1
|
Chen L, He Y, Lan J, Li Z, Gu D, Nie W, Zhang T, Ding Y. Advancements in nano drug delivery system for liver cancer therapy based on mitochondria-targeting. Biomed Pharmacother 2024; 180:117520. [PMID: 39395257 DOI: 10.1016/j.biopha.2024.117520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/26/2024] [Accepted: 10/04/2024] [Indexed: 10/14/2024] Open
Abstract
Based on poor efficacy and non-specific toxic side effects of conventional drug therapy for liver cancer, nano-based drug delivery system (NDDS) offers the advantage of drug targeting delivery. Subcellular targeting of nanomedicines on this basis enables more precise and effective termination of tumor cells. Mitochondria, as the crucial cell powerhouse, possesses distinctive physical and chemical properties in hepatoma cells different from that in hepatic cells, and controls apoptosis, tumor metastasis, and cellular drug resistance in hepatoma cells through metabolism and dynamics, which serves as a good choice for drug targeting delivery. Thus, mitochondria-targeting NDDS have become a recent research focus, showcasing the design of cationic nanoparticles, metal nanoparticles, mitochondrial peptide modification and so on. Although many studies have shown good results regarding anti-tumor efficacy, it is a long way to go before the successful translation of clinical application. Based on these, we summarized the specificity and importance of mitochondria in hepatoma cells, and reviewed the current mitochondria-targeting NDDS for liver cancer therapy, aiming to provide a better understanding for current development process, strengths and weaknesses of mitochondria-targeting NDDS as well as informing subsequent improvements and developments.
Collapse
Affiliation(s)
- Lixia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yitian He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Donghao Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Wenlong Nie
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
2
|
Xiao A, Yin L, Chen T, Qian H. Lipo/TK-CDN/TPP/Y6 nanoparticles inhibit cutaneous melanoma formation. J Drug Target 2024; 32:931-940. [PMID: 38838039 DOI: 10.1080/1061186x.2024.2365243] [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: 01/21/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Stimulation of the innate immune stimulator of interferon genes (STING) pathway has been shown to boost anti-tumour immunity. Nevertheless, the systemic delivery of STING agonists to the tumour presents challenges. Therefore, we designed a cyclic dinucleotide (CDN)-based drug delivery system (DDS) combined photothermal therapy (PTT)/photodynamic therapy (PDT)/immunotherapy for cutaneous melanoma. We coencapsulated a reactive oxygen species (ROS)-responsive prodrug thioketone-linked CDN (TK-CDN), and photoresponsive agents chlorin E6 (Y6) within mitochondria-targeting reagent triphenylphosphonium (TPP)-modified liposomes (Lipo/TK-CDN/TPP/Y6). Lipo/TK-CDN/TPP/Y6 exhibited a photothermal effect similar to Y6, along with a superior cellular uptake rate. Upon endocytosis by B16F10 cells, Lipo/TK-CDN/TPP/Y6 generated large amounts of ROS under laser irradiation for PDT. Mice bearing B16F10 tumours were intravenously injected with Lipo/TK-CDN/TPP/Y6 and exposed to irradiation, resulting in a substantial inhibition of tumour growth. Exploration of the mechanism of anti-tumour action showed that Lipo/TK-CDN/TPP/Y6 had a stronger stimulation of STING activation and anti-tumour immune cell infiltration compared to other groups. Hence, the Lipo/TK-CDN/TPP/Y6 nanoparticles offer great potential as a DDS for targeted and on-demand drug release at tumour sites. These nanoparticles exhibit promise as a candidate for precise and controllable combination therapy in the treatment of tumours.
Collapse
Affiliation(s)
- Anju Xiao
- Department of Dermatology and Venereology, Dejiang County People's Hospital, Affiliated to Zunyi Medical University, Dejiang, China
| | - Li Yin
- Department of Pathology, Dejiang County People's Hospital, Affiliated to Zunyi Medical University, Dejiang, China
| | - Ting Chen
- Department of Clinical Medicine, Dejiang County People's Hospital, Affiliated to Zunyi Medical University, Dejiang, China
| | - Huiling Qian
- Department of Endocrinology, Dejiang County People's Hospital, Affiliated to Zunyi Medical University, Dejiang, China
| |
Collapse
|
3
|
Jiang F, Liu S, Wang L, Chen H, Huang Y, Cao Y, Wang X, Lin M, Zhang J. ROS-Responsive Nanoprobes for Bimodal Imaging-Guided Cancer Targeted Combinatorial Therapy. Int J Nanomedicine 2024; 19:8071-8090. [PMID: 39130685 PMCID: PMC11317049 DOI: 10.2147/ijn.s467512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 07/17/2024] [Indexed: 08/13/2024] Open
Abstract
Purpose Chemotherapy mediated by Reactive oxygen species (ROS)-responsive drug delivery systems can potentially mitigate the toxic side effects of chemotherapeutic drugs and significantly enhance their therapeutic efficacy. However, achieving precise targeted drug delivery and real-time control of ROS-responsive drug release at tumor sites remains a formidable challenge. Therefore, this study aimed to describe a ROS-responsive drug delivery system with specific tumor targeting capabilities for mitigating chemotherapy-induced toxicity while enhancing therapeutic efficacy under guidance of Fluorescence (FL) and Magnetic resonance (MR) bimodal imaging. Methods Indocyanine green (ICG), Doxorubicin (DOX) prodrug pB-DOX and Superparamagnetic iron oxide (SPIO, Fe3O4) were encapsulated in poly(lactic-co-glycolic acid) (PLGA) by double emulsification method to prepare ICG/ pB-DOX/ Fe3O4/ PLGA nanoparticles (IBFP NPs). The surface of IBFP NPs was functionalized with mammaglobin antibodies (mAbs) by carbodiimide method to construct the breast cancer-targeting mAbs/ IBFP NPs (MIBFP NPs). Thereafter, FL and MR bimodal imaging ability of MIBFP NPs was evaluated in vitro and in vivo. Finally, the combined photodynamic therapy (PDT) and chemotherapy efficacy evaluation based on MIBFP NPs was studied. Results The multifunctional MIBFP NPs exhibited significant targeting efficacy for breast cancer. FL and MR bimodal imaging clearly displayed the distribution of the targeting MIBFP NPs in vivo. Upon near-infrared laser irradiation, the MIBFP NPs loaded with ICG effectively generated ROS for PDT, enabling precise tumor ablation. Simultaneously, it triggered activation of the pB-DOX by cleaving its sensitive moiety, thereby restoring DOX activity and achieving ROS-responsive targeted chemotherapy. Furthermore, the MIBFP NPs combined PDT and chemotherapy to enhance the efficiency of tumor ablation under guidance of bimodal imaging. Conclusion MIBFP NPs constitute a novel dual-modality imaging-guided drug delivery system for targeted breast cancer therapy and offer precise and controlled combined treatment options.
