nicotinic acid-based polymers were synthesized using varying amounts of dodecylsuccinic anhydride. Zn-coordinating polymers concealed their cationic charge and enhanced selectivity. These Zn-bound polymers were highly effective against liver and colon cancer cells (HepG2 and Colon 26, respectively) and prevented cancer cell migration. They also displayed potent anticancer activity against drug-resistant cell lines (COR-L23/R): their cationic structure facilitated cancer cell membrane disruption. Compared to these polymers, doxorubicin was less selective and less efficacious against drug-resistant cell lines and was unable to prevent cell migration. These polymers are potential cancer treatment agents, offering a promising solution for mitigating drug resistance and tumor metastasis and representing a novel approach to designing cancer therapeutics.
Collapse
Affiliation(s)
- Nishant Kumar
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Tadashi Nakaji-Hirabayashi
- Faculty of Engineering, Academic Assembly, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
- Graduate School of Innovative Life Science, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Moe Kato
- Graduate School of Innovative Life Science, University of Toyama, 3190 Gofuku, Toyama 930-8555, Japan
| | - Kazuaki Matsumura
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Robin Rajan
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| |
Collapse
|
12
|
Ngo TLH, Wang KL, Pan WY, Ruan T, Lin YJ. Immunomodulatory Prodrug Micelles Imitate Mild Heat Effects to Reshape Tumor Microenvironment for Enhanced Cancer Immunotherapy. ACS NANO 2024; 18:5632-5646. [PMID: 38344992 PMCID: PMC10883120 DOI: 10.1021/acsnano.3c11186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Physical stimulation with mild heat possesses the notable ability to induce immunomodulation within the tumor microenvironment (TME). It transforms the immunosuppressive TME into an immune-active state, making tumors more receptive to immune checkpoint inhibitor (ICI) therapy. Transient receptor potential vanilloid 1 (TRPV1), which can be activated by mild heat, holds the potential to induce these alterations in the TME. However, achieving precise temperature control within tumors while protecting neighboring tissues remains a significant challenge when using external heat sources. Taking inspiration from the heat sensation elicited by capsaicin-containing products activating TRPV1, this study employs capsaicin to chemically stimulate TRPV1, imitating immunomodulatory benefits akin to those induced by mild heat. This involves developing a glutathione (GSH)-responsive immunomodulatory prodrug micelle system to deliver capsaicin and an ICI (BMS202) concurrently. Following intravenous administration, the prodrug micelles accumulate at the tumor site through the enhanced permeability and retention effect. Within the GSH-rich TME, the micelles disintegrate and release capsaicin and BMS202. The released capsaicin activates TRPV1 expressed in the TME, enhancing programmed death ligand 1 expression on tumor cell surfaces and promoting T cell recruitment into the TME, rendering it more immunologically active. Meanwhile, the liberated BMS202 blocks immune checkpoints on tumor cells and T cells, activating the recruited T cells and ultimately eradicating the tumors. This innovative strategy represents a comprehensive approach to fine-tune the TME, significantly amplifying the effectiveness of cancer immunotherapy by exploiting the TRPV1 pathway and enabling in situ control of immunomodulation within the TME.
Collapse
Affiliation(s)
- Thi-Lan-Huong Ngo
- Research Center for Applied Sciences, Academia Sinica, Taipei, 115201, Taiwan
| | - Kuan-Lin Wang
- Research Center for Applied Sciences, Academia Sinica, Taipei, 115201, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, 242062, Taiwan
| | - Wen-Yu Pan
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, 110301, Taiwan
- Ph.D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, 110301, Taiwan
| | - Ting Ruan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City, 242062, Taiwan
| | - Yu-Jung Lin
- Research Center for Applied Sciences, Academia Sinica, Taipei, 115201, Taiwan
| |
Collapse
|
13
|
Sim R, Yang C, Yang YY. Chemical Proteomics and Morphological Profiling Revealing MYDGF as a Target for Synthetic Anticancer Macromolecules. Biomacromolecules 2024; 25:1047-1057. [PMID: 38225889 DOI: 10.1021/acs.biomac.3c01101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Biodegradable guanidinium-functionalized polycarbonates kill cancer cells via membrane translocation without causing resistance after repeated use, but the exact molecular targets of the polycarbonates are unknown. Here, we investigate the protein targets of the polycarbonates through affinity-based protein profiling and report myeloid-derived growth factor (MYDGF) as the main protein target. Direct binding of the polycarbonates to MYDGF protein is validated through biolayer interferometry. MYDGF is overexpressed in a range of cancer cells, and knockdown of MYDGF is shown to reduce cell proliferation in cancer cells. Through morphological profiling, we also identify similarities in phenotypic effects of the functionalized polycarbonates with topoisomerase I inhibitors, MDM2 inhibitors, and phosphatidylinositol 3kinase inhibitors against cancer cells, suggesting a common mechanism through the PIK3/AKT pathway leading to apoptosis. These findings present the first macromolecular compound targeting MYDGF and may serve as an example for MYDGF modulation as a potential new target for macromolecular chemotherapeutic development.
Collapse
Affiliation(s)
- Rachel Sim
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Chuan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| | - Yi Yan Yang
- Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Way, Centros #06-01, Singapore 138668, Singapore
| |
Collapse
|
14
|
Guo F, Jiao Y, Ding W, Du Y, Luo S, Wang M, Wang Y, Wu F, Wang L, Yang G. Synergistic effects of multidrug/material combination deliver system for anti-mutidrug-resistant tumor. Int J Pharm 2024; 649:123669. [PMID: 38056797 DOI: 10.1016/j.ijpharm.2023.123669] [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: 09/03/2023] [Revised: 11/04/2023] [Accepted: 12/02/2023] [Indexed: 12/08/2023]
Abstract
Multidrug resistance (MDR) is a public health issue of particular concern, for which nanotechnology-based multidrug delivery systems are considered among the most effective suppressive strategies for such resistance in tumors. However, for such strategies to be viable, the notable shortcomings of reduced loading efficiency and uncontrollable drug release ratio need to be addressed. To this end, we developed a novel "multidrug/material" co-delivery system, using d-α-tocopheryl polyethylene glycol 1000 succinate (TPGS, P-gp efflux pump inhibitor) and poly(amidoamine) (PAMAM) to fabricate a precursor material with the properties of reversing MDR and having a long-cycle. Further, to facilitate multidrug co-delivery, we loaded doxorubicin(Dox) and curcumin(Cur, cardiotoxicity modifier and P-gp inhibitor) into PAMAM-TPGS nano-micelles respectively, and mixed in appropriate proportions. The multidrug/material co-delivery system thus obtained was characterized by high drug loading and a controllable drug release ratio in the physiological environment. More importantly, in vitro and in vivo pharmacodynamic studies indicated that the multidrug/material co-delivery system facilitated the reversal of MDR. Moreover, the system has increased anti-tumor activity and is biologically safe. We accordingly propose that the "multidrug/material" co-delivery system developed in this study could serve as a potential platform for reversing MDR and achieving safe and effective clinical treatment.
Collapse
Affiliation(s)
- Fangyuan Guo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yunlong Jiao
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Wenqin Ding
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yinzhou Du
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shuai Luo
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Mengqi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yujia Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Fang Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Lianyi Wang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China
| | - Gensheng Yang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou 310014, China; Research Institute of Pharmaceutical Particle Technology, Zhejiang University of Technology, Hangzhou 310014, China.
| |
Collapse
|
15
|
Liu H, Shen W, Liu W, Yang Z, Yin D, Xiao C. From oncolytic peptides to oncolytic polymers: A new paradigm for oncotherapy. Bioact Mater 2024; 31:206-230. [PMID: 37637082 PMCID: PMC10450358 DOI: 10.1016/j.bioactmat.2023.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 07/18/2023] [Accepted: 08/08/2023] [Indexed: 08/29/2023] Open
Abstract
Traditional cancer therapy methods, especially those directed against specific intracellular targets or signaling pathways, are not powerful enough to overcome tumor heterogeneity and therapeutic resistance. Oncolytic peptides that can induce membrane lysis-mediated cancer cell death and subsequent anticancer immune responses, has provided a new paradigm for cancer therapy. However, the clinical application of oncolytic peptides is always limited by some factors such as unsatisfactory bio-distribution, poor stability, and off-target toxicity. To overcome these limitations, oncolytic polymers stand out as prospective therapeutic materials owing to their high stability, chemical versatility, and scalable production capacity, which has the potential to drive a revolution in cancer treatment. This review provides an overview of the mechanism and structure-activity relationship of oncolytic peptides. Then the oncolytic peptides-mediated combination therapy and the nano-delivery strategies for oncolytic peptides are summarized. Emphatically, the current research progress of oncolytic polymers has been highlighted. Lastly, the challenges and prospects in the development of oncolytic polymers are discussed.
