1
|
Lu X, Wan X, Lian J, Peng J, Jing P, Guo Q, Liao Y, Jiang Y, Yang C, Jin L, Shi S, Yao Y, Hu WW, Luo J. Antibiotic-based micelles with bone-targeting and pH-responsive properties for infectious osteomyelitis treatment. J Colloid Interface Sci 2025; 685:648-660. [PMID: 39862844 DOI: 10.1016/j.jcis.2025.01.136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
We developed antibiotic-based micelles with bone-targeting and charge-switchable properties (P-CASMs) for treating infectious osteomyelitis. The amphiphilic molecules are formed by combining ciprofloxacin (CIP) with ligand 1 through a mild salifying reaction, and spontaneously self-assemble into antibiotic-based micelles (ASMs) in aqueous solution. Acrylate groups on ligand 1 enable cross-linking of ASMs with pentaerythritol tetra(mercaptopropionate) via a click reaction, forming pH-sensitive cross-linked micelles (CASMs). The incorporation of vinylphosphonates imparts bone-targeting and charge-switchable properties of CASMs, creating P-CASMs. These P-CASMs exhibit good biocompatibility at physiological pH and strong adhesion to bone infection sites (pH 5.5) due to electrostatic interactions. They can effectively penetrate bacterial biofilms and release antibiotics in response to the local microenvironment, thereby eradicating bacteria. Compared to previous systems, the P-CASMs show higher drug loading (∼23 %), improved stability, and better biosafety. This innovative system holds substantial potential for clinical applications in the treatment of osteomyelitis.
Collapse
Affiliation(s)
- Xinyu Lu
- College of Chemistry and Environment, Southwest Minzu University, Chengdu 610041 China; Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu 610041 China
| | - Xiaohui Wan
- College of Chemistry and Environment, Southwest Minzu University, Chengdu 610041 China
| | - Jiali Lian
- College of Chemistry and Environment, Southwest Minzu University, Chengdu 610041 China
| | - Jiaoying Peng
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu 610041 China
| | - Pei Jing
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, China
| | - Qiao Guo
- College of Chemistry and Environment, Southwest Minzu University, Chengdu 610041 China
| | - Yulong Liao
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu 610041 China
| | - Yuchen Jiang
- College of Chemistry and Environment, Southwest Minzu University, Chengdu 610041 China
| | - Chengli Yang
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu 610041 China
| | - Lunqiang Jin
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu 610041 China
| | - Shaorui Shi
- Department of Laboratory Medicine, The Second People's Hospital of Yibin, West China Hospital, Yibin Hospital Sichuan University, Yibin 644000 China.
| | - Yongchao Yao
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu 610041 China.
| | - Wenchuang Walter Hu
- Precision Medicine Translational Research Center, West China Hospital, Sichuan University, Chengdu 610041 China.
| | - Jianbin Luo
- College of Chemistry and Environment, Southwest Minzu University, Chengdu 610041 China.
| |
Collapse
|
2
|
Wei HX, Liu MH, Wang TY, Shih MH, Yu J, Yeh YC. Fabrication of pH- and Ultrasound-Responsive Polymeric Micelles: The Effect of Amphiphilic Block Copolymers with Different Hydrophilic/Hydrophobic Block Ratios for Self-Assembly and Controlled Drug Release. Biomacromolecules 2025; 26:2116-2130. [PMID: 40067950 PMCID: PMC12004527 DOI: 10.1021/acs.biomac.4c01202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 04/15/2025]
Abstract
Stimuli-responsive polymeric vehicles can change their physical or chemical properties when exposed to internal or external triggers, enabling precise spatiotemporal control of drug release. Nevertheless, systematic research is lacking in preparing dual stimuli-responsive amphiphilic block copolymers with different hydrophilic/hydrophobic block ratios in forming self-assembled structures. Here, we synthesized two types of block copolymers consisting of the hydrophobic segments (i.e., pH-responsive 2-(diethylamino)ethyl methacrylate (DEA) and ultrasound-responsive 2-methoxyethyl methacrylate (MEMA)) and hydrophilic poly(ethylene glycol) methyl ether (mPEG) segments, forming mPEGX-b-P(DEAY-co-MEMAZ). These amphiphilic block copolymers can self-assemble to form polymeric micelles, and their structures (e.g., size) and properties (e.g., critical vesicle concentration, stability, stimuli-responsiveness to pH and ultrasound, drug loading efficiency, and controlled drug release performance) were thoroughly investigated. In vitro cell studies further demonstrate that ultrasound can efficiently trigger drug release from polymeric micelles, emphasizing their potential for controlled drug delivery in therapeutic applications.
Collapse
Affiliation(s)
- Hong-Xiang Wei
- Institute
of Polymer Science and Engineering, National
Taiwan University, Taipei 10617, Taiwan
| | - Ming-Hsin Liu
- Department
of Chemical Engineering, National Taiwan
University, Taipei 10617, Taiwan
| | - Tzu-Ying Wang
- Institute
of Polymer Science and Engineering, National
Taiwan University, Taipei 10617, Taiwan
| | - Meng-Hsiu Shih
- Department
of Chemical Engineering, National Taiwan
University, Taipei 10617, Taiwan
| | - Jiashing Yu
- Department
of Chemical Engineering, National Taiwan
University, Taipei 10617, Taiwan
| | - Yi-Cheun Yeh
- Institute
of Polymer Science and Engineering, National
Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
3
|
Hou J, Xue Z, Chen Y, Li J, Yue X, Zhang Y, Gao J, Hao Y, Shen J. Development of Stimuli-Responsive Polymeric Nanomedicines in Hypoxic Tumors and Their Therapeutic Promise in Oral Cancer. Polymers (Basel) 2025; 17:1010. [PMID: 40284275 PMCID: PMC12030766 DOI: 10.3390/polym17081010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/16/2025] [Accepted: 03/24/2025] [Indexed: 04/29/2025] Open
Abstract
Hypoxic tumors pose considerable obstacles to cancer treatment, as diminished oxygen levels can impair drug effectiveness and heighten therapeutic resistance. Oral cancer, a prevalent malignancy, encounters specific challenges owing to its intricate anatomical structure and the technical difficulties in achieving complete resection, thereby often restricting treatment efficacy. The impact of hypoxia is particularly critical in influencing both the treatment response and prognosis of oral cancers. This article summarizes and examines the potential of polymer nanomedicines to address these challenges. By engineering nanomedicines that specifically react to the hypoxic tumor microenvironment, these pharmaceuticals can markedly enhance targeting precision and therapeutic effectiveness. Polymer nanomedicines enhance therapeutic efficacy while reducing side effects by hypoxia-targeted accumulation. The article emphasizes that these nanomedicines can overcome the drug resistance frequently observed in hypoxic tumors by improving the delivery and bioavailability of anticancer agents. Furthermore, this review elucidates the design and application of polymer nanomedicines for treating hypoxic tumors, highlighting their transformative potential in cancer therapy. Finally, this article gives an outlook on stimuli-responsive polymeric nanomedicines in the treatment of oral cancer.
Collapse
Affiliation(s)
- Jialong Hou
- Department of Operative Dentistry and Endodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China; (J.H.); (Z.X.)
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Zhijun Xue
- Department of Operative Dentistry and Endodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China; (J.H.); (Z.X.)
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Yao Chen
- Department of Operative Dentistry and Endodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China; (J.H.); (Z.X.)
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Jisen Li
- Tianjin Key Laboratory for Disaster Medicine Technology, Institution of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China;
| | - Xin Yue
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
| | - Ying Zhang
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
| | - Jing Gao
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
| | - Yonghong Hao
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
- The Second Clinical Division, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
| | - Jing Shen
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
| |
Collapse
|
4
|
Weng L, Zhao M, Chen Z, Zhu L. Hypoxia-Targeted Responsive Delivery of Doxorubicin and Digoxin for Synergistic Treatment of Triple-Negative Breast Cancer. Mol Pharm 2025; 22:2142-2158. [PMID: 40059340 DOI: 10.1021/acs.molpharmaceut.4c01325] [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] [Indexed: 04/08/2025]
Abstract
To enhance the therapeutic efficacy and safety of triple-negative breast cancer (TNBC) treatment, we developed a hypoxia-responsive drug delivery system utilizing digoxin (DIG) to inhibit HIF-1α and sensitize TNBC to doxorubicin (DOX). DIG, a cardiac steroid with a well-characterized pharmacological mechanism, was encapsulated in micelles composed of methoxy-polyethylene glycol (mPEG) and poly(lactic acid) (PLA) copolymers, incorporating an azobenzene (AZO) trigger for hypoxia-sensitive drug release. The loading ratio of DOX to DIG was optimized based on DIG's minimum effective dose. In vitro and in vivo studies demonstrated that the micelles efficiently delivered their payload to hypoxic tumor regions, enabling rapid drug release. DIG-mediated HIF-1α inhibition enhanced TNBC sensitivity to DOX, leading to significant suppression of both primary tumor growth and pulmonary metastasis. This study presents a promising and clinically feasible strategy for TNBC and other hypoxia-driven malignancies.
Collapse
Affiliation(s)
- Lingyan Weng
- Institute of Special Environmental Medicine, Medical School, Nantong University, Nantong 226019, China
| | - Min Zhao
- Institute of Special Environmental Medicine, Medical School, Nantong University, Nantong 226019, China
| | - Zhongping Chen
- Institute of Special Environmental Medicine, Medical School, Nantong University, Nantong 226019, China
| | - Li Zhu
- Institute of Special Environmental Medicine, Medical School, Nantong University, Nantong 226019, China
| |
Collapse
|
5
|
Xie S, Zhou Y, Zhu H, Xu X, Zhang H, Yuan C, Huang M, Xu P, Li J, Liu Y. Interface-driven structural evolution on diltiazem as novel uPAR inhibitors: from in silico design to in vitro evaluation. Mol Divers 2025; 29:1261-1274. [PMID: 38935305 DOI: 10.1007/s11030-024-10908-7] [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: 04/21/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024]
Abstract
The urokinase-type plasminogen activator receptor (uPAR) emerges as a key target for anti-metastasis owing to its pivotal role in facilitating the invasive and migratory processes of cancer cells. Recently, we identified the uPAR-targeting anti-metastatic ability of diltiazem (22), a commonly used antihypertensive agent. Fine-tuning the chemical structures of known hits represents a vital branch of drug development. To develop novel anti-metastatic drugs, we performed an interface-driven structural evolution strategy on 22. The uPAR-targeting and anti-cancer abilities of this antihypertensive drug wereidentified by us recently. Based on in silico strategy, including extensive molecular dynamics (MD) simulations, hierarchical binding free energy predictions, and ADMET profilings, we designed, synthesized, and identified three new diltiazem derivatives (221-8, 221-57, and 221-68) as uPAR inhibitors. Indeed, all of these three derivatives exhibited uPAR-depending inhibitory activity against PC-3 cell line invasion at micromolar level. Particularly, derivatives 221-68 and 221-8 showed enhanced uPAR-dependent inhibitory activity against the tumor cell invasion compared to the original compound. Microsecond timesclae MD simulations demonstrated the optimized moiety of 221-68 and 221-8 forming more comprehensive interactions with the uPAR, highlighting the reasonability of our strategy. This work introduces three novel uPAR inhibitors, which not only pave the way for the development of effective anti-metastatic therapeutics, but also emphasize the efficacy and robustness of an in silico-based lead compound optimization strategy in drug design.
Collapse
Affiliation(s)
- Song Xie
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
- College of Chemistry, Fuzhou University, Fuzhou, 350116, China
| | - Yang Zhou
- College of Chemistry, Fuzhou University, Fuzhou, 350116, China
| | - Hao Zhu
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China
| | - Xinyi Xu
- College of Chemistry, Fuzhou University, Fuzhou, 350116, China
| | - Han Zhang
- College of Chemistry, Fuzhou University, Fuzhou, 350116, China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350116, China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, 350116, China
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, 350116, China
| | - Jinyu Li
- College of Chemistry, Fuzhou University, Fuzhou, 350116, China.