Collapse
Affiliation(s)
- Fujie Jiang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Shuling Liu
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Lu Wang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Huifang Chen
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Yao Huang
- School of Medicine, Chongqing University, Chongqing, 400030, People’s Republic of China
| | - Ying Cao
- School of Medicine, Chongqing University, Chongqing, 400030, People’s Republic of China
| | - Xiaoxia Wang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Meng Lin
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing, 400030, People’s Republic of China
| |
Collapse
|
4
|
Cho H, Huh KM, Shim MS, Cho YY, Lee JY, Lee HS, Kang HC. Beyond Nanoparticle-Based Intracellular Drug Delivery: Cytosol/Organelle-Targeted Drug Release and Therapeutic Synergism. Macromol Biosci 2024; 24:e2300590. [PMID: 38488862 DOI: 10.1002/mabi.202300590] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/07/2024] [Indexed: 07/16/2024]
Abstract
Nanoparticle (NP)-based drug delivery systems are conceived to solve poor water-solubility and chemical/physical instability, and their purpose expanded to target specific sites for maximizing therapeutic effects and minimizing unwanted events of payloads. Targeted sites are also narrowed from organs/tissues and cells to cytosol/organelles. Beyond specific site targeting, the particular release of payloads at the target sites is growing in importance. This review overviews various issues and their general strategies during multiple steps, from the preparation of drug-loaded NPs to their drug release at the target cytosol/organelles. In particular, this review focuses on current strategies for "first" delivery and "later" release of drugs to the cytosol or organelles of interest using specific stimuli in the target sites. Recognizing or distinguishing the presence/absence of stimuli or their differences in concentration/level/activity in one place from those in another is applied to stimuli-triggered release via bond cleavage or nanostructural transition. In addition, future directions on understanding the intracellular balance of stimuli and their counter-stimuli are demonstrated to synergize the therapeutic effects of payloads released from stimuli-sensitive NPs.
Collapse
Affiliation(s)
- Hana Cho
- Department of Pharmacy, College of Pharmacy and Regulated Cell Death (RCD) Control·Material Research Institute, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Kang Moo Huh
- Department of Polymer Science and Engineering, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Yong-Yeon Cho
- Department of Pharmacy, College of Pharmacy and Regulated Cell Death (RCD) Control·Material Research Institute, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Joo Young Lee
- Department of Pharmacy, College of Pharmacy and Regulated Cell Death (RCD) Control·Material Research Institute, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Hye Suk Lee
- Department of Pharmacy, College of Pharmacy and Regulated Cell Death (RCD) Control·Material Research Institute, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy and Regulated Cell Death (RCD) Control·Material Research Institute, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, Republic of Korea
| |
Collapse
|
5
|
Gautam S, Singh N, Marwaha D, Rai N, Sharma M, Tiwari P, Singh S, Kumar Bakshi A, Kumar A, Agarwal N, Prakash Shukla R, Ranjan Mishra P. Celastrol-loaded polymeric mixed micelles shows improved antitumor efficacy in 4 T1 bearing xenograft mouse model through spatial targeting. Int J Pharm 2024; 659:124234. [PMID: 38763310 DOI: 10.1016/j.ijpharm.2024.124234] [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: 12/09/2023] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
In this study, we have proposed a novel approach that combines hyaluronic acid (HA), folic acid (FA), and celastrol (CLS) within a polymeric micelle system (CLS-HF/MLs), offering a dual-action strategy against breast cancer. Polymeric mixed micelles were prepared through the thin-film hydration method, and comprehensive quality control parameters were established, encompassing particle size, polydispersity index, zeta potential, surface morphology, encapsulation efficiency, drug content, in vitro drug release, and storage stability assessment. The average particle size of CLS-HF/MLs micelles was found to be 120 nm and their drug loading and encapsulation efficiencies were 15.9 % and 89.52 %, respectively. The in vitro release data showed that the CLS-HF/MLs targeted mixed micelles displayed a prolonged release profile compared to the free drug. Additionally, the stability of the developed polymeric mixed micelles was maintained for up to 8 weeks of storage in terms of particle size and drug content. Furthermore, both flow cytometry and confocal laser scanning microscopy studies indicated a significant enhancement in the cellular uptake efficiency and cytotoxicity of CLS-HF/MLs mixed micelles against MCF-7 cell line. In terms of