Collapse
Affiliation(s)
- Hanmeng Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Wei Shen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, Anhui, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, Anhui, 230012, China
| | - Wanguo Liu
- Department of Orthopaedic Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033, China
| | - Zexin Yang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department (AUCM), Hefei, Anhui, 230012, China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, China
| |
Collapse
|
16
|
Kenawy ER, El-Khalafy SH, Abosharaf HA, El-Nshar EM, Ghazy AR, Azaam MM. Synthesis, Characterization, and Anticancer Potency of Branched Poly (p-Hydroxy Styrene) Schiff-Bases. Macromol Biosci 2023; 23:e2300090. [PMID: 37376773 DOI: 10.1002/mabi.202300090] [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: 03/06/2023] [Revised: 06/20/2023] [Indexed: 06/29/2023]
Abstract
A significant issue in cancer biology is finding anticancer therapies that effectively kill cancer cells. Through the use of several aldehydes, Schiff bases based on branched poly (p-hydroxy styrene) are created. The branched polymer is first chloroacetylated, then aminated with 1,4-phenylenediamine, and finally, aldehydes are reacted with the aminated polymer to produce the Schiff base compounds. Through the utilization of FTIR, TGA, XRD, NMR, and elemental analysis, all synthesized Schiff-bases are identified and characterized. Further, the antineoplastic potential of all Schiff bases is evaluated against different cancer cell lines. The results gained from this study indicate that the Schiff base polymers have cytotoxic power against cancer cells depending on cancer cell type and this antiproliferation potency is dose-concentration dependent. Importantly, the prepared S1 Schiff-base polymer shows potent cytotoxicity and is able to trigger the apoptosis and reactive oxygen species (ROS) in MCF-7 cells. Further, it downregulates VEGFR protein expression. The Schiff base polymers would have extensive applications in the biological disciplines.
Collapse
Affiliation(s)
- El-Refaie Kenawy
- Department of Chemistry, Faculty of Science, University of Tanta, Tanta, 31527, Egypt
| | - Sahar H El-Khalafy
- Department of Chemistry, Faculty of Science, University of Tanta, Tanta, 31527, Egypt
| | - Hamed A Abosharaf
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Esraa M El-Nshar
- Chemistry Department, Faculty of Dentistry, Sinai University, Kantara, 41612, Egypt
| | - Ahmed R Ghazy
- Laser Laboratory, Physics Department, Faculty of Sciences, Tanta University, Tanta, 31527, Egypt
| | - Mohamed M Azaam
- Department of Chemistry, Faculty of Science, University of Tanta, Tanta, 31527, Egypt
| |
Collapse
|
17
|
Chen L, Nabil A, Fujisawa N, Oe E, Li K, Ebara M. A facile, flexible, and multifunctional thermo-chemotherapy system for customized treatment of drug-resistant breast cancer. J Control Release 2023; 363:550-561. [PMID: 37804880 DOI: 10.1016/j.jconrel.2023.10.010] [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: 06/27/2023] [Revised: 10/02/2023] [Accepted: 10/04/2023] [Indexed: 10/09/2023]
Abstract
Anticancer drug resistance invariably emerges and poses a significant barrier to curative therapy for various breast cancers. This results in a lack of satisfactory therapeutic medicine for cancer treatment. Herein, a universal vector system for drug-resistance breast cancer was designed to meet the needs of reversed multidrug resistance, thermo-chemotherapy, and long-term drug release behavior. The vector system comprises polycaprolactone (PCL) nanofiber mesh and magnetic nanoparticles (MNPs). PCL has excellent biocompatibility and electrospinning performance. In this study, MNPs were tailored to be thermogenic in response to an alternating magnetic field (AMF). PCL nanofiber can deliver various chemotherapy drugs, and suitable MNPs encapsulated in the nanofiber can generate hyperthermia and synergistic effect with those chemotherapy drugs. Therefore, a more personalized treatment system can be developed for different breast malignancies. In addition, the PCL nanofiber mesh (NFM) enables sustained release of the drugs for up two months, avoiding the burden on patients caused by repeated administration. Through model drugs doxorubicin (DOX) and chemosensitizers curcumin (CUR), we systematically verified the therapeutic effect of DOX-resistance breast cancer and inhibition of tumor generation in vivo. These findings represent a multifaceted platform of importance for validating strategic reversed MDR in pursuit of promoted thermo-chemotherapeutic outcomes. More importantly, the low cost and excellent safety and efficacy of this nanofiber mesh demonstrate that this can be customized multi-function vector system may be a promising candidate for refractory cancer therapy in clinical.
Collapse
Affiliation(s)
- Lili Chen
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | - Ahmed Nabil
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt
| | - Nanami Fujisawa
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt
| | - Emiho Oe
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Biotechnology and Life Sciences Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef 62511, Egypt
| | - Kai Li
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan; Ph.D. Program in Humanics, School of Integrative and Global Majors, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Mitsuhiro Ebara
- Research Center for Macromolecules and Biomaterials, National Institute for Materials Science (NIMS), 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan; Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan; Department of Materials Science and Technology, Tokyo University of Science, Tokyo 125-8585, Japan.
| |
Collapse
|
18
|
Kong J, Xu S, Dai Y, Wang Y, Zhao Y, Zhang P. Study of the Fe 3O 4@ZIF-8@Sor Composite Modified by Tannic Acid for the Treatment of Sorafenib-Resistant Hepatocellular Carcinoma. ACS OMEGA 2023; 8:39174-39185. [PMID: 37901534 PMCID: PMC10601084 DOI: 10.1021/acsomega.3c04215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 08/30/2023] [Indexed: 10/31/2023]
Abstract
Chemotherapeutic agents fail in clinical chemotherapy in the absence of targeting and acquired resistance. We, therefore, synthesized Fe3O4@ZIF-8@Sor@TA nanocomposite drugs based on the drug delivery properties of nanomaterials. ZIF-8 is a nanomaterial with a porous structure that can load anticancer drugs. The nanodrug used the paramagnetic property of Fe3O4 to deliver sorafenib (Sor) precisely to the tumor site, then used the pH responsiveness of ZIF-8 to slowly release Sor in the tumor microenvironment, and finally used tannic acid (TA) to inhibit P-glycoprotein to suppress the Sor resistance. The results of material characterization presented that the prepared material was structurally stable and was able to achieve a cumulative drug release of 38.2% at pH 5.0 for 72 h. The good biocompatibility of the composite was demonstrated by in vitro and in vivo experiments, which could improve antitumor activity and reduce Sor resistance through magnetic targeting TA. In conclusion, the Fe3O4@ZIF-8@Sor@TA material prepared in this study demonstrated high antitumor activity in hepatocellular carcinoma treatment, promising to reduce drug resistance and providing a novel research approach for cancer treatment.
Collapse
Affiliation(s)
- Jianqiao Kong
- Department of General Surgery, Xiangyang No. 1 People’s Hospital, Hubei University
of Medicine, Xiangyang City 441000, China
| | - Song Xu
- Department of General Surgery, Xiangyang No. 1 People’s Hospital, Hubei University
of Medicine, Xiangyang City 441000, China
| | - Yang Dai
- Department of General Surgery, Xiangyang No. 1 People’s Hospital, Hubei University
of Medicine, Xiangyang City 441000, China
| | - Yi Wang
- Department of General Surgery, Xiangyang No. 1 People’s Hospital, Hubei University
of Medicine, Xiangyang City 441000, China
| | - Yun Zhao
- Department of General Surgery, Xiangyang No. 1 People’s Hospital, Hubei University
of Medicine, Xiangyang City 441000, China
| | - Peng Zhang
- Department of General Surgery, Xiangyang No. 1 People’s Hospital, Hubei University
of Medicine, Xiangyang City 441000, China
| |
Collapse
|
19
|
Wang W, Zhang Y, Wang Z, Liu X, Lu S, Hu X. A Native Drug-Free Macromolecular Therapeutic to Trigger Mutual Reinforcing of Endoplasmic Reticulum Stress and Mitochondrial Dysfunction for Cancer Treatment. ACS NANO 2023. [PMID: 37257082 DOI: 10.1021/acsnano.3c03450] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Drug-free macromolecular therapeutics are promising alternatives to traditional drugs. Nanomedicines with multiple organelles targeting can potentially increase the efficacy. Herein, a drug-free macromolecular therapeutic was designed to formulate endoplasmic reticulum (ER) and mitochondria dual-targeting nanoparticles (EMT-NPs), which can synergistically elicit ER stress and mitochondrial dysfunction. In vitro experiments indicated that EMT-NPs could effectively enter ER and mitochondria at an approximate ratio of 2 to 3. Subsequently, EMT-NPs could upregulate ER stress-related protein expression (IRE1α, CHOP), boosting calcium ion (Ca2+) efflux and activating the caspase-12 signaling cascade in cancer cells. In addition, EMT-NPs induced direct oxidative stress in mitochondria; some mitochondrial-related apoptotic events such as decreased mitochondrial membrane potential (MMP), upregulation of Bax, cytochrome c release, and caspase-3 activation were also observed for tumor cells upon incubation with EMT-NPs. Furthermore, the leaked Ca2+ from ER could induce mitochondrial Ca2+ overloading to further augment cancer cell apoptosis. In brief, mitochondrial and ER signaling networks collaborated well to promote cancer cell death. Extended photoacoustic and fluorescence imaging served well for the treatment of in vivo patient-derived xenografts cancer model. This drug-free macromolecular strategy with multiple subcellular targeting provides a potential paradigm for cancer theranostics in precision nanomedicine.