- Fujian Provincial Key Laboratory of Theoretical and Computational Chemistry, Xiamen, 361005, China.
| | - Yichang Liu
- School of Pharmacy, Nantong University, Nantong, 226001, Jiangsu, China.
| |
Collapse
|
6
|
Ma R, Zhang Y, Ji H, Fu H, Gu S, Su Q, Lin Y, Deng Q, Xue W, Yang Y. Glucose oxidase-driven self-accelerating drug release nanosystem based on metal-phenolic networks orchestrates tumor chemotherapy and ferroptosis-based therapy. Int J Biol Macromol 2025; 290:139103. [PMID: 39716697 DOI: 10.1016/j.ijbiomac.2024.139103] [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: 11/01/2024] [Revised: 12/14/2024] [Accepted: 12/20/2024] [Indexed: 12/25/2024]
Abstract
Nanocarriers responding to tumor microenvironment have been extensively exploited to improve the antitumor outcome of chemotherapeutic drugs. However, selectively and completely releasing drugs within the tumor remains a challenge, thereby limiting the therapeutic effect of drug delivery nanosystem. To tackle this challenge, a metal-phenolic networks (MPNs)-based nanosystem (F-MGD) showing the capability of self-accelerating drug release was originally fabricated in this study. Glucose oxidase (GOx) encapsulated in F-MGD could conduct the glucose transformation in tumor to cause the oxygen consumption and the production of gluconic acid and H2O2. Therefore, F-MGD with acid and hypoxia sensitivities thoroughly disintegrated under the aggravated acidity and hypoxia to achieve a more complete drug release. Besides, the product of H2O2 was readily decomposed into hydroxyl radicals via the iron ion-mediated Fenton reaction, which markedly augmented the oxidative stress in tumor cells and promoted ferroptosis. The results of both in vitro and in vivo assays demonstrated the significant antitumor efficacy of F-MGD. Collectively, this study proposes a strategy to expedite drug release in tumor and improve the tumor treatment effect by combining ferroptosis-based therapy and chemotherapy.
Collapse
Affiliation(s)
- Rongying Ma
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Yufei Zhang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Hongting Ji
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Huiling Fu
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Shuzhen Gu
- Department of Gynecology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Qianhong Su
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Yumian Lin
- School of Food Science and Engineering, Hainan University, Haikou 570228, China
| | - Qingchun Deng
- Department of Gynecology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Guangdong Provincial Engineering and Technological Research Center for Drug Carrier Development, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Yong Yang
- School of Food Science and Engineering, Hainan University, Haikou 570228, China.
| |
Collapse
|
7
|
Ying X, Chen Q, Yang Y, Wu Z, Zeng W, Miao C, Huang Q, Ai K. Nanomedicines harnessing cGAS-STING pathway: sparking immune revitalization to transform 'cold' tumors into 'hot' tumors. Mol Cancer 2024; 23:277. [PMID: 39710707 DOI: 10.1186/s12943-024-02186-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 11/26/2024] [Indexed: 12/24/2024] Open
Abstract
cGAS-STING pathway stands at the forefront of innate immunity and plays a critical role in regulating adaptive immune responses, making it as a key orchestrator of anti-tumor immunity. Despite the great potential, clinical outcomes with cGAS-STING activators have been disappointing due to their unfavorable in vivo fate, signaling an urgent need for innovative solutions to bridge the gap in clinical translation. Recent advancements in nanotechnology have propelled cGAS-STING-targeting nanomedicines to the cutting-edge of cancer therapy, leveraging precise drug delivery systems and multifunctional platforms to achieve remarkable region-specific biodistribution and potent therapeutic efficacy. In this review, we provide an in-depth exploration of the molecular mechanisms that govern cGAS-STING signaling and its potential to dynamically modulate the anti-tumor immune cycle. We subsequently introduced several investigational cGAS-STING-dependent anti-tumor agents and summarized their clinical trial progress. Additionally, we provided a comprehensive review of the unique advantages of cGAS-STING-targeted nanomedicines, highlighting the transformative potential of nanotechnology in this field. Furthermore, we comprehensively reviewed and comparatively analyzed the latest breakthroughs cGAS-STING-targeting nanomedicine, focusing on strategies that induce cytosolic DNA generation via exogenous DNA delivery, chemotherapy, radiotherapy, or dynamic therapies, as well as the nanodelivery of STING agonists. Lastly, we discuss the future prospects and challenges in cGAS-STING-targeting nanomedicine development, offering new insights to bridge the gap between mechanistic research and drug development, thereby opening new pathways in cancer treatment.
Collapse
Affiliation(s)
- Xiaohong Ying
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Qiaohui Chen
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Yongqi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Ziyu Wu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Wan Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Chenxi Miao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410013, China.
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Xiangya Hospital, Ministry of Education, Central South University, Changsha, 410008, China.
| |
Collapse
|
8
|
Gu J, Jiang L, Chen Z, Qi J. A simple nanoplatform of thermo-sensitive liposomes and gold nanorods to treat bone metastasis through improved chemotherapy combined with photothermal therapy. Int J Pharm X 2024; 8:100282. [PMID: 39286038 PMCID: PMC11403519 DOI: 10.1016/j.ijpx.2024.100282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024] Open
Abstract
Bone metastasis remains a clinical challenge and is still considered incurable. While nanoparticles-based drug delivery and photothermal therapy (PTT) show promise in treating subcutaneous solid tumor, their therapeutic outcome in treating bone metastasis is limited, due to the inaccessibility of bone metastatic site and the complexity of bone metastasis. Herein, we reported a simple nanoplatform composed of thermo-sensitive liposomes (TSL) and gold nanorods (GNR) to treat bone metastasis through improved chemotherapy combined with GNR-assisted PTT. Lipid combination of TSL was firstly tailored to regulate its stability under physiological condition as well as its sensitivity in responding to PTT-caused mild hyperthermia. The obtained TSL with loaded drug was then combined with GNR to form the nanoplatform through unsophisticated incubation. Cell experiments revealed that upon near-infrared (NIR) irradiation, the nanoplatform effectively inhibited the viability and migration ability of tumor cells through PTT, PTT-triggered thermo-sensitive drug release, and PTT-augmented sensitivity of tumor cells to drug. In a murine model of bone metastasis, the nanoplatform enabled effective delivery of loaded drug and GNR to bone metastatic site for rapid drug release upon local NIR irradiation. Through killing tumor cells and rebalancing the turnover of osteoclasts and osteoblasts, the nanoplatform largely preserved bone structure for pain relief and survival extension. Inspired by the simplicity of nanoplatform acquirement and treatment operation, the strategy of liposomes-based thermo-sensitive drug delivery in combination with GNR-assisted PTT is considered greatly promising in treating bone metastasis.
Collapse
Affiliation(s)
- Jia Gu
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
- Institute of Special Environmental Medicine, Nantong University, Nantong, People's Republic of China
| | - Lifan Jiang
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Zhongping Chen
- Institute of Special Environmental Medicine, Nantong University, Nantong, People's Republic of China
| | - Jun Qi
- Department of Burns and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| |
Collapse
|
9
|
Chenab KK, Malektaj H, Nadinlooie AAR, Mohammadi S, Zamani-Meymian MR. Intertumoral and intratumoral barriers as approaches for drug delivery and theranostics to solid tumors using stimuli-responsive materials. Mikrochim Acta 2024; 191:541. [PMID: 39150483 DOI: 10.1007/s00604-024-06583-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 07/15/2024] [Indexed: 08/17/2024]
Abstract
The solid tumors provide a series of biological barriers in cellular microenvironment for designing drug delivery methods based on advanced stimuli-responsive materials. These intertumoral and intratumoral barriers consist of perforated endotheliums, tumor cell crowding, vascularity, lymphatic drainage blocking effect, extracellular matrix (ECM) proteins, hypoxia, and acidosis. Triggering opportunities have been drawn for solid tumor therapies based on single and dual stimuli-responsive drug delivery systems (DDSs) that not only improved drug targeting in deeper sites of the tumor microenvironments, but also facilitated the antitumor drug release efficiency. Single and dual stimuli-responsive materials which are known for their lowest side effects can be categorized in 17 main groups which involve to internal and external stimuli anticancer drug carriers in proportion to microenvironments of targeted solid tumors. Development of such drug carriers can circumvent barriers in clinical trial studies based on their superior capabilities in penetrating into more inaccessible sites of the tumor tissues. In recent designs, key characteristics of these DDSs such as fast response to intracellular and extracellular factors, effective cytotoxicity with minimum side effect, efficient permeability, and rate and location of drug release have been discussed as core concerns of designing paradigms of these materials.
Collapse
Affiliation(s)
- Karim Khanmohammadi Chenab
- Department of Chemistry, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
- Department of Physics, Iran University of Science and Technology, Tehran, P.O. Box 16846-13114, Iran
| | - Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, 9220, Aalborg, Denmark
| | | | | | | |
Collapse
|
10
|
Serras A, Faustino C, Pinheiro L. Functionalized Polymeric Micelles for Targeted Cancer Therapy: Steps from Conceptualization to Clinical Trials. Pharmaceutics 2024; 16:1047. [PMID: 39204392 PMCID: PMC11359152 DOI: 10.3390/pharmaceutics16081047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 07/28/2024] [Accepted: 08/01/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer is still ranked among the top three causes of death in the 30- to 69-year-old age group in most countries and carries considerable societal and macroeconomic costs that differ depending on the cancer type, geography, and patient gender. Despite advances in several pharmacological approaches, the lack of stability and specificity, dose-related toxicity, and limited bioavailability of chemotherapy (standard therapy) pose major obstacles in cancer treatment, with multidrug resistance being a driving factor in chemotherapy failure. The past three decades have been the stage for intense research activity on the topic of nanomedicine, which has resulted in many nanotherapeutics with reduced toxicity, increased bioavailability, and improved pharmacokinetics and therapeutic efficacy employing smart drug delivery systems (SDDSs). Polymeric micelles (PMs) have become an auspicious DDS for medicinal compounds, being used to encapsulate hydrophobic drugs that also exhibit substantial toxicity. Through preclinical animal testing, PMs improved pharmacokinetic profiles and increased efficacy, resulting in a higher safety profile for therapeutic drugs. This review focuses on PMs that are already in clinical trials, traveling the pathways from preclinical to clinical studies until introduction to the market.
Collapse
Affiliation(s)
| | - Célia Faustino
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa (ULisboa), Avenida Professor Gama PintoGama Pinto, 1649-003 Lisboa, Portugal; (A.S.); (L.P.)
| | | |
Collapse
|
11
|
Nsanzamahoro S, Nan F, Shen L, Iradukunda Y, Li B, Yu WW. Designing a Hypoxia-Activated Sensing Platform Using an Azo Group-Triggered Reaction with the Formation of Silicon Nanoparticles. Anal Chem 2024; 96:11977-11984. [PMID: 38975827 DOI: 10.1021/acs.analchem.4c01857] [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: 07/09/2024]
Abstract
Hypoxia is known as a specific signal of various diseases, such as liver fibrosis. We designed a hypoxia-sensitive fluorometric approach that cleaved the azo bond (N═N) in the presence of hypoxia-controlled agents (sodium dithionite and azoreductase). 4-(2-Pyridylazo) resorcinol (Py-N═N-RC) bears a desirable hypoxia-responsive linker (N═N), and its azo bond breakup can only occur in the presence of sodium dithionite and azoreductase and leads to the release of 2,4-dihydroxyaniline, which can react with 3-[2-(2-aminoethylamino)ethylamino]propyltrimethoxysilane to generate yellow fluorescent silicon nanoparticles. This approach exhibited high selectivity and sensitivity toward both sodium dithionite and azoreductase over other potential interferences. The mouse liver microsome, which is known to contain azoreductase, was applied and confirmed the feasibility of the designed platform. Py-N═N-RC is expected to be a practical substrate for hypoxia-related biological analyses. Furthermore, silicon nanoparticles were successfully applied for Hela cell imaging owing to their negligible cytotoxicity and superb biocompatibility.