pharmacokinetic analysis, the half-life and AUC values of CLS-HF/MLs mixed micelles were found to be approximately 4.71- and 7.36-folds higher than the values of free drug (CLS), respectively. The CLS-HF/MLs micelles exhibited remarkable antitumor efficacy (almost complete ablation of the 4 T1-cell bearing tumor xenografts mouse model) due to the dual receptor (CD44 and folate) targeting effects with minimal side effects. When considering the cumulative findings of our present research, it becomes evident that mixed micelles designed for chemotherapy offer a promising and potentially effective therapeutic avenue for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Sanjay Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Ankit Kumar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Neha Agarwal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
6
|
Lin P, Lu Y, Zheng J, Lin Y, Zhao X, Cui L. Strategic disruption of cancer's powerhouse: precise nanomedicine targeting of mitochondrial metabolism. J Nanobiotechnology 2024; 22:318. [PMID: 38849914 PMCID: PMC11162068 DOI: 10.1186/s12951-024-02585-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/26/2024] [Indexed: 06/09/2024] Open
Abstract
Mitochondria occupy a central role in the biology of most eukaryotic cells, functioning as the hub of oxidative metabolism where sugars, fats, and amino acids are ultimately oxidized to release energy. This crucial function fuels a variety of cellular activities. Disruption in mitochondrial metabolism is a common feature in many diseases, including cancer, neurodegenerative conditions and cardiovascular diseases. Targeting tumor cell mitochondrial metabolism with multifunctional nanosystems emerges as a promising strategy for enhancing therapeutic efficacy against cancer. This review comprehensively outlines the pathways of mitochondrial metabolism, emphasizing their critical roles in cellular energy production and metabolic regulation. The associations between aberrant mitochondrial metabolism and the initiation and progression of cancer are highlighted, illustrating how these metabolic disruptions contribute to oncogenesis and tumor sustainability. More importantly, innovative strategies employing nanomedicines to precisely target mitochondrial metabolic pathways in cancer therapy are fully explored. Furthermore, key challenges and future directions in this field are identified and discussed. Collectively, this review provides a comprehensive understanding of the current state and future potential of nanomedicine in targeting mitochondrial metabolism, offering insights for developing more effective cancer therapies.
Collapse
Affiliation(s)
- Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
| | - Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, 510280, Guangdong, China.
- School of Dentistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
7
|
Gu M, Li C, Deng Q, Chen X, Lei R. Celastrol enhances the viability of random-pattern skin flaps by regulating autophagy through the AMPK-mTOR-TFEB axis. Phytother Res 2024; 38:3020-3036. [PMID: 38600729 DOI: 10.1002/ptr.8198] [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: 08/24/2023] [Revised: 01/06/2024] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
In reconstructive and plastic surgery, random-pattern skin flaps (RPSF) are often used to correct defects. However, their clinical usefulness is limited due to their susceptibility to necrosis, especially on the distal side of the RPSF. This study validates the protective effect of celastrol (CEL) on flap viability and explores in terms of underlying mechanisms of action. The viability of different groups of RPSF was evaluated by survival zone analysis, laser doppler blood flow, and histological analysis. The effects of CEL on flap angiogenesis, apoptosis, oxidative stress, and autophagy were evaluated by Western blot, immunohistochemistry, and immunofluorescence assays. Finally, its mechanistic aspects were explored by autophagy inhibitor and Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) inhibitor. On the seventh day after surgery, the survival area size, blood supply, and microvessel count of RPSF were augmented following the administration of CEL. Additionally, CEL stimulated angiogenesis, suppressed apoptosis, and lowered oxidative stress levels immediately after elevated autophagy in ischemic regions; These effects can be reversed using the autophagy inhibitor chloroquine (CQ). Specifically, CQ has been observed to counteract the protective impact of CEL on the RPSF. Moreover, it has also been discovered that CEL triggers the AMPK-mTOR-TFEB axis activation in the area affected by ischemia. In CEL-treated skin flaps, AMPK inhibitors were demonstrated to suppress the AMPK-mTOR-TFEB axis and reduce autophagy levels. This investigation suggests that CEL benefits the survival of RPSF by augmenting angiogenesis and impeding oxidative stress and apoptosis. The results are credited to increased autophagy, made possible by the AMPK-mTOR-TFEB axis activation.