Collapse
Affiliation(s)
- Wenhui Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science & Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Yongteng Zhang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Zeshu Wang
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| | - Xueping Liu
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life Science, Guangdong Provincial Key Laboratory of Laser Life Science & Guangzhou Key Laboratory of Spectral Analysis and Functional Probes, College of Biophotonics, South China Normal University, Guangzhou 510631, China
| | - Siyu Lu
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou 450000, China
| | - Xianglong Hu
- Key Laboratory of Precision and Intelligent Chemistry, University of Science and Technology of China, Hefei 230026, Anhui, China
- CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei 230026, Anhui, China
- School of Biomedical Engineering, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, Anhui, China
- Suzhou Institute for Advanced Research, University of Science and Technology of China, Suzhou 215123, China
| |
Collapse
|
20
|
Pan M, Lu C, Zhang W, Huang H, Shi X, Tang S, Liu D. Poly(l-Ornithine)-Based Polymeric Micelles as pH-Responsive Macromolecular Anticancer Agents. Pharmaceutics 2023; 15:pharmaceutics15041307. [PMID: 37111792 PMCID: PMC10143059 DOI: 10.3390/pharmaceutics15041307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Anticancer peptides and polymers represent an emerging field of tumor treatment and can physically interact with tumor cells to address the problem of multidrug resistance. In the present study, poly(l-ornithine)-b-poly(l-phenylalanine) (PLO-b-PLF) block copolypeptides were prepared and evaluated as macromolecular anticancer agents. Amphiphilic PLO-b-PLF self-assembles into nanosized polymeric micelles in aqueous solution. Cationic PLO-b-PLF micelles interact steadily with the negatively charged surfaces of cancer cells via electrostatic interactions and kill the cancer cells via membrane lysis. To alleviate the cytotoxicity of PLO-b-PLF, 1,2-dicarboxylic-cyclohexene anhydride (DCA) was anchored to the side chains of PLO via an acid-labile β-amide bond to fabricate PLO(DCA)-b-PLF. Anionic PLO(DCA)-b-PLF showed negligible hemolysis and cytotoxicity under neutral physiological conditions but recovered cytotoxicity (anticancer activity) upon charge reversal in the weakly acidic microenvironment of the tumor. PLO-based polypeptides might have potential applications in the emerging field of drug-free tumor treatment.
Collapse
Affiliation(s)
- Miao Pan
- Plastic Surgery Institute of Shantou University Medical College, Shantou 515041, China
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Shantou Plastic Surgery Clinical Research Center, Shantou 515041, China
| | - Chao Lu
- College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Wancong Zhang
- Plastic Surgery Institute of Shantou University Medical College, Shantou 515041, China
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Shantou Plastic Surgery Clinical Research Center, Shantou 515041, China
| | - Huan Huang
- Department of Pharmacy, Shantou University Medical College, Shantou 515041, China
| | - Xingyu Shi
- Department of Pharmacy, Shantou University Medical College, Shantou 515041, China
| | - Shijie Tang
- Plastic Surgery Institute of Shantou University Medical College, Shantou 515041, China
- Department of Plastic Surgery and Burn Center, Second Affiliated Hospital of Shantou University Medical College, Shantou 515041, China
- Shantou Plastic Surgery Clinical Research Center, Shantou 515041, China
| | - Daojun Liu
- Department of Pharmacy, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
21
|
Kumar N, Oqmhula K, Hongo K, Takagi K, Yusa SI, Rajan R, Matsumura K. Mechanistic insights and importance of hydrophobicity in cationic polymers for cancer therapy. J Mater Chem B 2023; 11:1456-1468. [PMID: 36661268 DOI: 10.1039/d2tb02059a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Development of molecules that can be effectively used for killing cancer cells remains a research topic of interest in drug discovery. However, various limitations of small molecules and nanotechnology-based drug-delivery systems hinder the development of chemotherapeutics. To resolve this issue, this study describes the potential application of polymeric molecules as anticancer drug candidates. We describe the design and synthesis of novel anticancer polymers containing hydrophobic groups. We established the fact that the cationic homopolymer (PAMPTMA) does not show any anticancer activity on its own; however, the insertion of hydrophobic moieties in copolymers (PAMPTMA-r-BuMA, PAMPTMA-r-HexMA, and PAMPTMA-r-OctMA) enhances their anticancer activity with a very low IC50 value (60 μg mL-1 for HepG2 cells). Mechanistic investigations were carried out using LDH leakage assay, cellular uptake, DOSY NMR and molecular dynamics to study the interaction between the polymer and the cell membrane as well as the role of hydrophobicity in enhancing this interaction. The results demonstrated that polymers are attracted by the anionic cancer cell membrane, which then leads to the insertion of hydrophobic groups inside the cell membrane, causing its disruption and ultimate lysis of the cell. This study demonstrates a novel and better approach for the rational design and discovery of new polymeric anticancer agents with improved efficacy.
Collapse
Affiliation(s)
- Nishant Kumar
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| | - Kenji Oqmhula
- School of Information Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Kenta Hongo
- Research Center for Advanced Computing Infrastructure, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan
| | - Kengo Takagi
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji, Hyogo 671-2280, Japan
| | - Shin-Ichi Yusa
- Department of Applied Chemistry, Graduate School of Engineering, University of Hyogo, 2167 Shosha, Himeji, Hyogo 671-2280, Japan
| | - Robin Rajan
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| | - Kazuaki Matsumura
- School of Materials Science, Japan Advanced Institute of Science and Technology, 1-1 Asahidai, Nomi, Ishikawa 923-1292, Japan.
| |
Collapse
|
22
|
Hu X, Li J, Zhang Y, Xiong M, Zhang H, Yuan Y. A helical oncolytic polypeptide with potent membranolytic activity for cancer therapy. Biomater Sci 2023; 11:1451-1458. [PMID: 36602031 DOI: 10.1039/d2bm01892f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Oncolytic peptides (OLPs) with membranolytic activity show great potential to combat multidrug-resistant cancer cells. Herein, we report a cationic helical oncolytic polypeptide (OLPP) with potent membranolytic activity for cancer therapy. The OLPP was synthesized by ring-opening polymerization of N-carboxyanhydrides (NCAs) and thiol-ene reaction. The OLPP was resistant to protease, showed high cytotoxicity to a series of cancer cells and caused cancer cell necrosis by quickly lysing cancer cell membrane independent of classic death-related intracellular pathways. Intra-tumoral injection of the OLPP effectively suppressed tumor growth in mice through the direct oncolytic effect. The OLPP represents a potential oncolytic chemotherapeutics for cancer therapy.
Collapse
Affiliation(s)
- Xue Hu
- School of Medicine, South China University of Technology, Guangzhou 510006, China.
| | - Jie Li
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China.
| | - Yuhao Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China.
| | - Menghua Xiong
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China. .,National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Houbing Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China. .,Key Laboratory of Biomedical Engineering of Guangdong Province, and Innovation Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, China
| | - Youyong Yuan
- School of Medicine, South China University of Technology, Guangzhou 510006, China. .,School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou 511442, China.
| |
Collapse
|
23
|
Wang C, Li F, Zhang T, Yu M, Sun Y. Recent advances in anti-multidrug resistance for nano-drug delivery system. Drug Deliv 2022; 29:1684-1697. [PMID: 35616278 PMCID: PMC9154776 DOI: 10.1080/10717544.2022.2079771] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
Chemotherapy for tumors occasionally results in drug resistance, which is the major reason for the treatment failure. Higher drug doses could improve the therapeutic effect, but higher toxicity limits the further treatment. For overcoming drug resistance, functional nano-drug delivery system (NDDS) has been explored to sensitize the anticancer drugs and decrease its side effects, which are applied in combating multidrug resistance (MDR) via a variety of mechanisms including bypassing drug efflux, controlling drug release, and disturbing metabolism. This review starts with a brief report on the major MDR causes. Furthermore, we searched the papers from NDDS and introduced the recent advances in sensitizing the chemotherapeutic drugs against MDR tumors. Finally, we concluded that the NDDS was based on several mechanisms, and we looked forward to the future in this field.
Collapse
Affiliation(s)
- Changduo Wang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Fashun Li
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Tianao Zhang
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Min Yu
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| | - Yong Sun
- Department of Pharmaceutics, School of Pharmacy, Qingdao University, Qingdao, China
| |
Collapse
|
24
|
Takahashi H, Sovadinova I, Yasuhara K, Vemparala S, Caputo GA, Kuroda K. Biomimetic antimicrobial polymers—Design, characterization, antimicrobial, and novel applications. WIRES NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 15:e1866. [PMID: 36300561 DOI: 10.1002/wnan.1866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 09/15/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022]
Abstract
Biomimetic antimicrobial polymers have been an area of great interest as the need for novel antimicrobial compounds grows due to the development of resistance. These polymers were designed and developed to mimic naturally occurring antimicrobial peptides in both physicochemical composition and mechanism of action. These antimicrobial peptide mimetic polymers have been extensively investigated using chemical, biophysical, microbiological, and computational approaches to gain a deeper understanding of the molecular interactions that drive function. These studies have helped inform SARs, mechanism of action, and general physicochemical factors that influence the activity and properties of antimicrobial polymers. However, there are still lingering questions in this field regarding 3D structural patterning, bioavailability, and applicability to alternative targets. In this review, we present a perspective on the development and characterization of several antimicrobial polymers and discuss novel applications of these molecules emerging in the field. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Therapeutic Approaches and Drug Discovery > Nanomedicine for Infectious Disease.