Collapse
Affiliation(s)
- Stanislas Nsanzamahoro
- School of Chemistry and Chemical Engineering, Ministry of Education Key Laboratory of Special Functional Aggregated Materials, Shandong Key Laboratory of Advanced Organosilicon Materials and Technologies, Shandong University, Jinan 250100, China
- Shandong Provincial Key Laboratory for Science of Material Creation and Energy Conversion, Science Center for Material Creation and Energy Conversion, Shandong University, Qingdao 266237, China
| | - Fuchun Nan
- School of Chemistry and Chemical Engineering, Ministry of Education Key Laboratory of Special Functional Aggregated Materials, Shandong Key Laboratory of Advanced Organosilicon Materials and Technologies, Shandong University, Jinan 250100, China
- Shandong Provincial Key Laboratory for Science of Material Creation and Energy Conversion, Science Center for Material Creation and Energy Conversion, Shandong University, Qingdao 266237, China
| | - Lanbo Shen
- Jinan Central Hospital, Shandong First Medical University, Jinan 250013, China
| | - Yves Iradukunda
- Key Laboratory of Chemistry of Northwestern Plant Resources, Key Laboratory for Natural Medicine of Gansu Province, Lanzhou Institute of Chemical Physics, Chinese Academy of Sciences, Lanzhou 730000, China
| | - Bin Li
- Jinan Central Hospital, Shandong First Medical University, Jinan 250013, China
| | - William W Yu
- School of Chemistry and Chemical Engineering, Ministry of Education Key Laboratory of Special Functional Aggregated Materials, Shandong Key Laboratory of Advanced Organosilicon Materials and Technologies, Shandong University, Jinan 250100, China
- Shandong Provincial Key Laboratory for Science of Material Creation and Energy Conversion, Science Center for Material Creation and Energy Conversion, Shandong University, Qingdao 266237, China
| |
Collapse
|
12
|
Wang L, Wang D, Lei W, Sun T, Gu B, Dong H, Taniguchi Y, Liu Y, Ling Y. Trigonometric Bundling Disulfide Unit Starship Synergizes More Effectively to Promote Cellular Uptake. Int J Mol Sci 2024; 25:7518. [PMID: 39062760 PMCID: PMC11277142 DOI: 10.3390/ijms25147518] [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: 05/20/2024] [Revised: 07/06/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
A small molecule disulfide unit technology platform based on dynamic thiol exchange chemistry at the cell membrane has the potential for drug delivery. However, the alteration of the CSSC dihedral angle of the disulfide unit caused by diverse substituents directly affects the effectiveness of this technology platform as well as its own chemical stability. The highly stable open-loop relaxed type disulfide unit plays a limited role in drug delivery due to its low dihedral angle. Here, we have built a novel disulfide unit starship based on the 3,4,5-trihydroxyphenyl skeleton through trigonometric bundling. The intracellular delivery results showed that the trigonometric bundling of the disulfide unit starship effectively promoted cellular uptake without any toxicity, which is far more than 100 times more active than that of equipment with a single disulfide unit in particular. Then, the significant reduction in cell uptake capacity (73-93%) using thiol erasers proves that the trigonometric bundling of the disulfide starship is an endocytosis-independent internalization mechanism via a dynamic covalent disulfide exchange mediated by thiols on the cell surface. Furthermore, analysis of the molecular dynamics simulations demonstrated that trigonometric bundling of the disulfide starship can significantly change the membrane curvature while pushing lipid molecules in multiple directions, resulting in a significant distortion in the membrane structure and excellent membrane permeation performance. In conclusion, the starship system we built fully compensates for the inefficiency deficiencies induced by poor dihedral angles.
Collapse
Affiliation(s)
- Lei Wang
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Dezhi Wang
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Wenzhuo Lei
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Tiantian Sun
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Bei Gu
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Han Dong
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Yosuke Taniguchi
- School of Pharmaceutical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan;
| | - Yichang Liu
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| | - Yong Ling
- School of Pharmacy, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, China; (D.W.); (W.L.); (T.S.); (B.G.); (H.D.); (Y.L.)
| |
Collapse
|
13
|
Ren C, Chen X, Hao X, Wu C, Xie L, Liu X. Integrated machine learning algorithms reveal a bone metastasis-related signature of circulating tumor cells in prostate cancer. Sci Data 2024; 11:701. [PMID: 38937469 PMCID: PMC11211408 DOI: 10.1038/s41597-024-03551-2] [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: 01/29/2024] [Accepted: 06/19/2024] [Indexed: 06/29/2024] Open
Abstract
Bone metastasis is an essential factor affecting the prognosis of prostate cancer (PCa), and circulating tumor cells (CTCs) are closely related to distant tumor metastasis. Here, the protein-protein interaction (PPI) networks and Cytoscape application were used to identify diagnostic markers for metastatic events in PCa. We screened ten hub genes, eight of which had area under the ROC curve (AUC) values > 0.85. Subsequently, we aim to develop a bone metastasis-related model relying on differentially expressed genes in CTCs for accurate risk stratification. We developed an integrative program based on machine learning algorithm combinations to construct reliable bone metastasis-related genes prognostic index (BMGPI). On the basis of BMGPI, we carefully evaluated the prognostic outcomes, functional status, tumor immune microenvironment, somatic mutation, copy number variation (CNV), response to immunotherapy and drug sensitivity in different subgroups. BMGPI was an independent risk factor for disease-free survival in PCa. The high risk group demonstrated poor survival as well as higher immune scores, higher tumor mutation burden (TMB), more frequent co-occurrence mutation, and worse efficacy of immunotherapy. This study highlights a new prognostic signature, the BMGPI. BMGPI is an independent predictor of prognosis in PCa patients and is closely associated with the immune microenvironment and the efficacy of immunotherapy.
Collapse
Affiliation(s)
- Congzhe Ren
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangyu Chen
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuexue Hao
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Changgui Wu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lijun Xie
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
14
|
Ding L, Agrawal P, Singh SK, Chhonker YS, Sun J, Murry DJ. Polymer-Based Drug Delivery Systems for Cancer Therapeutics. Polymers (Basel) 2024; 16:843. [PMID: 38543448 PMCID: PMC10974363 DOI: 10.3390/polym16060843] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 11/12/2024] Open
Abstract
Chemotherapy together with surgery and/or radiotherapy are the most common therapeutic methods for treating cancer. However, the off-target effects of chemotherapy are known to produce side effects and dose-limiting toxicities. Novel delivery platforms based on natural and synthetic polymers with enhanced pharmacokinetic and therapeutic potential for the treatment of cancer have grown tremendously over the past 10 years. Polymers can facilitate selective targeting, enhance and prolong circulation, improve delivery, and provide the controlled release of cargos through various mechanisms, including physical adsorption, chemical conjugation, and/or internal loading. Notably, polymers that are biodegradable, biocompatible, and physicochemically stable are considered to be ideal delivery carriers. This biomimetic and bio-inspired system offers a bright future for effective drug delivery with the potential to overcome the obstacles encountered. This review focuses on the barriers that impact the success of chemotherapy drug delivery as well as the recent developments based on natural and synthetic polymers as platforms for improving drug delivery for treating cancer.
Collapse
Affiliation(s)
- Ling Ding
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
| | - Prachi Agrawal
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (P.A.); (J.S.)
| | - Sandeep K. Singh
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
| | - Yashpal S. Chhonker
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
| | - Jingjing Sun
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; (P.A.); (J.S.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Daryl J. Murry
- Clinical Pharmacology Laboratory, Department of Pharmacy Practice and Science, University of Nebraska Medical Center, Omaha, NE 68198, USA; (L.D.); (S.K.S.); (Y.S.C.)
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
15
|
Ding Y, Yu W, Shen R, Zheng X, Zheng H, Yao Y, Zhang Y, Du C, Yi H. Hypoxia-Responsive Tetrameric Supramolecular Polypeptide Nanoprodrugs for Combination Therapy. Adv Healthc Mater 2024; 13:e2303308. [PMID: 37924332 DOI: 10.1002/adhm.202303308] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/30/2023] [Indexed: 11/06/2023]
Abstract
Despite the intense progress of photodynamic and chemotherapy, however, they cannot prevent solid tumor invasion, metastasis, and relapse, along with inferior efficacy and severe side effects. The hypoxia-responsive nanoprodrugs integrating photodynamic functions are highly sought to address the above-mentioned problems and overcome the tumor hypoxia-reduced efficacy. Herein, a hypoxia-responsive tetrameric supramolecular polypeptide nanoprodrug (SPN-TAPP-PCB4) is constructed from the self-assembly of tetrameric porphyrin-central poly(l-lysine-azobenzene-chlorambucil) (TAPP-(PLL-Azo-CB)4) and an anionic water-soluble [2]biphenyl-extended-pillar[6]arene (AWBpP6) via the synergy of hydrophobic, π-π stacking, and host-guest interactions. Upon laser irradiation, the central TAPP can convert oxygen to generate single oxygen (1 O2 ) to kill tumor cells. Furthermore, under the acidic and PDT-aggravated hypoxia tumor cell microenvironment, SPN-TAPP-PCB4 is rapidly disassembled, and then efficiently releases activated CB through the hypoxic-responsive cleavage of azobenzene linkages. Both in vitro and in vivo biological studies showcase synergistic cancer-killing actions between photodynamic therapy (PDT) and chemotherapy (CT) with negligible toxicity. Consequently, this supramolecular polypeptide nanoprodrug offers an effective strategy to design a hypoxia-responsive nanoprodrug for a potential combo PDT-CT transition.
Collapse
Affiliation(s)
- Yue Ding
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Wei Yu
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Rongkai Shen
- Department of Orthopedics, The First Affiliated Hospital of Fujian Medical University, 20, Chazhong Rd., Fuzhou, Fujian, 350005, China
| | - Xiangqin Zheng
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, National Key Clinical Specialty Construction Program of (Gynecology), Fujian Province Key Clinical Specialty for Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, 350001, China
| | - Hui Zheng
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, National Key Clinical Specialty Construction Program of (Gynecology), Fujian Province Key Clinical Specialty for Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, 350001, China
| | - Yong Yao
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Yuehua Zhang
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, 226019, P. R. China
| | - Chang Du
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Huan Yi
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, National Key Clinical Specialty Construction Program of (Gynecology), Fujian Province Key Clinical Specialty for Gynecology, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, 350001, China
| |
Collapse
|
16
|
Hu R, Lan J, Zhang D, Shen W. Nanotherapeutics for prostate cancer treatment: A comprehensive review. Biomaterials 2024; 305:122469. [PMID: 38244344 DOI: 10.1016/j.biomaterials.2024.122469] [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: 09/27/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
Prostate cancer (PCa) is the most prevalent solid organ malignancy and seriously affects male health. The adverse effects of prostate cancer therapeutics can cause secondary damage to patients. Nanotherapeutics, which have special targeting abilities and controlled therapeutic release profiles, may serve as alternative agents for PCa treatment. At present, many nanotherapeutics have been developed to treat PCa and have shown better treatment effects in animals than traditional therapeutics. Although PCa nanotherapeutics are highly attractive, few successful cases have been reported in clinical practice. To help researchers design valuable nanotherapeutics for PCa treatment and avoid useless efforts, herein, we first reviewed the strategies and challenges involved in prostate cancer treatment. Subsequently, we presented a comprehensive review of nanotherapeutics for PCa treatment, including their targeting methods, controlled release strategies, therapeutic approaches and mechanisms. Finally, we proposed the future prospects of nanotherapeutics for PCa treatment.