Collapse
Affiliation(s)
- Mingbao Gu
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Chenchao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Qingyu Deng
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ximiao Chen
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
- Zhejiang Provincial Key Laboratory of Orthopaedics, Wenzhou, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Rui Lei
- Department of Plastic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Klemt I, Reshetnikov V, Dutta S, Bila G, Bilyy R, Cuartero IC, Hidalgo A, Wünsche A, Böhm M, Wondrak M, Kunz-Schughart LA, Tietze R, Beierlein F, Imhof P, Gensberger-Reigl S, Pischetsrieder M, Körber M, Jost T, Mokhir A. A concept of dual-responsive prodrugs based on oligomerization-controlled reactivity of ester groups: an improvement of cancer cells versus neutrophils selectivity of camptothecin. RSC Med Chem 2024; 15:1189-1197. [PMID: 38665843 PMCID: PMC11042170 DOI: 10.1039/d3md00609c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/20/2024] [Indexed: 04/28/2024] Open
Abstract
Many known chemotherapeutic anticancer agents exhibit neutropenia as a dose-limiting side effect. In this paper we suggest a prodrug concept solving this problem for camptothecin (HO-cpt). The prodrug is programmed according to Boolean "AND" logic. In the absence of H2O2 (trigger T1), e.g. in the majority of normal cells, it exists as an inactive oligomer. In cancer cells and in primed neutrophils (high H2O2), the oligomer is disrupted forming intermediate (inactive) lipophilic cationic species. These are accumulated in mitochondria (Mit) of cancer cells, where they are activated by hydrolysis at mitochondrial pH 8 (trigger T2) with formation of camptothecin. In contrast, the intermediates remain stable in neutrophils lacking Mit and therefore a source of T2. In this paper we demonstrated a proof-of-concept. Our prodrug exhibits antitumor activity both in vitro and in vivo, but is not toxic to normal cell and neutrophils in contrast to known single trigger prodrugs and the parent drug HO-cpt.
Collapse
Affiliation(s)
- Insa Klemt
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU) 91058 Erlangen Germany
| | - Viktor Reshetnikov
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU) 91058 Erlangen Germany
| | - Subrata Dutta
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU) 91058 Erlangen Germany
| | - Galyna Bila
- Department of Histology, Cytology and Embryology, Danylo Halytsky Lviv National Medical University 79010 Lviv Ukraine
| | - Rostyslav Bilyy
- Department of Histology, Cytology and Embryology, Danylo Halytsky Lviv National Medical University 79010 Lviv Ukraine
| | - Itziar Cossío Cuartero
- Program of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) C. Melchor Fernández Almagro, 3 28029 Madrid Spain
| | - Andrés Hidalgo
- Program of Cardiovascular Regeneration, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC) C. Melchor Fernández Almagro, 3 28029 Madrid Spain
| | - Adrian Wünsche
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU) 91058 Erlangen Germany
| | - Maximilian Böhm
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU) 91058 Erlangen Germany
| | - Marit Wondrak
- OncoRay, National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden and Helmholtz-Zentrum Dresden-Rossendorf Dresden Germany
| | - Leoni A Kunz-Schughart
- OncoRay, National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden and Helmholtz-Zentrum Dresden-Rossendorf Dresden Germany
- National Center for Tumor Diseases (NCT) Partner Site Dresden Germany
| | - Rainer Tietze
- Department of Otorhinolaryngology, Head and Neck Surgery, Section of Experimental Oncology and Nanomedicine (SEON), FAU University Hospital 91054 Erlangen Germany
| | - Frank Beierlein
- Erlangen National High Performance Computing Center (NHR@FAU), FAU 91058 Erlangen Germany
- Computer-Chemistry-Center, Department of Chemistry and Pharmacy, FAU Germany
| | - Petra Imhof
- Erlangen National High Performance Computing Center (NHR@FAU), FAU 91058 Erlangen Germany
- Computer-Chemistry-Center, Department of Chemistry and Pharmacy, FAU Germany
| | | | | | - Marlies Körber
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU) 91058 Erlangen Germany
| | - Tina Jost
- Department of Radiation Oncology, FAU University Hospital 91054 Erlangen Germany
| | - Andriy Mokhir
- Department of Chemistry and Pharmacy, Organic Chemistry II, Friedrich-Alexander-University of Erlangen-Nürnberg (FAU) 91058 Erlangen Germany
| |
Collapse
|
9
|
Pegoraro C, Domingo-Ortí I, Conejos-Sánchez I, Vicent MJ. Unlocking the Mitochondria for Nanomedicine-based Treatments: Overcoming Biological Barriers, Improving Designs, and Selecting Verification Techniques. Adv Drug Deliv Rev 2024; 207:115195. [PMID: 38325562 DOI: 10.1016/j.addr.2024.115195] [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: 10/24/2023] [Revised: 01/13/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Enhanced targeting approaches will support the treatment of diseases associated with dysfunctional mitochondria, which play critical roles in energy generation and cell survival. Obstacles to mitochondria-specific targeting include the presence of distinct biological barriers and the need to pass through (or avoid) various cell internalization mechanisms. A range of studies have reported the design of mitochondrially-targeted nanomedicines that navigate the complex routes required to influence mitochondrial function; nonetheless, a significant journey lies ahead before mitochondrially-targeted nanomedicines become suitable for clinical use. Moving swiftly forward will require safety studies, in vivo assays confirming effectiveness, and methodologies to validate mitochondria-targeted nanomedicines' subcellular location/activity. From a nanomedicine standpoint, we describe the biological routes involved (from administration to arrival within the mitochondria), the features influencing rational design, and the techniques used to identify/validate successful targeting. Overall, rationally-designed mitochondria-targeted-based nanomedicines hold great promise for precise subcellular therapeutic delivery.