Collapse
Affiliation(s)
- Haruko Takahashi
- Graduate School of Integrated Sciences for Life Hiroshima University Higashi‐Hiroshima Hiroshima Japan
| | - Iva Sovadinova
- RECETOX, Faculty of Science Masaryk University Brno Czech Republic
| | - Kazuma Yasuhara
- Division of Materials Science, Graduate School of Science and Technology Nara Institute of Science and Technology Nara Japan
- Center for Digital Green‐Innovation Nara Institute of Science and Technology Nara Japan
| | - Satyavani Vemparala
- The Institute of Mathematical Sciences CIT Campus Chennai India
- Homi Bhabha National Institute Training School Complex Mumbai India
| | - Gregory A. Caputo
- Department of Chemistry & Biochemistry Rowan University Glassboro New Jersey USA
| | - Kenichi Kuroda
- Department of Biologic and Materials Sciences & Prosthodontics, School of Dentistry University of Michigan Ann Arbor Michigan USA
| |
Collapse
|
25
|
Guo J, Du X, Huang J, Liu C, Zhou Y, Li Y, Du B. Robust Dual Enzyme Cascade-Catalytic Cholesterol Depletion for Reverse Tumor Multidrug Resistance. Adv Healthc Mater 2022; 11:e2200859. [PMID: 35906730 DOI: 10.1002/adhm.202200859] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/30/2022] [Indexed: 01/27/2023]
Abstract
Although combination drugs and P-glycoprotein inhibitors are the main methods to solve multidrug resistance, these methods ignore the pathological structure of drug-resistant cells and extremely limit curative effect. Herein, a new paradigm of reversing multidrug resistance with abnormal expression of cholesterol as the target is proposed, which uses the cascade catalysis of "natural enzyme" cholesterol oxidase (COD) and "nanoenzyme" Cu2+ -modified zirconium-based metal-organic framework (ZrMOF(Cu)) to convert cholesterol into the highly cytotoxic hydroxyl radicals. The doxorubicin (DOX)-loaded nanoparticles (DOX@COD-MOF) can significantly reduce the cholesterol content of cancer cells via COD, which decrease the rigidity of drug resistant cancer cell membranes and restore the sensitivity of multidrug-resistant cells to DOX. Afterward, DOX@COD-MOF is encapsulated by cancer cell membranes (CCM) to construct a bionic "dual enzyme catalytic cascade nanoreactor" (DOX@COD-MOF@CCM). Such a rational design presents a preferential accumulation tendency to tumor sites due to the homologous targeting mechanism of CCM, and affords 94.4% in tumor growth suppression without systemic toxicity in vivo. This work aims to achieve the therapeutic purpose of high efficiency and low toxicity. It has the characteristics of "converting enemy into friend, " and opens up a promising way for effectively reversing multidrug resistance of tumors.
Collapse
Affiliation(s)
- Jialing Guo
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xiaoming Du
- Key Laboratory of Photochemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
| | - Jingshu Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chenxin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yingying Zhou
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Ying Li
- Department of Pharmacy, Zhengzhou Central Hospital Affiliated to Zhengzhou University, Zhengzhou, 450007, China
| | - Bin Du
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.,Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou, Henan Province, 450001, China
| |
Collapse
|
26
|
Raj W, Jerczynski K, Rahimi M, Pavlova E, Šlouf M, Przekora A, Pietrasik J. Stimuli-responsive vitamin E-based micelles: Effective drug carriers with a controlled anticancer drug release. POLYMER 2022. [DOI: 10.1016/j.polymer.2022.125001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
27
|
Tan J, Zhao Y, Hedrick JL, Yang YY. Effects of Hydrophobicity on Antimicrobial Activity, Selectivity, and Functional Mechanism of Guanidinium-Functionalized Polymers. Adv Healthc Mater 2022; 11:e2100482. [PMID: 33987953 DOI: 10.1002/adhm.202100482] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 04/21/2021] [Indexed: 11/06/2022]
Abstract
In this study, a series of guanidinium-functionalized polycarbonate random co-polymers is prepared from organocatalytic ring-opening polymerization to investigate the effect of the hydrophobic side chain (ethyl, propyl, isopropyl, benzyl, and hexyl) on their antimicrobial activity and selectivity. Although the polymers exhibit similar minimum inhibitory concentrations, the more hydrophobic polymers exhibit a faster rate of bacteria elimination. At higher percentage content (20 mol%), polymers with more hydrophobic side chains suffer from poor selectivity due to their high hemolytic activity. The highly hydrophobic co-polymer, containing the hydrophobic hexyl-functionalized cyclic carbonate, kills bacteria via a membrane-disruptive mechanism. Micelle formation leads to a lower extent of membrane disruption. This study unravels the effects of hydrophobic side chains on the activities of the polymers and their killing mechanism, providing insights into the design of new antimicrobial polymers.
Collapse
Affiliation(s)
- Jason Tan
- Institute of Bioengineering and Bioimaging 31 Biopolis Way Singapore 138669 Singapore
- Division of Chemistry and Biological Chemistry School of Physical and Mathematical Sciences Nanyang Technological University 21 Nanyang Link Singapore 637371 Singapore
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry School of Physical and Mathematical Sciences Nanyang Technological University 21 Nanyang Link Singapore 637371 Singapore
| | - James L. Hedrick
- IBM Almaden Research Center 650 Harry Road San Jose CA 95120 USA
| | - Yi Yan Yang
- Institute of Bioengineering and Bioimaging 31 Biopolis Way Singapore 138669 Singapore
| |
Collapse
|
28
|
Yang S, Leong J, Wang Y, Sim R, Tan KH, Chua YH, Tan N, Lee ALZ, Tay J, Yang YY. Drug-free neutrally charged polypeptide nanoparticles as anticancer agents. J Control Release 2022; 345:464-474. [PMID: 35331785 DOI: 10.1016/j.jconrel.2022.03.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 01/11/2023]
Abstract
Cationic synthetic anticancer polymers and peptides have attracted increasing attention for advancing cancer treatment without causing drug resistance development. To circumvent in vivo instability and toxicity caused by cationic charges of the anticancer polymers/peptides, we report, for the first time, a nanoparticulate delivery system self-assembled from a negatively charged pH-sensitive polypeptide poly(ethylene glycol)-b-poly(ʟ-lysine)-graft-cyclohexene-1,2-dicarboxylic anhydride and a cationic anticancer polypeptide guanidinium-functionalized poly(ʟ-lysine) (PLL-Gua) via electrostatic interaction. The formation of nanoparticles (Gua-NPs) neutralized the positive charges of PLL-Gua. Both PLL-Gua and Gua-NPs killed cancer cells in a dose- and time-dependent manner, and induced cell death via apoptosis. Confocal microscopic studies demonstrated that PLL-Gua and Gua-NPs readily entered cancer cells, and Gua-NPs were taken up by the cells via endocytosis. Notably, Gua-NPs and PLL-Gua exhibited similar in vitro anticancer efficacy against MCF-7 and resistant MCF-7/ADR. PLL-Gua and Gua-NPs also induced similar morphological changes in MCF-7/ADR cells compared to MCF-7 cells, further indicating their ability to bypass drug resistance mechanisms in the MCF-7/ADR cells. More importantly, Gua-NPs with higher LD50 and enhanced tumor accumulation significantly inhibited tumor growth with negligible side effects in vivo. Our findings shed light on the in vivo delivery of anticancer peptides and opened a new avenue for cancer treatment.
Collapse
Affiliation(s)
- Shengcai Yang
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Jiayu Leong
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Yanming Wang
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Rachel Sim
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Ko Hui Tan
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Yau Hong Chua
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Nathanael Tan
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore; School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Ashlynn L Z Lee
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Joyce Tay
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore
| | - Yi Yan Yang
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, Singapore 138669, Singapore; Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119288, Singapore.
| |
Collapse
|
29
|
Hedrick JL, Piunova V, Park NH, Erdmann T, Arrechea PL. Simple and Efficient Synthesis of Functionalized Cyclic Carbonate Monomers Using Carbon Dioxide. ACS Macro Lett 2022; 11:368-375. [PMID: 35575375 DOI: 10.1021/acsmacrolett.2c00060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Aliphatic polycarbonates represent an important class of materials with diverse applications ranging from battery electrolytes, polyurethane intermediates, and materials for biomedical applications. These materials can be produced via the ring-opening polymerization (ROP) of six- to eight-membered cyclic carbonates derived from precursor 1,3- and 1,5-diols. These diols can contain a range of functional groups depending on the desired thermal, mechanical, and solution properties. Generally, the ring closure to form the cyclic carbonate requires the use of undesirable and hazardous reagents. Advances in synthetic methodologies and catalysis have enabled the use of carbon dioxide (CO2) to perform these transformations with a high conversion of diol to cyclic carbonate, yet modest isolated yields due to oligomerization side reactions. In this Letter, we evaluate a series of bases in the presence of p-toluenesulfonyl chloride and the appropriate diol to better understand their effect on the yield and presence of oligomer byproducts during cyclic carbonate formation from CO2. From this study, N,N-tetramethylethylenediamine (TMEDA) was identified as an optimal base, facilitating the preparation of a diverse array of both six- and eight-membered carbonates from CO2 within 10 to 15 min. The robust conditions for both, the preparation of the diol precursor, and the TMEDA-mediated carbonate synthesis enabled readily telescoping the two-step reaction sequence, greatly simplifying the process of monomer preparation.