Collapse
Affiliation(s)
- Ruimin Hu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jin Lan
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Dinglin Zhang
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Wenhao Shen
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
17
|
Sun C, Bai S, Chen S, Chen J, Liu P, Wu Y, Zhao X, Wu Z. Insufficient Effective Time of Suberanilohydroxamic Acid, a Deacetylase Inhibitor, Treatment Promotes PC3 Cell Growth. Biol Pharm Bull 2024; 47:1708-1716. [PMID: 39462585 DOI: 10.1248/bpb.b24-00408] [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] [Indexed: 10/29/2024]
Abstract
Castration-resistant prostate cancer (CRPC) contributes mostly to prostate cancer-specific mortality, and conventional castration therapy is almost ineffective, new therapies are needed. As a new potential anti-cancer drug, histone deacetylases (HDACs) inhibitors were demonstrated to be effective in inhibiting drug-resistance cancers in preclinical studies, but the results from clinical trials on CRPC patients were disappointing, and the reasons are unknown. In this study, we investigated the effect of suberanilohydroxamic acid (SAHA), a broad-spectrum pan-HDAC inhibitor, on proliferation, apoptosis, cell cycle progression in PC3 cells, and found that, unlike significant inhibiting effects at high-dose, low-dose SAHA significantly promoted PC3 cell growth. Further colony formation assay showed that the inhibitory effect of SAHA is also dependent on the treatment time, high-dose SAHA also exhibited promoting effect on PC3 cells when the treatment time was insufficient. However, this effect was not observed in another CRPC cell line, DU145, or another HDAC inhibitor, Trichostatin A (TSA). Our results indicate that, instead of inhibitory effect, SAHA would promote PC3 cell growth if the dose is low or the treatment time is insufficient, but this effect has not been observed in other CRPC cell line or HDAC inhibitors.
Collapse
Affiliation(s)
- Chuan Sun
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital
- Oncology & Radiotherapy Department, Zhejiang Hospital
| | - Shiting Bai
- Department of Pain Medicine, Zhejiang Hospital
| | - Sisi Chen
- Second Clinical Medical College, Zhejiang Chinese Medical University
| | - Jianglin Chen
- Second Clinical Medical College, Zhejiang Chinese Medical University
| | - Pengyuan Liu
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital
- Oncology & Radiotherapy Department, Zhejiang Hospital
| | - Yajun Wu
- Department of TCM Pharmacy, Zhejiang Hospital
| | - Xinyuan Zhao
- Department of Occupational Medicine and Environmental Toxicology, School of Public Health, Nantong University
| | - Zhibing Wu
- Zhejiang Key Laboratory of Geriatrics and Geriatrics Institute of Zhejiang Province, Zhejiang Hospital
- Oncology & Radiotherapy Department, Zhejiang Hospital
| |
Collapse
|
18
|
Fang LR, Wang YH, Xiong ZZ, Wang YM. Research progress of nanomaterials in tumor-targeted drug delivery and imaging therapy. OPENNANO 2023; 14:100184. [DOI: 10.1016/j.onano.2023.100184] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
19
|
Liu S, Wang Z, Wei Q, Duan X, Liu Y, Wu M, Ding J. Biomaterials-enhanced bioactive agents to efficiently block spinal metastases of cancers. J Control Release 2023; 363:721-732. [PMID: 37741462 DOI: 10.1016/j.jconrel.2023.09.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 09/12/2023] [Accepted: 09/20/2023] [Indexed: 09/25/2023]
Abstract
The spine is the most common site of bone metastases, as 20%-40% of cancer patients suffer from spinal metastases. Treatments for spinal metastases are scarce and palliative, primarily aiming at relieving bone pain and preserving neurological function. The bioactive agents-mediated therapies are the most effective modalities for treating spinal metastases because they achieve systematic and specific tumor regression. However, the clinical applications of some bioactive agents are limited due to the lack of targeting capabilities, severe side effects, and vulnerability of drug resistance. Fortunately, advanced biomaterials have been developed as excipients to enhance these treatments, including chemotherapy, phototherapy, magnetic hyperthermia therapy, and combination therapy, by improving tumor targeting and enabling sustaining and stimuli-responsive release of various therapeutic agents. Herein, the review summarizes the development of biomaterials-mediated bioactive agents for enhanced treatments of spinal metastases and predicts future research trends.
Collapse
Affiliation(s)
- Shixian Liu
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, PR China; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China
| | - Zhonghan Wang
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, PR China
| | - Qi Wei
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China; Department of Thoracic Surgery, China-Japan Union Hospital of Jilin University, 126 Xiantai Street, Changchun 130033, PR China
| | - Xuefeng Duan
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, PR China; Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China
| | - Yang Liu
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China
| | - Minfei Wu
- Department of Orthopedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, PR China.
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 5625 Renmin Street, Changchun 130022, PR China; Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, 388 Yuhangtang Road, Hangzhou 310058, PR China.
| |
Collapse
|
20
|
Li D, Ren T, Wang X, Xiao Z, Sun G, Zhang N, Zhao L, Zhong R. Development and in vitro evaluation of carmustine delivery platform: A hypoxia-sensitive anti-drug resistant nanomicelle with BBB penetrating ability. Biomed Pharmacother 2023; 167:115631. [PMID: 37804814 DOI: 10.1016/j.biopha.2023.115631] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/29/2023] [Accepted: 10/03/2023] [Indexed: 10/09/2023] Open
Abstract
Glioma is extremely difficult to be completely excised by surgery due to its invasive nature. Thus, chemotherapy still is the mainstay in the treatment of glioma after surgery. However, the natural blood-brain barrier (BBB) greatly restricts the penetration of chemotherapeutic agents into the central nervous system. As a front-line anti-glioma agent in clinical, carmustine (BCNU) exerts antitumor effect by inducing DNA damage at the O6 position of guanine. However, the therapeutic effect of BCNU was largely decreased because of the drug resistance mediated by O6-alkylguanine-DNA alkyltransferase (AGT) and insufficient local drug concentrations. To overcome these obstacles, we synthesized a BCNU-loaded hypoxia-responsive nano-micelle with BBB penetrating capacity and AGT inhibitory activity, named as T80-HA-AZO-BG/BCNU NPs. In this nano-system, Tween 80 (T80) serves as a functional coating on the surface of the micelle, promoting transportation across the BBB. Hyaluronic acid (HA) with active tumor-targeting capability was linked with the hydrophobic O6-benzylguanine (BG) analog via a hypoxia-sensitive azo bond. Under hypoxic tumor microenvironment, the azo bond selectively breaks to release O6-BG as AGT inhibitor and BCNU as DNA alkylating agent. The synthesized T80-HA-AZO-BG/BCNU NPs showed good stability, favorable biocompatibility and hypoxia-responsive drug-releasing ability. T80 modification improved the transportation of the micelle across an in vitro BBB model. Moreover, T80-HA-AZO-BG/BCNU NPs exhibited significantly enhanced cytotoxicity against glioma cell lines with high AGT expression compared with traditional combined medication of BCNU plus O6-BG. We expect that the tumor-targeting nano-micelle designed for chloroethylnitrosourea will provide new tools for the development of effective glioma therapy.
Collapse
Affiliation(s)
- Duo Li
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Ting Ren
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Xiaoli Wang
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Zhixuan Xiao
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Guohui Sun
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Na Zhang
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| | - Lijiao Zhao
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China.
| | - Rugang Zhong
- Beijing Key Laboratory of Environmental & Viral Oncology, Faculty of Environment & Life, Beijing University of Technology, Beijing 100124, China
| |
Collapse
|
21
|
Li M, Ding C, Zhang D, Chen W, Yan Z, Chen Z, Guo Z, Guo L, Huang Y. Distinguishable Colorimetric Biosensor for Diagnosis of Prostate Cancer Bone Metastases. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303159. [PMID: 37840414 PMCID: PMC10646272 DOI: 10.1002/advs.202303159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 08/08/2023] [Indexed: 10/17/2023]
Abstract
Castration-resistant prostate cancer (PCa) causes severe bone metastasis (BM), which significantly increases mortality in men with PCa. Imaging tests and radiometric scanning require long analysis times, expensive equipment, specialized personnel, and a slow turnaround. New visualization technologies are expected to solve the above problems. Nonetheless, existing visualization techniques barely meet the urgency for precise diagnosis because the human eyes cannot recognize and capture even slight variations in visual information. By using dye differentiated superposition enhancement colorimetric biosensors, an effective method to diagnose prostate cancer bone metastases (PCa-BM) with excellent accuracy for naked-eye quantitative detection of alkaline phosphatase (ALP) is developed. The biomarker ALP specific hydrolytic product ascorbic acid can be detected by rhodamine derivatives (Rd) as gold nanobipyramids (Au NBPs) are deposited and grown. Color-recombining enhancement effects between Rd and Au NBPs significantly improved abundance. The 150 U L-1 threshold between normal and abnormal can be identified by color. And with color enhancement effect and double signal response, the ALP index is visually measured to diagnose PCa-BM and provide handy treatment recommendations. Additionally, the proposed colorimetric sensing strategy can be used to diagnose other diseases.
Collapse
Affiliation(s)
- Ming Li
- Department of Urology & NephrologyThe First Affiliated Hospital of Ningbo University59 Liuting StreetNingboZhejiang315010China
- College of Material Chemistry and Chemical EngineeringKey Laboratory of Organosilicon Chemistry and Material TechnologyMinistry of EducationKey Laboratory of Organosilicon Material Technology of Zhejiang ProvinceDepartment Hangzhou Normal UniversityHangzhouZhejiang311121China
| | - Caiping Ding
- College of Material Chemistry and Chemical EngineeringKey Laboratory of Organosilicon Chemistry and Material TechnologyMinistry of EducationKey Laboratory of Organosilicon Material Technology of Zhejiang ProvinceDepartment Hangzhou Normal UniversityHangzhouZhejiang311121China
| | - Dong Zhang
- Department of Urology & NephrologyThe First Affiliated Hospital of Ningbo University59 Liuting StreetNingboZhejiang315010China
| | - Weiwei Chen
- College of Material Chemistry and Chemical EngineeringKey Laboratory of Organosilicon Chemistry and Material TechnologyMinistry of EducationKey Laboratory of Organosilicon Material Technology of Zhejiang ProvinceDepartment Hangzhou Normal UniversityHangzhouZhejiang311121China
| | - Zejun Yan
- Department of Urology & NephrologyThe First Affiliated Hospital of Ningbo University59 Liuting StreetNingboZhejiang315010China
| | - Zikang Chen
- College of Material Chemistry and Chemical EngineeringKey Laboratory of Organosilicon Chemistry and Material TechnologyMinistry of EducationKey Laboratory of Organosilicon Material Technology of Zhejiang ProvinceDepartment Hangzhou Normal UniversityHangzhouZhejiang311121China
| | - Zhiyong Guo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐productsState Key Laboratory Base of Novel Functional Materials and Preparation ScienceSchool of Materials Science and Chemical EngineeringNingbo UniversityNingboZhejiang315211China
| | - Longhua Guo
- College of BiologicalChemical Sciences and EngineeringJiaxing UniversityJiaxingZhejiang314001China
| | - Youju Huang
- College of Material Chemistry and Chemical EngineeringKey Laboratory of Organosilicon Chemistry and Material TechnologyMinistry of EducationKey Laboratory of Organosilicon Material Technology of Zhejiang ProvinceDepartment Hangzhou Normal UniversityHangzhouZhejiang311121China
| |
Collapse
|
22
|
Karmakar R, Dey S, Alam A, Khandelwal M, Pati F, Rengan AK. Attributes of Nanomaterials and Nanotopographies for Improved Bone Tissue Engineering and Regeneration. ACS APPLIED BIO MATERIALS 2023; 6:4020-4041. [PMID: 37691480 DOI: 10.1021/acsabm.3c00549] [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] [Indexed: 09/12/2023]
Abstract
Bone tissue engineering (BTE) is a multidisciplinary area that can solve the limitation of conventional grafting methods by developing viable and biocompatible bone replacements. The three essential components of BTE, i.e., Scaffold material and Cells and Growth factors altogether, facilitate support and guide for bone formation, differentiation of the bone tissues, and enhancement in the cellular activities and bone regeneration. However, there is a scarcity of the appropriate materials that can match the mechanical property as well as functional similarity to native tissue, considering the bone as hard tissue. In such scenarios, nanotechnology can be leveraged upon to achieve the desired aspects of BTE, and that is the key point of this review article. This review article examines the significant areas of nanotechnology research that have an impact on regeneration of bone: (a) scaffold with nanomaterials helps to enhance physicochemical interactions, biocompatibility, mechanical stability, and attachment; (b) nanoparticle-based approaches for delivering bioactive chemicals, growth factors, and genetic material. The article begins with the introduction of components and healing mechanisms of bone and the factors associated with them. The focus of this article is on the various nanotopographies that are now being used in scaffold formation, by describing how they are made, and how these nanotopographies affect the immune system and potential underlying mechanisms. The advantages of 4D bioprinting in BTE by using nanoink have also been mentioned. Additionally, we have investigated the importance of an in silico approach for finding the interaction between drugs and their related receptors, which can help to formulate suitable systems for delivery. This review emphasizes the role of nanoscale approach and how it helps to increase the efficacy of parameters of scaffold as well as drug delivery system for tissue engineering and bone regeneration.