Collapse
Affiliation(s)
- Camilla Pegoraro
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inés Domingo-Ortí
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - Inmaculada Conejos-Sánchez
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| | - María J Vicent
- Polymer Therapeutics Laboratory and CIBERONC, Príncipe Felipe Research Center, Av. Eduardo Primo Yúfera 3, E-46012 Valencia, Spain.
| |
Collapse
|
10
|
Liu J, Cabral H, Mi P. Nanocarriers address intracellular barriers for efficient drug delivery, overcoming drug resistance, subcellular targeting and controlled release. Adv Drug Deliv Rev 2024; 207:115239. [PMID: 38437916 DOI: 10.1016/j.addr.2024.115239] [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: 11/22/2023] [Revised: 01/16/2024] [Accepted: 02/27/2024] [Indexed: 03/06/2024]
Abstract
The cellular barriers are major bottlenecks for bioactive compounds entering into cells to accomplish their biological functions, which limits their biomedical applications. Nanocarriers have demonstrated high potential and benefits for encapsulating bioactive compounds and efficiently delivering them into target cells by overcoming a cascade of intracellular barriers to achieve desirable therapeutic and diagnostic effects. In this review, we introduce the cellular barriers ahead of drug delivery and nanocarriers, as well as summarize recent advances and strategies of nanocarriers for increasing internalization with cells, promoting intracellular trafficking, overcoming drug resistance, targeting subcellular locations and controlled drug release. Lastly, the future perspectives of nanocarriers for intracellular drug delivery are discussed, which mainly focus on potential challenges and future directions. Our review presents an overview of intracellular drug delivery by nanocarriers, which may encourage the future development of nanocarriers for efficient and precision drug delivery into a wide range of cells and subcellular targets.
Collapse
Affiliation(s)
- Jing Liu
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan.
| | - Peng Mi
- Department of Radiology, Huaxi MR Research Center (HMRRC), State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, No.17 South Renmin Road, Chengdu, Sichuan 610041, China.
| |
Collapse
|
11
|
Bai S, Zhang XD, Zou YQ, Lin YX, Liu ZY, Li KW, Huang P, Yoshida T, Liu YL, Li MS, Zhang W, Wang XJ, Zhang M, Du C. Development of high-efficiency superparamagnetic drug delivery system with MPI imaging capability. Front Bioeng Biotechnol 2024; 12:1382085. [PMID: 38572358 PMCID: PMC10987818 DOI: 10.3389/fbioe.2024.1382085] [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: 02/05/2024] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
In this study, a high-efficiency superparamagnetic drug delivery system was developed for preclinical treatment of bladder cancer in small animals. Two types of nanoparticles with magnetic particle imaging (MPI) capability, i.e., single- and multi-core superparamagnetic iron oxide nanoparticles (SPIONs), were selected and coupled with bladder anti-tumor drugs by a covalent coupling scheme. Owing to the minimal particle size, magnetic field strengths of 270 mT with a gradient of 3.2 T/m and 260 mT with a gradient of 3.7 T/m were found to be necessary to reach an average velocity of 2 mm/s for single- and multi-core SPIONs, respectively. To achieve this, a method of constructing an in vitro magnetic field for drug delivery was developed based on hollow multi-coils arranged coaxially in close rows, and magnetic field simulation was used to study the laws of the influence of the coil structure and parameters on the magnetic field. Using this method, a magnetic drug delivery system of single-core SPIONs was developed for rabbit bladder therapy. The delivery system consisted of three coaxially and equidistantly arranged coils with an inner diameter of Φ50 mm, radial height of 85 mm, and width of 15 mm that were positioned in close proximity to each other. CCK8 experimental results showed that the three types of drug-coupled SPION killed tumor cells effectively. By adjusting the axial and radial positions of the rabbit bladder within the inner hole of the delivery coil structure, the magnetic drugs injected could undergo two-dimensional delivery motions and were delivered and aggregated to the specified target location within 12 s, with an aggregation range of about 5 mm × 5 mm. In addition, the SPION distribution before and after delivery was imaged using a home-made open-bore MPI system that could realistically reflect the physical state. This study contributes to the development of local, rapid, and precise drug delivery and the visualization of this process during cancer therapy, and further research on MPI/delivery synchronization technology is planned for the future.