Collapse
Affiliation(s)
- James L. Hedrick
- IBM Almaden Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Victoria Piunova
- IBM Almaden Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Nathaniel H. Park
- IBM Almaden Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Tim Erdmann
- IBM Almaden Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Pedro L. Arrechea
- IBM Almaden Research Center, 650 Harry Road, San Jose, California 95120, United States
| |
Collapse
|
30
|
Jung K, Corrigan N, Wong EHH, Boyer C. Bioactive Synthetic Polymers. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105063. [PMID: 34611948 DOI: 10.1002/adma.202105063] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/13/2021] [Indexed: 05/21/2023]
Abstract
Synthetic polymers are omnipresent in society as textiles and packaging materials, in construction and medicine, among many other important applications. Alternatively, natural polymers play a crucial role in sustaining life and allowing organisms to adapt to their environments by performing key biological functions such as molecular recognition and transmission of genetic information. In general, the synthetic and natural polymer worlds are completely separated due to the inability for synthetic polymers to perform specific biological functions; in some cases, synthetic polymers cause uncontrolled and unwanted biological responses. However, owing to the advancement of synthetic polymerization techniques in recent years, new synthetic polymers have emerged that provide specific biological functions such as targeted molecular recognition of peptides, or present antiviral, anticancer, and antimicrobial activities. In this review, the emergence of this generation of bioactive synthetic polymers and their bioapplications are summarized. Finally, the future opportunities in this area are discussed.
Collapse
Affiliation(s)
- Kenward Jung
- Cluster for Advanced Macromolecular Design (CAMD), Australian Centre for Nanomedicine (ACN), and School of Chemical Engineering, University of New South Wales (UNSW) Sydney, Sydney, NSW, 2052, Australia
| | - Nathaniel Corrigan
- Cluster for Advanced Macromolecular Design (CAMD), Australian Centre for Nanomedicine (ACN), and School of Chemical Engineering, University of New South Wales (UNSW) Sydney, Sydney, NSW, 2052, Australia
| | - Edgar H H Wong
- Cluster for Advanced Macromolecular Design (CAMD), Australian Centre for Nanomedicine (ACN), and School of Chemical Engineering, University of New South Wales (UNSW) Sydney, Sydney, NSW, 2052, Australia
| | - Cyrille Boyer
- Cluster for Advanced Macromolecular Design (CAMD), Australian Centre for Nanomedicine (ACN), and School of Chemical Engineering, University of New South Wales (UNSW) Sydney, Sydney, NSW, 2052, Australia
| |
Collapse
|
31
|
Kumar N, Fazal S, Miyako E, Matsumura K, Rajan R. Avengers against cancer: A new era of nano-biomaterial-based therapeutics. MATERIALS TODAY 2021; 51:317-349. [DOI: 10.1016/j.mattod.2021.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
32
|
Cheng X, Li D, Xu J, Wei B, Fang Q, Yang L, Xue Y, Wang X, Tang R. Self-assembled ternary hybrid nanodrugs for overcoming tumor resistance and metastasis. Acta Pharm Sin B 2021; 11:3595-3607. [PMID: 34900539 PMCID: PMC8642601 DOI: 10.1016/j.apsb.2021.03.041] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 02/07/2023] Open
Abstract
Traditional chemotherapy exhibits a certain therapeutic effect toward malignant cancer, but easily induce tumor multidrug resistance (MDR), thereby resulting in the progress of tumor recurrence or metastasis. In this work, we deigned ternary hybrid nanodrugs (PEI/DOX@CXB-NPs) to simultaneously combat against tumor MDR and metastasis. In vitro results demonstrate this hybrid nanodrugs could efficiently increase cellular uptake at pH 6.8 by the charge reversal, break lysosomal sequestration by the proton sponge effect and trigger drugs release by intracellular GSH, eventually leading to higher drugs accumulation and cell-killing in drug-sensitive/resistant cells. In vivo evaluation revealed that this nanodrugs could significantly inhibit MDR tumor growth and simultaneously prevent A549 tumor liver/lung metastasis owing to the specifically drugs accumulation. Mechanism studies further verified that hybrid nanodrugs were capable of down-regulating the expression of MDR or metastasis-associated proteins, lead to the enhanced anti-MDR and anti-metastasis effect. As a result, the multiple combination strategy provided an option for effective cancer treatment, which could be potentially extended to other therapeutic agents or further use in clinical test.
Collapse
|
33
|
Tay J, Zhao Y, Hedrick JL, Yang YY. Elucidating the anticancer activities of guanidinium-functionalized amphiphilic random copolymers by varying the structure and composition in the hydrophobic monomer. Theranostics 2021; 11:8977-8992. [PMID: 34522222 PMCID: PMC8419055 DOI: 10.7150/thno.60711] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 07/01/2021] [Indexed: 01/14/2023] Open
Abstract
Rationale: Use of traditional anticancer chemotherapeutics has been hindered by the multifactorial nature of multi-drug resistance (MDR) development and metastasis. Recently, cationic polycarbonates were reported as novel unconventional anticancer agents that mitigated MDR and inhibited metastasis. The aim of this study is to explore structure-anticancer activity relationship. Specifically, a series of cationic guanidinium-based random copolymers of varying hydrophobicity was synthesized with a narrow polydispersity (Ð = 1.12-1.27) via organocatalytic ring-opening polymerization (OROP) of functional cyclic carbonate monomers, and evaluated for anticancer activity, killing kinetics, degradability and functional mechanism. Methods: Linear, branched and aromatic hydrophobic side chain units, such as ethyl, benzyl, butyl, isobutyl and hexyl moieties were explored as comonomer units for modulating anticancer activity. As hydrophobicity/hydrophilicity balance of the polymers determines their anticancer efficacy, the feed ratio between the two monomers was varied to tune their hydrophobicity. Results: Notably, incorporating the hexyl moiety greatly enhanced anticancer efficiency and killing kinetics on cancer cells. Degradation studies showed that the polymers degraded completely within 4-6 days. Flow cytometry and lactate dehydrogenase (LDH) release analyses demonstrated that anticancer mechanism of the copolymers containing a hydrophobic co-monomer was concentration dependent, apoptosis at IC50, and both apoptosis and necrosis at 2 × IC50. In contrast, the homopolymer without a hydrophobic comonomer killed cancer cells predominantly via apoptotic mechanism. Conclusion: The hydrophobicity of the polymers played an important role in anticancer efficacy, killing kinetics and anticancer mechanism. This study provides valuable insights into designing novel anticancer agents utilizing polymers.
Collapse
Affiliation(s)
- Joyce Tay
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Yanli Zhao
- Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - James L. Hedrick
- IBM Almaden Research Center, 650 Harry Road, San Jose, California 95120, United States
| | - Yi Yan Yang
- Institute of Bioengineering and Bioimaging, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| |
Collapse
|
34
|
Liang L, Peng Y, Qiu L. Mitochondria-targeted vitamin E succinate delivery for reversal of multidrug resistance. J Control Release 2021; 337:117-131. [PMID: 34274383 DOI: 10.1016/j.jconrel.2021.07.023] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 07/11/2021] [Accepted: 07/14/2021] [Indexed: 12/31/2022]
Abstract
Inducing mitochondrial malfunction is an appealing strategy to overcome tumor multidrug resistance (MDR). Reported here a versatile mitochondrial-damaging molecule, vitamin E succinate (VES), is creatively utilized to assist MDR reversal of doxorubicin hydrochloride (DOX·HCl) via a nanovesicle platform self-assembled from amphiphilic polyphosphazenes containing pH-sensitive 1H-benzo-[d]imidazol-2-yl) methanamine (BIMA) groups. Driven by multiple non-covalent interactions, VES is fully introduced into the hydrophobic membrane of DOX·HCl-loaded nanovesicles with loading content of 23.5%. The incorporated VES also offers robust anti-leakage property toward DOX·HCl under normal physiological conditions. More importantly, upon release within acidic tumor cells, VES can target mitochondria and result in various dysfunctions including excessive generation of reactive oxygen species (ROS), mitochondrial membrane potential (ΔΨm) loss, and inhibited adenosine triphosphate (ATP) synthesis, which contribute to cell apoptosis and insufficient energy supply for drug efflux pumps. Consequently, the killing-effect of DOX·HCl is significantly enhanced toward drug resistant cancer cells at the optimal mass ratio of DOX·HCl to VES. Further in vivo antitumor investigation on nude mice bearing xenograft drug-resistant human chronic myelogenous leukemia K562/ADR tumors verifies the extremely enhanced anti-tumor efficacy of the dual drug-loaded nanovesicle with the tumor inhibition rate (TIR) of 82.38%. Collectively, this study provides a s safe, facile and promising strategy for both precise drug delivery and MDR eradication to improve cancer therapy.
Collapse
Affiliation(s)
- Lina Liang
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Yan Peng
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China
| | - Liyan Qiu
- Ministry of Education (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, China.
| |
Collapse
|
35
|
Ansari I, Singh P, Mittal A, Mahato RI, Chitkara D. 2,2-Bis(hydroxymethyl) propionic acid based cyclic carbonate monomers and their (co)polymers as advanced materials for biomedical applications. Biomaterials 2021; 275:120953. [PMID: 34218051 DOI: 10.1016/j.biomaterials.2021.120953] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 12/15/2022]
Abstract
Designing grafted biodegradable polymers with tailored multi-functional properties is one of the most researched fields with extensive biomedical applications. Among many biodegradable polymers, polycarbonates have gained much attention due to their ease of synthesis, high drug loading, and excellent biocompatibility profiles. Among various monomers, 2,2-bis(hydroxymethyl) propionic acid (bis-MPA) derived cyclic carbonate monomers have been extensively explored in terms of their synthesis as well as their polymerization. Since the late 90s, significant advancements have been made in the design of bis-MPA derived cyclic carbonate monomers as well as in their reaction schemes. Currently, bis-MPA derived polycarbonates have taken a form of an entire platform with a multitude of applications, the latest being in the field of nanotechnology, targeted drug, and nucleic acid delivery. The present review outlines an up to date developments that have taken place in the last two decades in the design, synthesis, and biomedical applications of bis-MPA derived cyclic carbonates and their (co)polymers.