Collapse
Affiliation(s)
- Rounik Karmakar
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Sreenath Dey
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Aszad Alam
- Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology, Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Mudrika Khandelwal
- Department of Materials Science and Metallurgical Engineering, Indian Institute of Technology, Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology (IIT), Hyderabad, Kandi-502285, Sangareddy, Telangana, India
| |
Collapse
|
23
|
Shamsipur M, Ghavidast A, Pashabadi A. Phototriggered structures: Latest advances in biomedical applications. Acta Pharm Sin B 2023; 13:2844-2876. [PMID: 37521863 PMCID: PMC10372844 DOI: 10.1016/j.apsb.2023.04.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 03/12/2023] [Accepted: 04/11/2023] [Indexed: 08/01/2023] Open
Abstract
Non-invasive control of the drug molecules accessibility is a key issue in improving diagnostic and therapeutic procedures. Some studies have explored the spatiotemporal control by light as a peripheral stimulus. Phototriggered drug delivery systems (PTDDSs) have received interest in the past decade among biological researchers due to their capability the control drug release. To this end, a wide range of phototrigger molecular structures participated in the DDSs to serve additional efficiency and a high-conversion release of active fragments under light irradiation. Up to now, several categories of PTDDSs have been extended to upgrade the performance of controlled delivery of therapeutic agents based on well-known phototrigger molecular structures like o-nitrobenzyl, coumarinyl, anthracenyl, quinolinyl, o-hydroxycinnamate and hydroxyphenacyl, where either of one endows an exclusive feature and distinct mechanistic approach. This review conveys the design, photochemical properties and essential mechanism of the most important phototriggered structures for the release of single and dual (similar or different) active molecules that have the ability to quickly reason of the large variety of dynamic biological phenomena for biomedical applications like photo-regulated drug release, synergistic outcomes, real-time monitoring, and biocompatibility potential.
Collapse
|
24
|
Wang X, Shi G, Fan S, Ma J, Yan Y, Wang M, Tang X, Lv P, Zhang Y. Targeted delivery of food functional ingredients in precise nutrition: design strategy and application of nutritional intervention. Crit Rev Food Sci Nutr 2023; 64:7854-7877. [PMID: 36999956 DOI: 10.1080/10408398.2023.2193275] [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] [Indexed: 04/01/2023]
Abstract
With the high incidence of chronic diseases, precise nutrition is a safe and efficient nutritional intervention method to improve human health. Food functional ingredients are an important material base for precision nutrition, which have been researched for their application in preventing diseases and improving health. However, their poor solubility, stability, and bad absorption largely limit their effect on nutritional intervention. The establishment of a stable targeted delivery system is helpful to enhance their bioavailability, realize the controlled release of functional ingredients at the targeted action sites in vivo, and provide nutritional intervention approaches and methods for precise nutrition. In this review, we summarized recent studies about the types of targeted delivery systems for the delivery of functional ingredients and their digestion fate in the gastrointestinal tract, including emulsion-based delivery systems and polymer-based delivery systems. The building materials, structure, size and charge of the particles in these delivery systems were manipulated to fabricate targeted carriers. Finally, the targeted delivery systems for food functional ingredients have gained some achievements in nutritional intervention for inflammatory bowel disease (IBD), liver disease, obesity, and cancer. These findings will help in designing fine targeted delivery systems, and achieving precise nutritional intervention for food functional ingredients on human health.
Collapse
Affiliation(s)
- Xu Wang
- Hebei Food Inspection and Research Institute, Hebei Food Safety Key Laboratory, Key Laboratory of Special Food Supervision Technology for State Market Regulation, Hebei Engineering Research Center for Special Food Safety and Health, Shijiazhuang, China
- Department of Cell Biology, Cardiovascular Medical Science Center, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Guohua Shi
- Hebei Food Inspection and Research Institute, Hebei Food Safety Key Laboratory, Key Laboratory of Special Food Supervision Technology for State Market Regulation, Hebei Engineering Research Center for Special Food Safety and Health, Shijiazhuang, China
| | - Sufang Fan
- Hebei Food Inspection and Research Institute, Hebei Food Safety Key Laboratory, Key Laboratory of Special Food Supervision Technology for State Market Regulation, Hebei Engineering Research Center for Special Food Safety and Health, Shijiazhuang, China
| | - Junmei Ma
- Hebei Food Inspection and Research Institute, Hebei Food Safety Key Laboratory, Key Laboratory of Special Food Supervision Technology for State Market Regulation, Hebei Engineering Research Center for Special Food Safety and Health, Shijiazhuang, China
| | - Yonghuan Yan
- Hebei Food Inspection and Research Institute, Hebei Food Safety Key Laboratory, Key Laboratory of Special Food Supervision Technology for State Market Regulation, Hebei Engineering Research Center for Special Food Safety and Health, Shijiazhuang, China
- School of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Mengtian Wang
- Hebei Food Inspection and Research Institute, Hebei Food Safety Key Laboratory, Key Laboratory of Special Food Supervision Technology for State Market Regulation, Hebei Engineering Research Center for Special Food Safety and Health, Shijiazhuang, China
- School of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| | - Xiaozhi Tang
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Key Laboratory of Grains and Oils Quality Control and Processing, Nanjing University of Finance and Economics, Nanjing, China
| | - Pin Lv
- Department of Cell Biology, Cardiovascular Medical Science Center, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, China
| | - Yan Zhang
- Hebei Food Inspection and Research Institute, Hebei Food Safety Key Laboratory, Key Laboratory of Special Food Supervision Technology for State Market Regulation, Hebei Engineering Research Center for Special Food Safety and Health, Shijiazhuang, China
- School of Forensic Medicine, Hebei Key Laboratory of Forensic Medicine, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
25
|
Negut I, Bita B. Polymeric Micellar Systems-A Special Emphasis on "Smart" Drug Delivery. Pharmaceutics 2023; 15:976. [PMID: 36986837 PMCID: PMC10056703 DOI: 10.3390/pharmaceutics15030976] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
Concurrent developments in anticancer nanotechnological treatments have been observed as the burden of cancer increases every year. The 21st century has seen a transformation in the study of medicine thanks to the advancement in the field of material science and nanomedicine. Improved drug delivery systems with proven efficacy and fewer side effects have been made possible. Nanoformulations with varied functions are being created using lipids, polymers, and inorganic and peptide-based nanomedicines. Therefore, thorough knowledge of these intelligent nanomedicines is crucial for developing very promising drug delivery systems. Polymeric micelles are often simple to make and have high solubilization characteristics; as a result, they seem to be a promising alternative to other nanosystems. Even though recent studies have provided an overview of polymeric micelles, here we included a discussion on the "intelligent" drug delivery from these systems. We also summarized the state-of-the-art and the most recent developments of polymeric micellar systems with respect to cancer treatments. Additionally, we gave significant attention to the clinical translation potential of polymeric micellar systems in the treatment of various cancers.
Collapse
Affiliation(s)
- Irina Negut
- National Institute for Laser, Plasma and Radiation Physics, 409 Atomistilor Street, Magurele, 077125 Bucharest, Romania
| | - Bogdan Bita
- National Institute for Laser, Plasma and Radiation Physics, 409 Atomistilor Street, Magurele, 077125 Bucharest, Romania
- Faculty of Physics, University of Bucharest, 077125 Măgurele, Romania
| |
Collapse
|
26
|
Wang Q, Atluri K, Tiwari AK, Babu RJ. Exploring the Application of Micellar Drug Delivery Systems in Cancer Nanomedicine. Pharmaceuticals (Basel) 2023; 16:ph16030433. [PMID: 36986532 PMCID: PMC10052155 DOI: 10.3390/ph16030433] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/07/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
Various formulations of polymeric micelles, tiny spherical structures made of polymeric materials, are currently being investigated in preclinical and clinical settings for their potential as nanomedicines. They target specific tissues and prolong circulation in the body, making them promising cancer treatment options. This review focuses on the different types of polymeric materials available to synthesize micelles, as well as the different ways that micelles can be tailored to be responsive to different stimuli. The selection of stimuli-sensitive polymers used in micelle preparation is based on the specific conditions found in the tumor microenvironment. Additionally, clinical trends in using micelles to treat cancer are presented, including what happens to micelles after they are administered. Finally, various cancer drug delivery applications involving micelles are discussed along with their regulatory aspects and future outlooks. As part of this discussion, we will examine current research and development in this field. The challenges and barriers they may have to overcome before they can be widely adopted in clinics will also be discussed.
Collapse
Affiliation(s)
- Qi Wang
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
| | - Keerthi Atluri
- Product Development Department, Alcami Corporation, Morrisville, NC 27560, USA
| | - Amit K. Tiwari
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
- Department of Cell and Cancer Biology, University of Toledo, Toledo, OH 43614, USA
| | - R. Jayachandra Babu
- Department of Drug Discovery and Development, Auburn University, Auburn, AL 36849, USA
- Correspondence:
| |
Collapse
|
27
|
Zhang X, Xu X, Wang X, Lin Y, Zheng Y, Xu W, Liu J, Xu W. Hepatoma-targeting and reactive oxygen species-responsive chitosan-based polymeric micelles for delivery of celastrol. Carbohydr Polym 2023; 303:120439. [PMID: 36657834 DOI: 10.1016/j.carbpol.2022.120439] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/01/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022]
Abstract
A glycyrrhetinic acid-modified carboxymethyl chitosan-thioketal-rhein (GCTR) conjugate was designed and synthesized for the in vivo delivery of celastrol (Cela). Cela was encapsulated into polymeric micelles (PMs) formed by GCTR conjugates self-assembly in water to form Cela/GCTR PMs with high drug loading capacity and small particle size. Cela/GCTR PMs had a sustained-release characteristic in the blood environment and a rapid-release feature in the tumor microenvironment. Cela/GCTR PMs had a significant proliferation inhibitory effect on HepG2 and BEL-7402 cells, but a negligible impact on L-02 cells at low concentrations. Cela/GCTR PMs possessed reactive oxygen species (ROS)-responsive properties in vitro and in cells, could improve the bioavailability of Cela, and exert remarkable hepatoma-targeting properties. Cela/GCTR PMs could also effectively inhibit tumor growth with no apparent damage to different organs. In summary, GCTR PMs with good ROS-responsive and hepatoma-targeting properties are expected to be possible delivery carriers for hydrophobic antineoplastic drugs for hepatocellular carcinoma therapy.