Collapse
Affiliation(s)
- Shi Bai
- Department of Information Engineering, Shenyang University of Technology, Shenyang, China
| | - Xiao-dan Zhang
- Department of Information Engineering, Shenyang University of Technology, Shenyang, China
| | - Yu-qi Zou
- Department of Information Engineering, Shenyang University of Technology, Shenyang, China
| | - Yu-xi Lin
- Department of Information Engineering, Shenyang University of Technology, Shenyang, China
| | - Zhi-yao Liu
- Department of Information Engineering, Shenyang University of Technology, Shenyang, China
| | - Ke-wen Li
- Department of Information Engineering, Shenyang University of Technology, Shenyang, China
| | - Ping Huang
- Department of Information Engineering, Shenyang University of Technology, Shenyang, China
| | - Takashi Yoshida
- Department of Electrical Engineering, Kyushu University, Fukuoka, Japan
| | - Yi-li Liu
- Department of Urology, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Ming-shan Li
- Department of Urology, Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Zhang
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiao-ju Wang
- Department of Foreign Languages, Liaoning Vocational and Technical College of Economics, Shenyang, China
| | - Min Zhang
- First Affiliated Hospital, China Medical University, Shenyang, China
| | - Cheng Du
- Department of Oncology, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
12
|
Zhang F, Chen J, Luo W, Wen C, Mao W, Yang Y, Liu C, Xu Y, Chen W, Wen L. Mitochondria targeted biomimetic platform for chemo/photodynamic combination therapy against osteosarcoma. Int J Pharm 2024; 652:123865. [PMID: 38286195 DOI: 10.1016/j.ijpharm.2024.123865] [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: 11/07/2023] [Revised: 01/13/2024] [Accepted: 01/26/2024] [Indexed: 01/31/2024]
Abstract
Clinical treatment for osteosarcoma (OS) is still lacking effective means, and no significant progress in OS treatment have been made in recent years. Single chemotherapy has serious side effects and can produce drug resistance easily, resulting poor therapeutic effect. As a modern and non-invasive treatment form, photodynamic therapy (PDT) is widely used to treat diverse cancers. Chemotherapy in combination with PDT is a particularly efficient antitumor method that could overcome the defects of monotherapies. Since mitochondria is a key subcellular organelle involved in cell apoptosis regulation, targeting tumor cells mitochondria for drug delivery has become an important entry point for anti-tumor therapy. Herein, we rationally designed a core-shell structured biomimetic nanoplatform, i.e., D@SLNP@OSM-IR780, to achieve tumor homologous targeting and mitochondria targeted drug release for chemotherapy combined with PDT against OS. Upon 808 nm laser irradiation, D@SLNP@OSM-IR780 exhibited excellent photo-cytotoxicity in vitro. The excellent targeting effect of D@SLNP@OSM-IR780 in tumor tissues produced a tumor inhibition rate of 98.9% in vivo. We further indicated that synergistic chemo-photodynamic effect induced by D@SLNP@OSM-IR780 could activate mitochondria-mediated apoptosis pathway, along with host immune response and potential photothermal effect. On the whole, D@SLNP@OSM-IR780 is revealed to be a promising platform for OS targeted combination therapeutics.
Collapse
Affiliation(s)
- Fengtian Zhang
- Department of Orthopaedics, Second Affiliated Hospital of Soochow University Osteoporosis Research Institute of Soochow University, 1055 Sanxiang Road, Suzhou 215000, People's Republic of China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China; Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Jinling East Avenue, Zhanggong District, Ganzhou 341000, People's Republic of China
| | - Jiaoting Chen
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Weihong Luo
- College of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Changlong Wen
- Department of Infectious Diseases, Ganzhou People's Hospital, 17 Hongqi Avenue, Zhanggong District, Ganzhou 341000, People's Republic of China
| | - Wei Mao
- College of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Yutian Yang
- College of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China
| | - Chunting Liu
- People's Hospital of Shicheng County, Xihua Middle Road, Qinjiang Town, Ganzhou 342700, People's Republic of China
| | - Youjia Xu
- Department of Orthopaedics, Second Affiliated Hospital of Soochow University Osteoporosis Research Institute of Soochow University, 1055 Sanxiang Road, Suzhou 215000, People's Republic of China.
| | - Weiliang Chen
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China; College of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China.
| | - Lijuan Wen
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases of Ministry of Education, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China; College of Pharmacy, Gannan Medical University, University Park in Rongjiang New District, Ganzhou 341000, People's Republic of China.
| |
Collapse
|
13
|
Xiao S, Huang S, Yang X, Lei Y, Chang M, Hu J, Meng Y, Zheng G, Chen X. The development and evaluation of hyaluronic acid coated mitochondrial targeting liposomes for celastrol delivery. Drug Deliv 2023; 30:2162156. [PMID: 36600637 PMCID: PMC9828745 DOI: 10.1080/10717544.2022.2162156] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In order to precisely deliver celastrol into mitochondria of tumor cells, improve antitumor efficacy of celastrol and overcome its troublesome problems in clinical application, a novel multistage-targeted celastrol delivery system (C-TL/HA) was developed via electrostatic binding of hyaluronic acid (HA) to celastrol-loaded cationic liposomes composed of natural soybean phosphatidylcholine and cholesterol modified with mitochondrial targeting molecular TPP. Study results in this article showed that C-TL/HA successfully transported celastrol into mitochondria, effectively activated apoptosis of mitochondrial pathway, exerted higher tumor inhibition efficiency and lower toxic side effects compared with free celastrol. More importantly, HA coating not only enabled this delivery system to have good stability and safety in vivo, but also increased drug uptake and facilitated tumor targeting through recognizing CD44 receptors rich on the surface of tumor cells. Conclusively, this HA-coated mitochondrial targeting liposomes may provide a prospect for the clinical application of celastrol in tumor therapy.