Collapse
Affiliation(s)
- Imran Ansari
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Vidya Vihar Campus, Pilani, 333 031, Rajasthan, India
| | - Prabhjeet Singh
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Vidya Vihar Campus, Pilani, 333 031, Rajasthan, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Vidya Vihar Campus, Pilani, 333 031, Rajasthan, India
| | - Ram I Mahato
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani (BITS Pilani), Vidya Vihar Campus, Pilani, 333 031, Rajasthan, India.
| |
Collapse
|
36
|
Leong J, Yang C, Tan J, Tan BQ, Hor S, Hedrick JL, Yang YY. Combination of guanidinium and quaternary ammonium polymers with distinctive antimicrobial mechanisms achieving a synergistic antimicrobial effect. Biomater Sci 2021; 8:6920-6929. [PMID: 32959808 DOI: 10.1039/d0bm00752h] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
The increasing emergence and spread of antimicrobial resistance are urgent and important global challenges today. The clinical pipeline is lacking in innovative drugs that avoid the development of drug resistance. Macromolecular antimicrobials kill bacteria and fungi through physical disruptions to the cell membrane, which is difficult for microbes to overcome. Recently, we reported antimicrobial polycarbonates that kill microbes via two different mechanisms. Polycarbonates functionalized with quaternary ammonium disrupted the lipid bilayer membrane of the microbes, while polycarbonates functionalized with guanidinium translocated the membrane and precipitated cytosolic components. We hypothesized that the combination of these two distinct mechanisms would result in a more than additive increase in antimicrobial efficacy. Block and random copolymers containing both cationic groups had similar minimum inhibitory concentrations (MICs) as the guanidinium homopolymer on 5 representatives of the ESKAPE pathogens. Interestingly, the random copolymer killed P. aeruginosa and A. baumannii more rapidly than the block copolymer and the guanidinium homopolymer with the same number of guanidinium groups. Like quaternary ammonium homopolymer, the copolymers killed the bacteria via a membrane-disruptive mechanism. Then, we simply mixed quaternary ammonium homopolymer and guanidinium homopolymer, and studied antimicrobial activity of the combination at various concentrations. Checkerboard assay results showed that the combination of the polymers, in general, achieved a synergistic or additive effect in inhibiting the growth of bacteria. At concentrations where it exibited a synergistic or additive effect in inhibiting bacterial growth, the combination killed the bacteria effectively (99%-99.9% killing efficiency) although the individual polymers at these concentrations did not exert bactericidal activity. Therefore, it is essential to have the two functional groups on separate molecules to provide synergism. This study provides a basic understanding of polymer design with different cationic groups.
Collapse
Affiliation(s)
- Jiayu Leong
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore 138669, Singapore.
| | | | | | | | | | | | | |
Collapse
|
37
|
Fan F, Jin L, Yang L. pH-Sensitive Nanoparticles Composed Solely of Membrane-Disruptive Macromolecules for Treating Pancreatic Cancer. ACS APPLIED MATERIALS & INTERFACES 2021; 13:12824-12835. [PMID: 33689289 DOI: 10.1021/acsami.0c16576] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Pancreatic tumor is extremely lethal because its cancerous structures are sheltered by dense stromal barriers that hinder the infiltration of therapeutics. To facilitate the infiltration of therapeutics through the stromal barrier, remodeling the stroma with an adjuvant prior to or together with gemcitabine-the current chemotherapeutic standard for pancreatic cancer-is a widely studied strategy; nevertheless, the intrinsic nonuniformity in distribution (spatial and/or temporal) of the adjuvant and gemcitabine has raised the increased risk of tumor metastasis as a major concern. In this work, we propose long-circulating, pH-sensitive nanoparticles composed solely of cellular membrane-disruptive molecules as a new approach for treating pancreatic cancer. Using a micelle of a polymeric mimetic of host defense peptides as the model for such nanoparticles, we showed that this nanoparticle exhibited acid-activated cytotoxicity indiscriminately to both cancerous and fibroblast cells, and the underlying activity mode was acid-activatable disruption of cellular membrane integrity. As a result, our acid-activatable nanoparticle effectively permeabilized the stromal barrier and eradicated the otherwise sheltered pancreatic cancer cells, as demonstrated with a three-dimensional spheroid in which a shell of fibroblast NIH-3T3 cells was cultured over a core of pancreatic BxPC-3 cells. When administered intravenously into mouse models bearing xenograft pancreatic BxPC-3 tumors, our acid-activatable nanoparticle efficiently inhibited tumor growth without causing noticeable off-target adverse effects or promoting tumor metastasis. Notably, this nanoparticle permeabilized the otherwise dense pancreatic tumor tissue while significantly suppressing the expression of extracellular matrix components and activated cancer-associated fibroblasts. Although the feasibility of our approach was demonstrated with a micelle of a polymeric molecule, we trust that future research efforts in this pathway may eventually offer translational formulations for improving the therapeutic efficacy of pancreatic cancer.
Collapse
Affiliation(s)
- Feng Fan
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Soft Matter Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Lijun Jin
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Soft Matter Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Lihua Yang
- Hefei National Laboratory for Physical Sciences at the Microscale, CAS Key Laboratory of Soft Matter Chemistry, School of Chemistry and Materials Science, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
38
|
Zheng Y, Kng J, Yang C, Hedrick JL, Yang YY. Cationic polymer synergizing with chemotherapeutics and re-purposing antibiotics against cancer cells. Biomater Sci 2021; 9:2174-2182. [PMID: 33502409 DOI: 10.1039/d0bm02155e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Chemotherapy is one of the most effective treatments for cancer. However, toxicity and the development of drug resistance have become the major hurdles to the commonly used chemotherapeutics such as doxorubicin and paclitaxel. Antibiotics have also been used as anti-cancer drugs due to their anti-proliferative and cytotoxic effects. However, these anti-tumor antibiotics like ciprofloxacin face the similar resistance and toxicity issues. In this study, we used a quaternary ammonium-functionalized cationic polycarbonate to synergize with the existing chemotherapeutics and re-purpose antibiotics to address the resistance and toxicity issues. When used in combination with the drugs, the cationic polymer induced 2-3 fold more damage in the cancer cell membrane within 2 hours, thus enhancing the uptake of chemotherapeutics up to 2.5 fold more into the breast, liver and even chemotherapeutics-resistant cancer cells. On the other hand, the chemotherapeutics increased the cellular uptake of polymer. The combined effects resulted in 3-10 fold reduction in IC50 of chemotherapy drugs and yielded therapeutic synergy at a clinically-relevant concentration range of drugs when treating multiple types of cancer cells, while the use of guanidinium-functionalized polymer capable of membrane translocation did not lead to a synergistic effect. Thus, the quaternary ammonium-functionalized cationic polymer can increase the therapeutic efficacies of existing drugs, mitigating toxicities by lowering required dosage and circumventing drug resistance via its membrane disruption mechanism. The findings of this study provide insights into designing future anticancer therapy.
Collapse
Affiliation(s)
- Yiran Zheng
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, P. R. China.
| | | | | | | | | |
Collapse
|
39
|
Yu W, Maynard E, Chiaradia V, Arno MC, Dove AP. Aliphatic Polycarbonates from Cyclic Carbonate Monomers and Their Application as Biomaterials. Chem Rev 2021; 121:10865-10907. [DOI: 10.1021/acs.chemrev.0c00883] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Wei Yu
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Edward Maynard
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Viviane Chiaradia
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Maria C. Arno
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
- Institute of Cancer and Genomic Sciences, University of Birmingham, Edgbaston, B15 2TT U.K
| | - Andrew P. Dove
- School of Chemistry, University of Birmingham, Edgbaston, B15 2TT U.K
| |
Collapse
|
40
|
Tomás RMF, Gibson MI. Covalent cell surface recruitment of chemotherapeutic polymers enhances selectivity and activity. Chem Sci 2021; 12:4557-4569. [PMID: 34163721 PMCID: PMC8179505 DOI: 10.1039/d0sc06580c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/12/2021] [Indexed: 01/22/2023] Open
Abstract
Synthetic macromolecular chemotherapeutics inspired by host defence peptides can disrupt cell membranes and are emerging as agents for the treatment of cancer and infections. However, their off-target effects remain a major unmet challenge. Here we introduce a covalent recruitment strategy, whereby metabolic oligosaccharide engineering is used to label targeted cells with azido glycans, to subsequently capture chemotherapeutic polymers by a bio-orthogonal click reaction. This results in up to 10-fold reduction in EC50 and widening of the therapeutic window. Cell death is induced by not only membrane leakage, but also by apoptosis due to the conjugated chemotherapeutic being internalised by glycan recycling. Covalent recruitment also lead to increased penetration and significant cell death in a 3-D tumour model in just 3 hours, whereas doxorubicin required 24 hours. This conceptual approach of 'engineering cells to capture polymers' rather than 'engineering polymers to target cells' will bring new opportunities in non-traditional macromolecular therapeutics.