Collapse
Affiliation(s)
- Xue Zhang
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Xueya Xu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Xiaoying Wang
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| | - Yajuan Lin
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Yaling Zheng
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Wen Xu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Jian Liu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Wei Xu
- Pharmacy College, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| |
Collapse
|
28
|
Yang SB, Lee DN, Lee JH, Seo M, Shin DW, Lee S, Lee YH, Park J. Design and Evaluation of a Carrier-Free Prodrug-Based Palmitic-DEVD-Doxorubicin Conjugate for Targeted Cancer Therapy. Bioconjug Chem 2023; 34:333-344. [PMID: 36735902 DOI: 10.1021/acs.bioconjchem.2c00490] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In the development of new drugs, typical polymer- or macromolecule-based nanocarriers suffer from manufacturing process complexity, unwanted systematic toxicity, and low loading capacity. However, carrier-free nanomedicines have made outstanding progress in drug delivery and pharmacokinetics, demonstrating most of the advantages associated with nanoparticles when applied in targeted anticancer therapy. Here, to overcome the problems of nanocarriers and conventional cytotoxic drugs, we developed a novel, carrier-free, self-assembled prodrug consisting of a hydrophobic palmitic (16-carbon chain n-hexadecane chain) moiety and hydrophilic group (or moiety) which is included in a caspase-3-specific cleavable peptide (Asp-Glu-Val-Asp, DEVD) and a cytotoxic drug (doxorubicin, DOX). The amphiphilic conjugate, the palmitic-DEVD-DOX, has the ability to self-assemble into nanoparticles in saline without the need for any carriers or nanoformulations. Additionally, the inclusion of doxorubicin is in its prodrug form and the apoptosis-specific DEVD peptide lead to the reduced side effects of doxorubicin in normal tissue. Furthermore, the carrier-free palmitic-DEVD-DOX nanoparticles could passively accumulate in the tumor tissues of tumor-bearing mice due to an enhanced permeation and retention (EPR) effect. As a result, the palmitic-DEVD-DOX conjugate showed an enhanced therapeutic effect compared with the unmodified DEVD-DOX conjugate. Therefore, this carrier-free palmitic-DEVD-DOX prodrug has great therapeutic potential to treat solid tumors, overcoming the problems of conventional chemotherapy and nanoparticles.
Collapse
Affiliation(s)
- Seong-Bin Yang
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
| | - Dong-Nyeong Lee
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
| | - Jun-Hyuck Lee
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Minho Seo
- Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Dong Wook Shin
- College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea
| | - Seokwoo Lee
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Young-Ho Lee
- Research Center for Bioconvergence Analysis, Korea Basic Science Institute (KBSI), Ochang, Chungbuk 28119, Republic of Korea.,Bio-Analytical Science, University of Science and Technology, Daejeon 34113, Republic of Korea.,Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Jooho Park
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea.,Department of Biomedical Chemistry, College of Biomedical and Health Science, Konkuk University, Chungju 27478, Republic of Korea
| |
Collapse
|
29
|
Shen Y, Zou Y, Bie B, Dong C, Lv Y. Combining dual-targeted liquid metal nanoparticles with autophagy activation and mild photothermal therapy to treat metastatic breast cancer and inhibit bone destruction. Acta Biomater 2023; 157:578-592. [PMID: 36442822 DOI: 10.1016/j.actbio.2022.11.044] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/26/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Although mild photothermal therapy (mild-PTT) avoids treatment bottleneck of the traditional PTT, the application of mild-PTT in deep and internal tumors is severely restricted due to thermal resistance, limited irradiation area and penetration depth. In addition, bone resorption caused by tumor colonization in distal bone tissue exacerbates tumor progression. Here, a strategy was developed for the treatment of bone metastasis and alleviation of bone resorption, which was based on liquid metal (LM) nanoparticle to resist thermal resistance induced by mild-PTT via autophagy activation. Briefly, LM and autophagy activator (Curcumin, Cur) were loaded into zeolitic imidazolate framework-8 (ZIF-8), which was then functionalized with hyaluronic acid/alendronate (CLALN). CLALN exhibited good photothermal performance, drug release ability under acidic environment, specifical recognition and aggregation at bone metastasis sites. CLALN combined with mild-PPT dramatically inhibited tumor progress by inducing the impaired autophagy and reduced the expression of programmed cell death ligand 1 (PD-L1) protein triggered by mild-PTT, resisting thermal resistance and alleviating the immunosuppression. Besides, CLALN combined with mild-PPT effectively alleviated osteolysis compared with only CLALN or mild-PPT. Our experiments demonstrated that this multi-functional LM-based nanoparticle combined with autophagy activation provided a promising therapeutic strategy for bone metastasis treatment. STATEMENT OF SIGNIFICANCE: Due to the limited light penetration, photothermal therapy (PTT) has limited inhibitory effect on tumor cells colonized in the bone. In addition, nonspecific heat diffusion of PTT may accidentally burn normal tissues and damage peripheral blood vessels, which can block the accumulation of drugs in deep tumors. Here, a multifunctional liquid metal based mild-PTT delivery system is designed to inhibit tumor growth and bone resorption by modulating the bone microenvironment and activating autophagy "on demand". It can overcome the treatment bottleneck of traditional PTT and improve the treatment effect of mild-PTT by resisting photothermal resistance and immune suppression. In addition, it also exhibits favorable heat/acid-responsive drug release performance and can specifically target tumor cells at the site of bone metastases.
Collapse
Affiliation(s)
- Yaping Shen
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Yang Zou
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, No. 1 Sunshine Avenue, Jiangxia District, Wuhan, Hubei 430200, PR China; College of Environmental Engineering, Wuhan Textile University, Wuhan 430200, PR China
| | - Binglin Bie
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, No. 1 Sunshine Avenue, Jiangxia District, Wuhan, Hubei 430200, PR China
| | - Chanjuan Dong
- Mechanobiology and Regenerative Medicine Laboratory, Bioengineering College, Chongqing University, Chongqing 400044, PR China
| | - Yonggang Lv
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, No. 1 Sunshine Avenue, Jiangxia District, Wuhan, Hubei 430200, PR China.
| |
Collapse
|
30
|
Yu J, Wang L, Xie X, Zhu W, Lei Z, Lv L, Yu H, Xu J, Ren J. Multifunctional Nanoparticles Codelivering Doxorubicin and Amorphous Calcium Carbonate Preloaded with Indocyanine Green for Enhanced Chemo-Photothermal Cancer Therapy. Int J Nanomedicine 2023; 18:323-337. [PMID: 36700147 PMCID: PMC9869790 DOI: 10.2147/ijn.s394896] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 01/07/2023] [Indexed: 01/20/2023] Open
Abstract
Background Multifunctional stimuli-responsive nanoparticles with photothermal-chemotherapy provided a powerful tool for improving the accuracy and efficiency in the treatment of malignant tumors. Methods Herein, photosensitizer indocyanine green (ICG)-loaded amorphous calcium-carbonate (ICG@) nanoparticle was prepared by a gas diffusion reaction. Doxorubicin (DOX) and ICG@ were simultaneously encapsulated into poly(lactic-co-glycolic acid)-ss-chondroitin sulfate A (PSC) nanoparticles by a film hydration method. The obtained PSC/ICG@+DOX hybrid nanoparticles were characterized and evaluated by Fourier transform infrared spectroscopy (FTIR), dynamic light scattering (DLS), transmission electron microscopy (TEM), and differential scanning calorimetry (DSC). The cellular uptake and cytotoxicity of PSC/ICG@+DOX nanoparticles were analyzed by confocal laser scanning microscopy (CLSM) and MTT assay in 4T1 cells. In vivo antitumor activity of the nanoparticles was evaluated in 4T1-bearing Balb/c mice. Results PSC/ICG@+DOX nanoparticles were nearly spherical in shape by TEM observation, and the diameter was 407 nm determined by DLS. Owing to calcium carbonate and disulfide bond linked copolymer, PSC/ICG@+DOX nanoparticles exhibited pH and reduction-sensitive drug release. Further, PSC/ICG@+DOX nanoparticles showed an effective photothermal effect under near-infrared (NIR) laser irradiation, and improved cellular uptake and cytotoxicity in breast cancer 4T1 cells. Importantly, PSC/ICG@+DOX nanoparticles demonstrated the most effective suppression of tumor growth in orthotopic 4T1-bearing mice among the treatment groups. In contrast with single chemotherapy or photothermal therapy, chemo-photothermal treatment by PSC/ICG@+DOX nanoparticles synergistically inhibited the growth of 4T1 cells. Conclusion This study demonstrated that PSC/ICG@+DOX nanoparticles with active targeting and stimuli-sensitivity would be a promising strategy to enhance chemo-photothermal cancer therapy.
Collapse
Affiliation(s)
- Jingmou Yu
- Huzhou Key Laboratory of Medical and Environmental Applications Technologies, School of Life Sciences, Huzhou University, Huzhou, People’s Republic of China,Jiangxi Provincial Laboratory Laboratory of System Biomedicine, Jiujiang University, Jiujiang, People’s Republic of China,School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China,Correspondence: Jingmou Yu; Jin Ren, Email ;
| | - Liangliang Wang
- Affiliated Hospital of Jiujiang University, Jiujiang, People’s Republic of China
| | - Xin Xie
- Jiangxi Provincial Laboratory Laboratory of System Biomedicine, Jiujiang University, Jiujiang, People’s Republic of China
| | - Wenjing Zhu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| | - Zhineng Lei
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| | - Linghui Lv
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| | - Hongling Yu
- School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| | - Jing Xu
- Affiliated Hospital of Jiujiang University, Jiujiang, People’s Republic of China
| | - Jin Ren
- Jiangxi Provincial Laboratory Laboratory of System Biomedicine, Jiujiang University, Jiujiang, People’s Republic of China,School of Pharmacy and Life Sciences, Jiujiang University, Jiujiang, People’s Republic of China
| |
Collapse
|
31
|
Yang DC, Yang XZ, Luo CM, Wen LF, Liu JY, Lin Z. A promising strategy for synergistic cancer therapy by integrating a photosensitizer into a hypoxia-activated prodrug. Eur J Med Chem 2022; 243:114749. [PMID: 36115207 DOI: 10.1016/j.ejmech.2022.114749] [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: 06/02/2022] [Revised: 08/28/2022] [Accepted: 09/03/2022] [Indexed: 11/24/2022]
Abstract
Herein, we fabricate a multifunctional molecular prodrug BAC where the chemotherapeutical agent camptothecin (CPT) is linked with a boron dipyrromethene (BODIPY)-based photosensitizer by an azobenzene chain which is sensitive to over-expressed azoreductase in hypoxic tumor cells. This prodrug was further loaded into biodegradable monomethoxy poly(ethylene glycol)-b-poly(caprolactone) (mPEG-b-PCL) to improve its solubility and tumor accumulation. The formed BAC nanoparticles (BAC NPs) can destroy aerobic tumor cells with relatively short distance from blood vessels by photodynamic therapy (PDT) under illumination. The PDT action inevitably leads to consumption of O2, and subsequently acute hypoxia which can induce cleavage of azobenzene linkage to boost release of CPT killing the other hypoxic interior tumor cells survived from PDT. Both in vitro and in vivo studies have verified that BAC NPs possess remarkable antitumor activity by a synergistic action of PDT and chemotherapy.
Collapse
Affiliation(s)
- De-Chao Yang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Xiao-Zhen Yang
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Cheng-Miao Luo
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Lin-Feng Wen
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Jian-Yong Liu
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350108, China; Key Laboratory of Molecule Synthesis and Function Discovery, Fujian Province University, College of Chemistry, Fuzhou University, Fuzhou, 350108, China; State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou, 350108, China.
| | - Zhonghui Lin
- National & Local Joint Biomedical Engineering Research Center on Photodynamic Technologies, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| |
Collapse
|
32
|
Guo Y, Fan Y, Wang Z, Li G, Zhan M, Gong J, Majoral JP, Shi X, Shen M. Chemotherapy Mediated by Biomimetic Polymeric Nanoparticles Potentiates Enhanced Tumor Immunotherapy via Amplification of Endoplasmic Reticulum Stress and Mitochondrial Dysfunction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2206861. [PMID: 36125843 DOI: 10.1002/adma.202206861] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/11/2022] [Indexed: 06/15/2023]
Abstract
Construction of multifunctional nanoplatforms to elevate chemotherapeutic efficacy and induce long-term antitumor immunity still remains to be an extreme challenge. Herein, the design of an advanced redox-responsive nanomedicine formulation based on phosphorus dendrimer-copper(II) complexes (1G3 -Cu)- and toyocamycin (Toy)-loaded polymeric nanoparticles (GCT NPs) coated with cancer cell membranes (CM) are reported. The designed GCT@CM NPs with a size of 210 nm are stable under physiological conditions but are rapidly dissociated in the reductive tumor microenvironment to deplete glutathione and release drugs. The co-loading of 1G3 -Cu and Toy within the NPs causes significant tumor cell apoptosis and immunogenic cell death through 1G3 -Cu-induced mitochondrial dysfunction and Toy-mediated amplification of endoplasmic reticulum stress, respectively, thus effectively suppressing tumor growth, promoting dendritic cell maturation, and increasing tumor-infiltrating cytotoxic T lymphocytes. Likewise, the coated CM and the loaded 1G3 -Cu render the GCT@CM NPs with homotypic targeting and T1 -weighted magnetic resonance imaging of tumors, respectively. With the assistance of programmed cell death ligand 1 antibody, the GCT@CM NP-mediated chemotherapy can significantly potentiate tumor immunotherapy for effective inhibition of tumor recurrence and metastasis. The developed GCT@CM NPs hold a great potential for chemotherapy-potentiated immunotherapy of different tumor types through different mechanisms or synergies.