Collapse
Affiliation(s)
- Simeng Xiao
- Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan, China
| | - Siying Huang
- Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan, China
| | - Xiaojing Yang
- Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan, China
| | - Yujie Lei
- Pharmacy Department, Wuxue No.1 People’s Hospital, Wuxue, China
| | - Mingxiang Chang
- Laboratory of Cell and Molecular Biology, Hubei Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Junjie Hu
- Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan, China
| | - Yan Meng
- Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan, China
| | - Guohua Zheng
- Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan, China,CONTACT Xinyan Chen Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan430065, China; Guohua Zheng Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan430065, China
| | - Xinyan Chen
- Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan, China,CONTACT Xinyan Chen Pharmacy Faculty, Hubei University of Chinese Medicine, Wuhan430065, China; Guohua Zheng Key Laboratory of Chinese Medicine Resource and Compound Prescription, Ministry of Education, Hubei University of Chinese Medicine, Wuhan430065, China
| |
Collapse
|
14
|
Zhang S, Zeng Y, Wang K, Song G, Yu Y, Meng T, Yuan H, Hu F. Chitosan-based nano-micelles for potential anti-tumor immunotherapy: Synergistic effect of cGAS-STING signaling pathway activation and tumor antigen absorption. Carbohydr Polym 2023; 321:121346. [PMID: 37739513 DOI: 10.1016/j.carbpol.2023.121346] [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: 04/13/2023] [Revised: 08/09/2023] [Accepted: 08/28/2023] [Indexed: 09/24/2023]
Abstract
Cyclic GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) signaling pathway is an essential DNA-sensing pathway to regulate the innate and adaptive immune response, which plays an important role in tumor immunotherapy. Although the STING agonists can be used, they are limited by their inability to target immune cells and systemic immunotoxicity, calling for novel strategies to accurately and effectively activate the cGAS-STING signaling pathway. Herein, mannose-modified stearic acid-grafted chitosan (M-CS-SA) micelles with the ability to activate the cGAS-STING signaling pathway and absorb tumor antigens were constructed. The chitosan-based nano-micelles showed valid dendritic cell (DCs) targeting and could escape from lysosomes leading to the activation of the cGAS-STING signaling pathway and the maturation of DCs. In addition, a combinatorial therapy was presented based on the programmed administration of oxaliplatin and M-CS-SA. M-CS-SA adsorbed tumor antigens released by chemotherapy to construct an autologous tumor vaccine and built a comprehensive antitumor immune response. In vivo, the combinatorial therapy achieved a tumor inhibition rate of 76.31 % at the oxaliplatin dose of 5 mg/kg and M-CS-SA dose of 15 mg/kg, and increased the CD3+ CD8+ T cell infiltration. This work demonstrated that M-CS-SA and its co-treatment with oxaliplatin showed great potential in tumor immunotherapy.
Collapse
Affiliation(s)
- Shufen Zhang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Yingping Zeng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Kai Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Guangtao Song
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Yiru Yu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China; Jinhua Institute of Zhejiang University, Jinhua 321299, China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China; Jinhua Institute of Zhejiang University, Jinhua 321299, China.
| |
Collapse
|
15
|
Wang Z, Qin Y, Wang X, Zhang T, Hu Y, Wang D, Zhang L, Zhu Y. Glutathione Programmed Mitochondria Targeted Delivery of Lonidamine for Effective Against Triple Negative Breast Cancer. Int J Nanomedicine 2023; 18:4023-4042. [PMID: 37520302 PMCID: PMC10378575 DOI: 10.2147/ijn.s413217] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/11/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Mitochondria are a significant target of lonidamine (LND). However, its limited solubility and inability to specifically target mitochondria, LND can lead to hepatic toxicity and has shown only modest anticancer activity. The objective of this study is to establish a glutathione programmed mitochondria targeted delivery of LND for the effective treatment of triple negative breast cancer (TNBC). Methods In this study, LND was encapsulated in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) wrapped with mitochondria-targeting short-chain triphenylphosphonium-tocopherol polyethylene glycol succinate (TPP-TPGS, TPS) and tumor-targeting long-chain 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-S-S-polyethylene glycol-R6RGD (DSPE-S-S-PEG2000-R6RGD, DSSR), which were designated as LND-PLGA/TPS/DSSR NPs. The release behavior was evaluated, and cellular uptake, in vitro and in vivo antitumor activity of nanoparticles were investigated. The mechanism, including apoptosis of tumor cells and mitochondrial damage and respiratory rate detection, was also further investigated. Results LND-PLGA/TPS/DSSR NPs were successfully prepared, and characterization revealed that they are globular particles with smooth surfaces and an average diameter of about 250 nm. Long-chain DSSR in LND-PLGA/TPS/DSSR NPs prevented positively charged LND-PLGA/TPS NPs from being cleared by the reticuloendothelial system. Furthermore, LND release rate from NPs at pH 8.0 was significantly higher than that at pH 7.4 and 5.5, which demonstrated specific LND release in mitochondria and prevented LND leakage in cytoplasm and lysosome. NPs could locate in mitochondria, and the released LND triggered apoptosis of tumor cells by damaging mitochondria and releasing apoptosis-related proteins. In addition, in TNBC mice model, programmed mitochondria targeted NPs improved efficacy and reduced LND toxicity. Conclusion LND-PLGA/TPS/DSSR NPs may be a useful system and provide an effective approach for the treatment of TNBC.