Collapse
Affiliation(s)
- Ruben M F Tomás
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
- MAS CDT, University of Warwick Coventry CV4 7AL UK
| | - Matthew I Gibson
- Department of Chemistry, University of Warwick Coventry CV4 7AL UK
- Warwick Medical School, University of Warwick Coventry CV4 7AL UK
| |
Collapse
|
41
|
Tan EWP, Hedrick JL, Arrechea PL, Erdmann T, Kiyek V, Lottier S, Yang YY, Park NH. Overcoming Barriers in Polycarbonate Synthesis: A Streamlined Approach for the Synthesis of Cyclic Carbonate Monomers. Macromolecules 2021. [DOI: 10.1021/acs.macromol.0c02880] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Eddy W. P. Tan
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore 138669, Singapore
| | - James L. Hedrick
- IBM Research-Almaden, 650 Harry Road, San Jose, California 95120, United States
| | - Pedro L. Arrechea
- IBM Research-Almaden, 650 Harry Road, San Jose, California 95120, United States
| | - Tim Erdmann
- IBM Research-Almaden, 650 Harry Road, San Jose, California 95120, United States
| | - Vivien Kiyek
- IBM Research-Almaden, 650 Harry Road, San Jose, California 95120, United States
| | - Simon Lottier
- IBM Research-Almaden, 650 Harry Road, San Jose, California 95120, United States
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore 138669, Singapore
| | - Nathaniel H. Park
- IBM Research-Almaden, 650 Harry Road, San Jose, California 95120, United States
| |
Collapse
|
42
|
Alven S, Aderibigbe BA. The Therapeutic Efficacy of Dendrimer and Micelle Formulations for Breast Cancer Treatment. Pharmaceutics 2020; 12:E1212. [PMID: 33333778 PMCID: PMC7765183 DOI: 10.3390/pharmaceutics12121212] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/16/2020] [Accepted: 10/16/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is among the most common types of cancer in women and it is the cause of a high rate of mortality globally. The use of anticancer drugs is the standard treatment approach used for this type of cancer. However, most of these drugs are limited by multi-drug resistance, drug toxicity, poor drug bioavailability, low water solubility, poor pharmacokinetics, etc. To overcome multi-drug resistance, combinations of two or more anticancer drugs are used. However, the combination of two or more anticancer drugs produce toxic side effects. Micelles and dendrimers are promising drug delivery systems that can overcome the limitations associated with the currently used anticancer drugs. They have the capability to overcome drug resistance, reduce drug toxicity, improve the drug solubility and bioavailability. Different classes of anticancer drugs have been loaded into micelles and dendrimers, resulting in targeted drug delivery, sustained drug release mechanism, increased cellular uptake, reduced toxic side effects of the loaded drugs with enhanced anticancer activity in vitro and in vivo. This review article reports the biological outcomes of dendrimers and micelles loaded with different known anticancer agents on breast cancer in vitro and in vivo.
Collapse
Affiliation(s)
| | - Blessing Atim Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Eastern Cape 5700, South Africa;
| |
Collapse
|
43
|
Lee ALZ, Yang C, Gao S, Wang Y, Hedrick JL, Yang YY. Biodegradable Cationic Polycarbonates as Vaccine Adjuvants. ACS APPLIED MATERIALS & INTERFACES 2020; 12:52285-52297. [PMID: 33179910 DOI: 10.1021/acsami.0c09649] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In this study, biodegradable cationic polycarbonate and polylactide block copolymers were synthesized and successfully used as novel vaccine adjuvants to provide enhanced anticancer immunity. The polymers formed nanoparticles with the model vaccine, ovalbumin (OVA), and the immunostimulant toll-like receptor 3 agonist poly(I:C) (a synthetic analog of the double-stranded RNA). Higher uptake of poly(I:C) by the bone marrow-derived dendritic cells and macrophages and OVA by dendritic cells was observed when delivered using the polymer adjuvant. In vivo experiments showed that these nanoparticles remained longer in the subcutaneous injection site as compared to OVA alone and led to higher production of anti-OVA specific antibodies with prolonged immunostimulation. When OVA was combined with poly(I:C) that was either co-entrapped in the same particles or as separate particles, a comparable level of anti-OVA IgG1 antibodies and interleukin-6 (IL-6) was produced in mouse blood plasma, and a similar level of cytotoxic T lymphocyte (CTL) response in mice was stimulated as compared to OVA/Alum particles. Furthermore, tumor rejection in the mice that were vaccinated for 9 months with the formulations containing the polymer adjuvant was stronger than the other treatment groups without the polymer. Notably, the cationic polycarbonates were not associated with any adverse in vivo effects. Thus, these biodegradable polymers may be promising substitutes for aluminum-based adjuvants in vaccine formulations.
Collapse
Affiliation(s)
- Ashlynn L Z Lee
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Chuan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - Shujun Gao
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
- NanoBio Lab, 31 Biopolis Way, #09-01 The Nanos, Singapore 138669, Singapore
| | - Yanming Wang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | - James L Hedrick
- IBM Almaden Research Center, 650 Harry Road, San Jose, California 95120 United States
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| |
Collapse
|
44
|
Shen W, Zhang Y, Wan P, An L, Zhang P, Xiao C, Chen X. Antineoplastic Drug-Free Anticancer Strategy Enabled by Host-Defense-Peptides-Mimicking Synthetic Polypeptides. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e2001108. [PMID: 32700437 DOI: 10.1002/adma.202001108] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/22/2020] [Indexed: 06/11/2023]
Abstract
An antineoplastic drug-free anticancer strategy enabled by host defense peptides (HDPs)-mimicking synthetic polypeptides is reported. The polypeptide exhibits a broad spectrum of anticancer activity in 12 cancer cell lines, including drug-resistant and highly metastatic tumor cells. Detailed mechanistic studies reveal that the cationic anticancer polypeptide (ACPP) can directly induce rapid necrosis of cancer cells within minutes through a membrane-lytic mechanism. Moreover, a pH-sensitive zwitterionic derivative of ACPP (DA-ACPP) is prepared for in vivo application. DA-ACPP shows negligible hemolysis under neutral physiological conditions, and can be converted back to ACPP in slightly acidic tumor environments, resulting in selective killing of cancer cells. Consequently, DA-ACPP shows an effective inhibition of tumor growth in both 4T1 orthotopic breast tumor models and B16-F10 melanoma pulmonary metastatic models. Overall, these findings demonstrate that synthetic HDPs-mimicking polypeptides represent safe and effective antineoplastic agents, which sheds new light on the development of drug-free synthetic polymers for cancer therapy.
Collapse
Affiliation(s)
- Wei Shen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yu Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Pengqi Wan
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- Department of Chemistry, Northeast Normal University, Changchun, Jilin, 130022, P. R. China
| | - Lin An
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- Department of Dermatology, China-Japan Union Hospital, Jilin University, Changchun, Jilin, 130033, P. R. China
| | - Peng Zhang
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| | - Chunsheng Xiao
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| | - Xuesi Chen
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
- Jilin Biomedical Polymers Engineering Laboratory, Changchun, 130022, P. R. China
| |
Collapse
|
45
|
Hou X, Shou C, He M, Xu J, Cheng Y, Yuan Z, Lan M, Zhao Y, Yang Y, Chen X, Gao F. A combination of LightOn gene expression system and tumor microenvironment-responsive nanoparticle delivery system for targeted breast cancer therapy. Acta Pharm Sin B 2020; 10:1741-1753. [PMID: 33088693 PMCID: PMC7564032 DOI: 10.1016/j.apsb.2020.04.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/11/2020] [Accepted: 03/27/2020] [Indexed: 01/02/2023] Open
Abstract
A light-switchable transgene system called LightOn gene expression system could regulate gene expression with a high on/off ratio under blue light, and have great potential for spatiotemporally controllable gene expression. We developed a nanoparticle drug delivery system (NDDS) to achieve tumor microenvironment-responsive and targeted delivery of diphtheria toxin A (DTA) fragment-encoded plasmids to tumor sites. The expression of DTA was induced by exposure to blue light. Nanoparticles composed of polyethylenimine and vitamin E succinate linked by a disulfide bond, and PEGylated hyaluronic acid modified with RGD peptide, accumulated in tumor tissues and were actively internalized into 4T1 cells via dual targeting to CD44 and αvβ3 receptors. The LightOn gene expression system was able to control target protein expression through regulation of the intensity or duration of blue light exposure. In vitro studies showed that light-induced DTA expression reduced 4T1 cell viability and induced apoptosis. Furthermore, the LightOn gene expression system enabled spatiotemporal control of the expression of DTA in a mouse 4T1 tumor xenograft model, which resulted in excellent antitumor effects, reduced tumor angiogenesis, and no systemic toxicity. The combination of the LightOn gene expression system and NDDS may be an effective strategy for treatment of breast cancer.