Collapse
Affiliation(s)
- Yunqi Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Yu Fan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Zhiqiang Wang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Gaoming Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Mengsi Zhan
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Junli Gong
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | | | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| | - Mingwu Shen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, P. R. China
| |
Collapse
|
33
|
Tian H, Zhang T, Qin S, Huang Z, Zhou L, Shi J, Nice EC, Xie N, Huang C, Shen Z. Enhancing the therapeutic efficacy of nanoparticles for cancer treatment using versatile targeted strategies. J Hematol Oncol 2022; 15:132. [PMID: 36096856 PMCID: PMC9469622 DOI: 10.1186/s13045-022-01320-5] [Citation(s) in RCA: 166] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/20/2022] [Indexed: 12/24/2022] Open
Abstract
Poor targeting of therapeutics leading to severe adverse effects on normal tissues is considered one of the obstacles in cancer therapy. To help overcome this, nanoscale drug delivery systems have provided an alternative avenue for improving the therapeutic potential of various agents and bioactive molecules through the enhanced permeability and retention (EPR) effect. Nanosystems with cancer-targeted ligands can achieve effective delivery to the tumor cells utilizing cell surface-specific receptors, the tumor vasculature and antigens with high accuracy and affinity. Additionally, stimuli-responsive nanoplatforms have also been considered as a promising and effective targeting strategy against tumors, as these nanoplatforms maintain their stealth feature under normal conditions, but upon homing in on cancerous lesions or their microenvironment, are responsive and release their cargoes. In this review, we comprehensively summarize the field of active targeting drug delivery systems and a number of stimuli-responsive release studies in the context of emerging nanoplatform development, and also discuss how this knowledge can contribute to further improvements in clinical practice.
Collapse
Affiliation(s)
- Hailong Tian
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Tingting Zhang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Siyuan Qin
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Zhao Huang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jiayan Shi
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, 3800, VIC, Australia
| | - Edouard C Nice
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China
| | - Na Xie
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China. .,West China School of Basic Medical Sciences and Forensic Medicine, Sichuan university, Chengdu, 610041, China.
| | - Canhua Huang
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China. .,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, 315040, Ningbo, Zhejiang, China.
| |
Collapse
|
34
|
Hu B, Zhang Y, Zhang G, Li Z, Jing Y, Yao J, Sun S. Research progress of bone-targeted drug delivery system on metastatic bone tumors. J Control Release 2022; 350:377-388. [PMID: 36007681 DOI: 10.1016/j.jconrel.2022.08.034] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 10/15/2022]
Abstract
Bone metastases are common in malignant tumors and the effect of conventional treatment is limited. How to effectively inhibit tumor bone metastasis and deliver the drug to the bone has become an urgent issue to be solved. While bone targeting drug delivery systems have obvious advantages in the treatment of bone tumors. The research on bone-targeted anti-tumor therapy has made significant progress in recent years. We introduced the related tumor pathways of bone metastases. The tumor microenvironment plays an important role in metastatic bone tumors. We introduce a drug-loading systems based on different environment-responsive nanocomposites for anti-tumor and anti-metastatic research. According to the process of bone metastases and the structure of bone tissue, we summarized the information on bone-targeting molecules. Bisphosphate has become the first choice of bone-targeted drug delivery carrier because of its affinity with hydroxyapatite in bone. Therefore, we sought to summarize the bone-targeting molecule of bisphosphate to identify the modification effect on bone-targeting. And this paper discusses the relationship between bisphosphate bone targeting molecular structure and drug delivery carriers, to provide some new ideas for the research and development of bone-targeting drug delivery carriers. Targeted therapy will make a more outstanding contribution to the treatment of tumors.
Collapse
Affiliation(s)
- Beibei Hu
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China; State Key Laboratory Breeding Base-Hebei Province, Key Laboratory of Molecular Chemistry for Drug, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Yongkang Zhang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Guogang Zhang
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Zhongqiu Li
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Yongshuai Jing
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China
| | - Jun Yao
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China.
| | - Shiguo Sun
- College of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, 26 Yuxiang Road, Shijiazhuang 050018, PR China.
| |
Collapse
|
35
|
Liu T, Lang M. Preparation and characterization of novel functional tri-block copolymer for constructing temperature/redox dual-stimuli responsive micelles. JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2022. [DOI: 10.1080/10601325.2022.2092409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- Tianyue Liu
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Meidong Lang
- Key Laboratory for Ultrafine Materials of Ministry of Education, School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
36
|
Li Z, Zhang W, Zhang Z, Gao H, Qin Y. Cancer bone metastases and nanotechnology-based treatment strategies. Expert Opin Drug Deliv 2022; 19:1217-1232. [PMID: 35737871 DOI: 10.1080/17425247.2022.2093856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Bone metastases have gradually been recognized as common metastases that affect patient quality of life and survival due to the increased incidence of primary tumors. However, there is still a lack of effective clinical treatment methods for bone metastases because of their particularity and complexity. Nanomedicine provides a new strategy for the treatment of bone metastases and shows great therapeutic potential. Thus, it is important to review the latest nanomedicine treatments for bone metastases. AREAS COVERED This review introduces the mechanistic relationships of bone metastases and summarizes nanotechnology-based treatments of bone metastases according to targeting strategies. EXPERT OPINION As we start to understand the mechanisms that enable bone metastases, we can better develop nanomedicine treatments. However, many of the mechanisms behind bone metastasis remain unclear. The application of nanomedicine shows promising anti-bone metastasis efficacy and helps to explore the pathogenesis of bone metastases. The optimized construction of nanomedicine according to bone metastatic properties is crucial to ensure the desired anti-bone metastasis efficacy and good biosafety. Therefore, the transition from bench to bedside still requires continued exploration.
Collapse
Affiliation(s)
- Zhaofeng Li
- Department of Orthopedic, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, China.,Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Wei Zhang
- Department of Orthopedics, Sichuan Provincial People's Hospital & Sichuan Academy of Medical Sciences & Affiliated Hospital of University of Electronic Science and Technology, Chengdu, Sichuan, China
| | - Zhong Zhang
- Department of Orthopedic, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, China
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Yi Qin
- Department of Orthopedic, Zhuhai People's Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, Guangdong, China
| |
Collapse
|
37
|
Sun L, Zhao P, Chen M, Leng J, Luan Y, Du B, Yang J, Yang Y, Rong R. Taxanes prodrug-based nanomedicines for cancer therapy. J Control Release 2022; 348:672-691. [PMID: 35691501 DOI: 10.1016/j.jconrel.2022.06.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/04/2022] [Accepted: 06/04/2022] [Indexed: 11/16/2022]
Abstract
Malignant tumor remains a huge threat to human health and chemotherapy still occupies an important place in clinical tumor treatment. As a kind of potent antimitotic agent, taxanes act as the first-line broad-spectrum cancer drug in clinical use. However, disadvantages such as prominent hydrophobicity, severe off-target toxicity or multidrug resistance lead to unsatisfactory therapeutic effects, which restricts its wider usage. The efficient delivery of taxanes is still quite a challenge despite the rapid developments in biomaterials and nanotechnology. Great progress has been made in prodrug-based nanomedicines (PNS) for cancer therapy due to their outstanding advantages such as high drug loading efficiency, low carrier induced immunogenicity, tumor stimuli-responsive drug release, combinational therapy and so on. Based on the numerous developments in this filed, this review summarized latest updates of taxanes prodrugs-based nanomedicines (TPNS), focusing on polymer-drug conjugate-based nanoformulations, small molecular prodrug-based self-assembled nanoparticles and prodrug-encapsulated nanosystems. In addition, the new trends of tumor stimuli-responsive TPNS were also discussed. Moreover, the future challenges of TPNS for clinical translation were highlighted. We here expect this review will inspire researchers to explore more practical taxanes prodrug-based nano-delivery systems for clinical use.
Collapse
Affiliation(s)
- Linlin Sun
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Pan Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Menghan Chen
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jiayi Leng
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yixin Luan
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Baoxiang Du
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Jia Yang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China
| | - Yong Yang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, PR China.
| |
Collapse
|
38
|
Tiburcius S, Krishnan K, Jose L, Patel V, Ghosh A, Sathish CI, Weidenhofer J, Yang JH, Verrills NM, Karakoti A, Vinu A. Egg-yolk core-shell mesoporous silica nanoparticles for high doxorubicin loading and delivery to prostate cancer cells. NANOSCALE 2022; 14:6830-6845. [PMID: 35441642 DOI: 10.1039/d2nr00783e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Mesoporous silica-based nanoparticles (MSNs) have gained rapid interest as a drug delivery system (DDS) and demonstrated their versatility in delivering drugs for the treatment of various cancers. However, the drug loading efficiency of MSNs is low and is usually improved by improving textural properties through complicated synthesis methods or by post synthesis modification of the surface that can result in the loss of surface area and modify its drug release properties. In this study, we report a direct single-step synthesis of MSNs with a unique egg-yolk core-shell morphology, large pore volume and a hydrophilic surface, decorated with nitrogen rich surface functionalities for increasing its drug loading capacity. This combination of excellent textural properties and surface functionalisation was achieved by a simple soft templating method using dual surfactants and the silica sources assisted by employing either triethylamine (TEA) or triethanolamine (TEO) as the hydrolysis agent. The morphology and well-ordered mesoporous structure can simply be tuned by changing the pH of the synthesis medium that affects the self-assembly mechanism of the micelles. HRTEM image of samples clearly revealed an egg-yolk core-shell morphology with a thin mesoporous silica shell. The optimised MSN samples synthesized at a pH of 11 using either TEA or TEO depicted a higher doxorubicin (Dox) loading capacity of 425 μg mg-1 and 481 μg mg-1 respectively, as compared to only 347 μg mg-1 for MSN samples due to the uniform distribution of nitrogen functionalities. The anticancer activity of Dox loaded MSNs evaluated in two different prostate cancer cell lines (PC-3 and LNCaP) showed a higher cytotoxicity of the drug loaded on optimised MSN samples as compared to pristine MSNs without affecting the cellular uptake of the particles. These results suggest that the unique single-step synthesis and functionalisation method resulted in successfully achieving higher drug loading in egg-yolk core-shell nitrogen functionalised MSNs and could be implemented as an effective carrier of chemotherapeutic drugs.