Collapse
Affiliation(s)
- Xinyu Hou
- Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Chenting Shou
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Muye He
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Jiajun Xu
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yi Cheng
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zeting Yuan
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200062, China
| | - Minbo Lan
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
| | - Yuzheng Zhao
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
- Optogenetics & Molecular Imaging Interdisciplinary Research Center, CAS Center for Excellence in Brain Science, East China University of Science and Technology, Shanghai 200237, China
| | - Yi Yang
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
- Optogenetics & Molecular Imaging Interdisciplinary Research Center, CAS Center for Excellence in Brain Science, East China University of Science and Technology, Shanghai 200237, China
| | - Xianjun Chen
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
- Optogenetics & Molecular Imaging Interdisciplinary Research Center, CAS Center for Excellence in Brain Science, East China University of Science and Technology, Shanghai 200237, China
- Corresponding author. Tel.: +86 21 64252449; fax: +86 21 64258277.
| | - Feng Gao
- Shanghai Key Laboratory of New Drug Design, East China University of Science and Technology, Shanghai 200237, China
- Department of Pharmaceutics, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Corresponding author. Tel.: +86 21 64252449; fax: +86 21 64258277.
| |
Collapse
|
46
|
Tariq H, Bokhari SAI. Surface-functionalised hybrid nanoparticles for targeted treatment of cancer. IET Nanobiotechnol 2020; 14:537-547. [PMID: 33010128 PMCID: PMC8676046 DOI: 10.1049/iet-nbt.2020.0073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/11/2020] [Accepted: 07/03/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer is a leading cause of death worldwide. Despite the great advancement in understanding the pharmacology and biology of cancer, it still signifies one of the most serious human-health related problems. The current treatments for cancer may include surgery, radiotherapy, and chemotherapy, but these procedures have several limitations. Current studies have shown that nanoparticles (NPs) can be used as a novel strategy for cancer treatment. Developing nanosystems that allow lower doses of therapeutic agents, as well as their selective release in tumour cells, may resolve the challenges of targeted cancer therapy. In this review, the authors discuss the role of the size, shape, and surface modifications of NPs in cancer treatment. They also address the challenges associated with cancer therapies based on NPs. The overall purpose of this review is to summarise the recent developments in designing different hybrid NPs with promising therapeutic properties for different types of cancer.
Collapse
Affiliation(s)
- Hasnat Tariq
- Department of Biotechnology, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Syed Ali Imran Bokhari
- Department of Biological Sciences, International Islamic University, Islamabad, Pakistan
| |
Collapse
|
47
|
Tan J, Tay J, Hedrick J, Yang YY. Synthetic macromolecules as therapeutics that overcome resistance in cancer and microbial infection. Biomaterials 2020; 252:120078. [PMID: 32417653 DOI: 10.1016/j.biomaterials.2020.120078] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/24/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023]
Abstract
Synthetic macromolecular antimicrobials have shown efficacy in the treatment of multidrug resistant (MDR) pathogens. These synthetic macromolecules, inspired by Nature's antimicrobial peptides (AMPs), mitigate resistance by disrupting microbial cell membrane or targeting multiple intracellular proteins or genes. Unlike AMPs, these polymers are less prone to degradation by proteases and are easier to synthesize on a large scale. Recently, various studies have revealed that cancer cell membrane, like that of microbes, is negatively charged, and AMPs can be used as anticancer agents. Nevertheless, efforts in developing polymers as anticancer agents has remained limited. This review highlights the recent advancement in the development of synthetic biodegradable antimicrobial polymers (e.g. polycarbonates, polyesters and polypeptides) and anticancer macromolecules including peptides and polymers. Additionally, strategies to improve their in vivo bioavailability and selectivity towards bacteria and cancer cells are examined. Lastly, future perspectives, including use of artificial intelligence or machine learning, in the development of antimicrobial and anticancer macromolecules are discussed.
Collapse
Affiliation(s)
- Jason Tan
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore, 138669, Singapore; Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - Joyce Tay
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore, 138669, Singapore; Division of Chemistry and Biological Chemistry, School of Physical and Mathematical Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore, 637371, Singapore
| | - James Hedrick
- IBM Almaden Research Center, 650 Harry Road, San Jose, CA, 95120, United States
| | - Yi Yan Yang
- Institute of Bioengineering and Nanotechnology, 31 Biopolis Way, Singapore, 138669, Singapore.
| |
Collapse
|
48
|
Santa-González GA, Patiño-González E, Manrique-Moreno M. Cell cycle progression data on human skin cancer cells with anticancer synthetic peptide LTX-315 treatment. Data Brief 2020; 30:105443. [PMID: 32258289 PMCID: PMC7118296 DOI: 10.1016/j.dib.2020.105443] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/23/2022] Open
Abstract
Skin cancer, including melanoma and non-melanoma (NMSC), represents the most common type of malignancy in the white population [1]. The incidence rate of melanoma is increasing worldwide, while the associated mortality remains stable. On the other hand, the incidence of NMSC varies widely [1,2]. Camilio and collaborators recently described the anticancer properties of LTX-315, a novel synthetic anticancer peptide, commercialized as Oncopore™ [3,4]. Despite various studies demonstrating the efficiency of LTX-315 therapy in inducing cancer cell death, the effects on cell cycle progression of this antitumoral peptide are poorly understood. In this research, we present data about the effect of LTX-315 on the cell cycle of two skin cancer cell lines: epidermoid carcinoma cells (A431) and melanoma cells (A375); as well as on an immortalized normal keratinocyte cell line, HaCaT. Additionally, its cytotoxicity on the cells was determined by measuring the uptake of propidium iodide, in order to establish its relationship with cell cycle progression. The analysed data obtained by flow cytometry show different cell cycle distributions in non-tumoral and skin cancer-derived cell lines in response to LTX-315 treatment. Non-tumoral cells showed a sub-G1 peak, while for tumoral cells there was a shift in the G1peak without producing an obvious distant and distinct sub-G1 peak. This data is in accordance with a major decrease in cell viability in non-cancer cells.
Collapse
Affiliation(s)
- Gloria A. Santa-González
- Genetic Regeneration and Cancer Group, Faculty of Exact and Natural Sciences, Biology Institute, University of Antioquia, A.A. 1226, Medellin, Colombia
- Structural Biochemistry of Macromolecules Group, Faculty of Exact and Natural Sciences, Chemistry Institute, University of Antioquia, A.A. 1226, Medellin, Colombia
| | - Edwin Patiño-González
- Genetic Regeneration and Cancer Group, Faculty of Exact and Natural Sciences, Biology Institute, University of Antioquia, A.A. 1226, Medellin, Colombia
- Structural Biochemistry of Macromolecules Group, Faculty of Exact and Natural Sciences, Chemistry Institute, University of Antioquia, A.A. 1226, Medellin, Colombia
| | - Marcela Manrique-Moreno
- Structural Biochemistry of Macromolecules Group, Faculty of Exact and Natural Sciences, Chemistry Institute, University of Antioquia, A.A. 1226, Medellin, Colombia
- Corresponding author at: Calle 67 # 53 – 108, Bloque 2-235, Medellin, Colombia.
| |
Collapse
|
49
|
Fan F, Piao JG, Zhao Y, Jin L, Li M, Wang Y, Yang L. Bioinspired Membrane-Disruptive Macromolecules as Drug-Free Therapeutics. ACS APPLIED BIO MATERIALS 2020; 3:1267-1275. [DOI: 10.1021/acsabm.9b01143] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
50
|
Huang Y, Yan J, Peng S, Tang Z, Tan C, Ling J, Lin W, Lin X, Zu X, Yi G. pH/Reduction Dual-Stimuli-Responsive Cross-Linked Micelles Based on Multi-Functional Amphiphilic Star Copolymer: Synthesis and Controlled Anti-Cancer Drug Release. Polymers (Basel) 2020; 12:E82. [PMID: 31947729 PMCID: PMC7023672 DOI: 10.3390/polym12010082] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 12/03/2022] Open
Abstract
Novel approach has been constructed for preparing the amphiphilic star copolymer pH/reduction stimuli-responsive cross-linked micelles (SCMs) as a smart drug delivery system for the well-controlled anti-tumor drug doxorubicin (DOX) release. The SCMs had a low CMC value of 5.3 mg/L. The blank and DOX-loaded SCMs both had a spherical shape with sizes around 100-180 nm. In addition, the good stability and well pH/reduction-sensitivity of the SCMs were determined by dynamic light scattering (DLS) as well. The SCMs owned a low release of DOX in bloodstream and normal tissues while it had a fast release in tumor higher glutathione (GSH) concentration and/or lower pH value conditions, which demonstrates their pH/reduction dual-responsiveness. Furthermore, we conducted the thermodynamic analysis to study the interactions between the DOX and polymer micelles in the DOX release process. The values of the thermodynamic parameters at pH 7.4 and at pH 5.0 conditions indicated that the DOX release was endothermic and controlled mainly by the forces of an electrostatic interaction. At pH 5.0 with 10 mM GSH condition, electrostatic interaction, chemical bond, and hydrophobic interactions contributed together on DOX release. With the low cytotoxicity of blank SCMs and well cytotoxicity of DOX-loaded SCMs, the results indicated that the SCMs could form a smart cancer microenvironment-responsive drug delivery system. The release kinetic and thermodynamic analysis offer a theoretical foundation for the interaction between drug molecules and polymer matrices, which helps provide a roadmap for the oriented design and control of anti-cancer drug release for cancer therapy.
Collapse
Affiliation(s)
- Yunwei Huang
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Jingye Yan
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Shiyuan Peng
- Guangdong Provincial Key Lab of Green Chemical Product Technology, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Zilun Tang
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Cuiying Tan
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Jiabao Ling
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Wenjing Lin
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaofeng Lin
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Xihong Zu
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| | - Guobin Yi
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|