Collapse
Affiliation(s)
- Steffi Tiburcius
- Global Innovative Centre for Advanced Nanomaterials, Faculty of Engineering and Built Environment, The University of Newcastle, Callaghan, 2308, NSW, Australia.
| | - Kannan Krishnan
- Global Innovative Centre for Advanced Nanomaterials, Faculty of Engineering and Built Environment, The University of Newcastle, Callaghan, 2308, NSW, Australia.
| | - Linta Jose
- Global Innovative Centre for Advanced Nanomaterials, Faculty of Engineering and Built Environment, The University of Newcastle, Callaghan, 2308, NSW, Australia.
| | - Vaishwik Patel
- Global Innovative Centre for Advanced Nanomaterials, Faculty of Engineering and Built Environment, The University of Newcastle, Callaghan, 2308, NSW, Australia.
| | - Arnab Ghosh
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, 2308, NSW, Australia
| | - C I Sathish
- Global Innovative Centre for Advanced Nanomaterials, Faculty of Engineering and Built Environment, The University of Newcastle, Callaghan, 2308, NSW, Australia.
| | - Judith Weidenhofer
- Hunter Medical Research Institute (HMRI), New Lambton Heights, 2305, NSW, Australia
| | - Jae-Hun Yang
- Global Innovative Centre for Advanced Nanomaterials, Faculty of Engineering and Built Environment, The University of Newcastle, Callaghan, 2308, NSW, Australia.
| | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, The University of Newcastle, Callaghan, 2308, NSW, Australia
| | - Ajay Karakoti
- Global Innovative Centre for Advanced Nanomaterials, Faculty of Engineering and Built Environment, The University of Newcastle, Callaghan, 2308, NSW, Australia.
| | - Ajayan Vinu
- Global Innovative Centre for Advanced Nanomaterials, Faculty of Engineering and Built Environment, The University of Newcastle, Callaghan, 2308, NSW, Australia.
| |
Collapse
|
39
|
Zhou J, Sun X, Zhang X, Yang H, Jiang Z, Luo Q, Liu Y, Wang G. miR-107 is involved in the regulation of NEDD9-mediated invasion and metastasis in breast cancer. BMC Cancer 2022; 22:533. [PMID: 35549691 PMCID: PMC9097419 DOI: 10.1186/s12885-022-09603-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 04/24/2022] [Indexed: 11/25/2022] Open
Abstract
Background As a metastasis-related protein, NEDD9 has been reported in breast cancer (BC) metastasis research. However, there are few studies on the upstream regulators of NEDD9, especially involving the potential role of miRNAs. The purpose of this study was to explain whether miR-107 potentially regulates NEDD9, which may lead to invasion and metastasis of BC. Methods MCF-7 and MDA-MB-231 cells were transduced with lentiviruses to construct stably transduced cells with miR-107 overexpression, miR-107 silencing or empty vectors. A luciferase reporter assay was performed to verify the binding of miR-107 and NEDD9. The scratch test and Transwell assay were used to measure cell migration and invasion ability, respectively. For the study of metastasis in vivo, we injected MDA-MB-231 cells into the fat pad of nude mice to develop an orthotopic breast cancer model. Results We found that NEDD9 expression correlates with the prognosis of BC patients. In BC cell lines, NEDD9 was positively correlated with cell migration ability. Further research revealed that miR-107 inhibited NEDD9 expression by targeting the 3′-untranslated region of NEDD9. Overexpression of miR-107 suppressed the expression of NEDD9, thereby inhibiting the invasion, migration and proliferation of BC cells, but interference with miR-107 promoted the expression of NEDD9 as well as invasion, migration and proliferation. In an in vivo model, overexpression of miR-107 decreased the expression of NEDD9 and inhibited tumour growth, invasion and metastasis; however, these effects were reversed by inhibiting miR-107. Conclusions These findings indicated the potential role of miR-107 in regulating NEDD9 in the invasion, migration and proliferation of BC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09603-3.
Collapse
Affiliation(s)
- Jiamin Zhou
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China
| | - Xianglin Sun
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China
| | - Xinyu Zhang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China
| | - Huan Yang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China.
| | - Zhenglin Jiang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China
| | - Qianqian Luo
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China
| | - Yifei Liu
- Department of Pathology, Affiliated Hospital of Nantong University, 20 Xisi Road, Nantong, 226001, China
| | - Guohua Wang
- Department of Physiology and Hypoxic Biomedicine, Institute of Special Environmental Medicine, Nantong University, 9 Seyuan Road, Chongchuan District, Nantong, 226019, Jiangsu, China.
| |
Collapse
|
40
|
Lu M, Huang X, Cai X, Sun J, Liu X, Weng L, Zhu L, Luo Q, Chen Z. Hypoxia-Responsive Stereocomplex Polymeric Micelles with Improved Drug Loading Inhibit Breast Cancer Metastasis in an Orthotopic Murine Model. ACS APPLIED MATERIALS & INTERFACES 2022; 14:20551-20565. [PMID: 35476401 DOI: 10.1021/acsami.1c23737] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Tumor metastasis is a leading cause of breast cancer-related death. Taxane-loaded polymeric formulations, such as Genexol PM and Nanoxel M using poly(ethylene glycol)-poly(d,l-lactide) (PEG-PLA) micelles as drug carriers, have been approved for the treatment of metastatic breast cancer. Unfortunately, the physical instability of PEG-PLA micelles, leading to poor drug loading, premature drug leakage, and consequently limited drug delivery to tumors, largely hinders their therapeutic outcome. Inspired by the enantiomeric nature of PLA, this work developed stereocomplex PEG-PLA micelles through stereoselective interactions of enantiomeric PLA, which are further incorporated with a hypoxia-responsive moiety used as a hypoxia-cleavable linker of PEG and PLA, to maximize therapeutic outcomes. The results showed that the obtained micelles had high structural stability, showing improved drug loading for effective drug delivery to tumors as well as other tissues. Especially, they were capable of sensitively responding to the hypoxic tumor environment for drug release, reversing hypoxia-induced drug resistance and hypoxia-promoted cell migration for enhanced bioavailability under hypoxia. In vivo results further showed that the micelles, especially at a high dose, inhibited the growth of the primary tumor and improved tumor pathological conditions, consequently remarkably inhibiting its metastasis to the lungs and liver, while not causing any systemic toxicity. Hypoxia-responsive stereocomplex micelles thus emerge as a reliable drug delivery system to treat breast cancer metastasis.
Collapse
Affiliation(s)
- Min Lu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Xu Huang
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Xiaohui Cai
- Department of Hematology, Nanjing Medical University, Affiliated Changzhou No. 2 People's Hospital, Changzhou 213000, People's Republic of China
| | - Jiajia Sun
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Xuemeng Liu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Lingyan Weng
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Li Zhu
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Qianqian Luo
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| | - Zhongping Chen
- Institute of Special Environmental Medicine and Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226019, People's Republic of China
| |
Collapse
|
41
|
Zhu J, Guo T, Wang Z, Zhao Y. Triggered azobenzene-based prodrugs and drug delivery systems. J Control Release 2022; 345:475-493. [PMID: 35339578 DOI: 10.1016/j.jconrel.2022.03.041] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/18/2022] [Accepted: 03/20/2022] [Indexed: 12/18/2022]
Abstract
Azobenzene-based molecules show unique trans-cis isomerization upon ultraviolet light irradiation, which induce the change of polarity, crystallinity, stability, and binding affinity with pharmacological target. Moreover, azobenzene is the substrate of azoreductase that is often overexpressed in many pathological sites, e.g. hypoxic solid tumor. Therefore, azobenzene can be a multifunctional molecule in material science, pharmaceutical science and biomedicine because of its sensitivity to light, hypoxia and certain enzymes, hence showing potential application in site-specific smart therapy. Herein we focus on the employment of azobenzene and its derivatives for engineering triggered prodrug and drug delivery systems, and provide an overview of photoswitchable azo-based prodrugs, the associated problems regarding ultraviolet light and reversible isomerization, as well as the potential solutions. We also present the advance of azo-bearing delivery vehicles wherein azobenzene act as the linker, capping agent, and building block, and discuss the corresponding mechanisms for controlled cargo release, endocytosis enhancement and sensitization of free radical cancer therapy.
Collapse
Affiliation(s)
- Jundong Zhu
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China
| | - Tao Guo
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300120, China
| | - Zheng Wang
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China.
| | - Yanjun Zhao
- School of Pharmaceutical Science & Technology, Tianjin Key Laboratory for Modern Drug Delivery & High Efficiency, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin University, Tianjin 300072, China.
| |
Collapse
|
42
|
Zhang X, Liu Q, Zhang T, Gao P, Wang H, Yao L, Huang J, Jiang S. Bone-targeted nanoplatform enables efficient modulation of bone tumor microenvironment for prostate cancer bone metastasis treatment. Drug Deliv 2022; 29:889-905. [PMID: 35285760 PMCID: PMC8928789 DOI: 10.1080/10717544.2022.2050845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
As there is currently no effective therapy for patients with prostate cancer (PCa) bone metastasis, it was stringent to explore the relevant treatment strategies. Actually, the interaction between cancer cells and bone microenvironment plays important role in prostate cancer bone metastasis, especially the Sonic hedgehog protein (SHH) signaling in the bone microenvironment. The SHH promotes osteoblast maturation and osteoblast then secretes RANKL to induce osteoclastogenesis. Herein, this study develops bone-targeting calcium phosphate lipid hybrid nanoparticles (NPs) loaded with docetaxel (DTXL) and SHH siRNA for PCa bone metastasis treatment. For bone targeting purposes, the nanoplatform was modified with alendronate (ALN). (DTXL + siRNA)@NPs-ALN NPs effectively change the bone microenvironment by inhibiting the SHH paracrine and autocrine signaling, enhancing the anti-tumor effects of DTXL. Besides showing good in vitro cellular uptake, the NPs-ALN also inhibited tumor growth both in vitro and in vivo by inducing apoptosis, cell cycle arrest, and autophagy. This DDS comprised of (DTXL + siRNA)-loaded NPs provides an excellent strategy to treat PCa bone metastasis.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Postdoctoral of Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Pathology, Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Qingbin Liu
- Clinical Medical Laboratory Center, Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Tingting Zhang
- Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Pei Gao
- Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Hui Wang
- Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Lu Yao
- Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| | - Jingwen Huang
- The First Affiliated Hospital of Bengbu Medical College, Tumor Hospital Affiliated to Bengbu Medical College, Bengbu, China
| | - Shulong Jiang
- Clinical Medical Laboratory Center, Jining No. 1 People's Hospital, Jining Medical University, Jining, China
| |
Collapse
|
43
|
Niu Y, Yang H, Yu Z, Gao C, Ji S, Yan J, Han L, Huo Q, Xu M, Liu Y. Intervention with the Bone-Associated Tumor Vicious Cycle through Dual-Protein Therapeutics for Treatment of Skeletal-Related Events and Bone Metastases. ACS NANO 2022; 16:2209-2223. [PMID: 35077154 DOI: 10.1021/acsnano.1c08269] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Bone metastasis is a common metastasis site such as lung cancer, prostate cancer, and other malignant tumors. The occurrence of bone metastases of lung cancer is often accompanied by bone loss, fracture, and other skeletal-related events (SREs) caused by tumor proliferation and osteoclast activation. Furthermore, along with the differentiation and maturation of osteoclasts in the bone microenvironment, it will further promote the occurrence and development of bone metastasis. Protein drugs are one of the most promising therapeutic pharmaceuticals, but in vivo delivery of protein therapeutics still confronts great challenges. In order to more effectively conquer bone metastases and alleviate SREs, herein, we constructed biomineralized metal-organic framework (MOF) nanoparticles carrying protein toxins with both bone-seeking and CD44-receptor-targeting abilities. More importantly, through combination with Receptor Activator of Nuclear Factor-κ B Ligand (RANKL) antibody, in vivo results demonstrated that these two protein agents not only enhanced the detraction effects of protein toxin agents as ribosome-inactivating protein (RIP) on bone metastatic tumor cells but also exhibited synergistic intervention of the crosstalk between bone cells and tumor cells and reduced SREs such as bone loss. Collectively, we expect that this strategy can provide an effective and safe option in regulating bone-tumor microenvironments to overcome bone metastasis and SREs.
Collapse
Affiliation(s)
- Yimin Niu
- Department of Pharmacy, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
- Department of Neurology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, Jiangsu, China
| | - Hongbin Yang
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, Anhui, China
| | - Zhenyan Yu
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, Anhui, China
| | - Cuicui Gao
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu, China
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, Liaoning, China
| | - Shuaishuai Ji
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, Anhui, China
| | - Jie Yan
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, Anhui, China
| | - Lei Han
- Department of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, Jiangsu, China
| | - Qiang Huo
- School of Pharmacy, Bengbu Medical College, Bengbu 233030, Anhui, China
| | - Ming Xu
- Department of Occupational Disease Prevention, Jiangsu Provincial Center for Disease Control and Prevention, Nanjing 210009, Jiangsu, China
| | - Yang Liu
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| |
Collapse
|
44
|
Shariatinia Z. Big family of nano- and microscale drug delivery systems ranging from inorganic materials to polymeric and stimuli-responsive carriers as well as drug-conjugates. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102790] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|