Collapse
Affiliation(s)
- Zhongjie Wang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, People’s Republic of China
| | - Yanru Qin
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, People’s Republic of China
| | - Xueyuan Wang
- College of Life Science, Nanjing Normal University, Nanjing, 210023, People’s Republic of China
| | - Tianyu Zhang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, People’s Republic of China
| | - Yixue Hu
- College of Life Science, Nanjing Normal University, Nanjing, 210023, People’s Republic of China
| | - Dongna Wang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, People’s Republic of China
| | - Liefeng Zhang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, People’s Republic of China
| | - Yongqiang Zhu
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, People’s Republic of China
- College of Life Science, Nanjing Normal University, Nanjing, 210023, People’s Republic of China
| |
Collapse
|
16
|
Wang C, Dai S, Zhao X, Zhang Y, Gong L, Fu K, Ma C, Peng C, Li Y. Celastrol as an emerging anticancer agent: Current status, challenges and therapeutic strategies. Biomed Pharmacother 2023; 163:114882. [PMID: 37196541 DOI: 10.1016/j.biopha.2023.114882] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/19/2023] Open
Abstract
Celastrol is a pentacyclic triterpenoid extracted from the traditional Chinese medicine Tripterygium wilfordii Hook F., which has multiple pharmacological activities. In particular, modern pharmacological studies have demonstrated that celastrol exhibits significant broad-spectrum anticancer activities in the treatment of a variety of cancers, including lung cancer, liver cancer, colorectal cancer, hematological malignancies, gastric cancer, prostate cancer, renal carcinoma, breast cancer, bone tumor, brain tumor, cervical cancer, and ovarian cancer. Therefore, by searching the databases of PubMed, Web of Science, ScienceDirect and CNKI, this review comprehensively summarizes the molecular mechanisms of the anticancer effects of celastrol. According to the data, the anticancer effects of celastrol can be mediated by inhibiting tumor cell proliferation, migration and invasion, inducing cell apoptosis, suppressing autophagy, hindering angiogenesis and inhibiting tumor metastasis. More importantly, PI3K/Akt/mTOR, Bcl-2/Bax-caspase 9/3, EGFR, ROS/JNK, NF-κB, STAT3, JNK/Nrf2/HO-1, VEGF, AR/miR-101, HSF1-LKB1-AMPKα-YAP, Wnt/β-catenin and CIP2A/c-MYC signaling pathways are considered as important molecular targets for the anticancer effects of celastrol. Subsequently, studies of its toxicity and pharmacokinetic properties showed that celastrol has some adverse effects, low oral bioavailability and a narrow therapeutic window. In addition, the current challenges of celastrol and the corresponding therapeutic strategies are also discussed, thus providing a theoretical basis for the development and application of celastrol in the clinic.
Collapse
Affiliation(s)
- Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shu Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xingtao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yafang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lihong Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
17
|
Gu X, Shu T, Deng W, Shen C, Wu Y. An X-ray activatable gold nanorod encapsulated liposome delivery system for mitochondria-targeted photodynamic therapy (PDT). J Mater Chem B 2023; 11:4539-4547. [PMID: 37161717 DOI: 10.1039/d3tb00608e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
In this work, we developed a mitochondria-targeted nanomaterial for neoadjuvant X-ray-triggered photodynamic therapy of rectal cancer. Herein, we designed a biodegradable liposome incorporating a photosensitizer, verteporfin, to generate X-ray-induced reactive oxygen species, gold nanorods as radiation enhancers, and triphenylphosphonium as the mitochondrial targeting moiety. The average size of the nanocarrier was about 150 nm. Due to the synergetic effect between X-ray and a combination of verteporfin and gold nanorods, as well as precise site-targeted TPP-modified liposomal nanocarriers, our nanoconjugates generated sufficient cytotoxic singlet oxygen within the mitochondria under X-ray irradiation, triggering the loss of membrane potential and mitochondria-related apoptosis of cancer cells.
Collapse
Affiliation(s)
- Xuefan Gu
- College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, Shaanxi, 710065, P. R. China
- ARC Centre of Excellence for Nanoscale Biophotonics, Graduate School of Biomedical Engineering, University of New South, Wales Kensington, 2052 NSW, Australia
- Faculty of Science and Engineering, Macquarie University, Sydney, 2109 NSW, Australia
| | - Tiantian Shu
- College of Chemistry and Chemical Engineering, Xi'an Shiyou University, Xi'an, Shaanxi, 710065, P. R. China
| | - Wei Deng
- School of Biomedical Engineering, University of Technology Sydney, Sydney, Australia
| | - Chao Shen
- Faculty of Science and Engineering, Macquarie University, Sydney, 2109 NSW, Australia
| | - Youshen Wu
- Department of Chemistry, School of Science, Xi'an Jiaotong University, Xi'an, Shaanxi, 710049, P. R. China.
| |
Collapse
|
18
|
Liu Y, Li J. Self-assembling nanoarchitectonics of size-controllable celastrol nanoparticles for efficient cancer chemotherapy with reduced systemic toxicity. J Colloid Interface Sci 2023; 636:216-222. [PMID: 36634391 DOI: 10.1016/j.jcis.2022.12.162] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 12/28/2022] [Accepted: 12/29/2022] [Indexed: 01/02/2023]
Abstract
Celastrol, extracted from Tripterygium wilfordii Hook F, is one of the most promising natural extract for cancer treatment. Nevertheless, insufficient tumor retention and severe systemic toxicity still hinder its application. Herein, we report for the first time that Celastrol can directly self-assemble into size-controllable nanoparticles through the anti-solvent method by using different good solvent or by the variation of Celastrol concentrations. In vitro anti-cancer experiment revealed that the as-prepared nanoparticles can kill MCF-7 cells more effectively. Moreover, the nanoparticles can efficiently accumulate in tumors of the tumor bearing mice after tail vein injection. Under the administration of lethal dosage of Celastrol, the tumors are greatly suppressed and the mice maintain the activity. These results demonstrate that anti-solvent method may be a promising strategy to fabricate Celastrol nano-drugs with controllable size and less systemic toxicity for further clinical cancer treatment.
Collapse
Affiliation(s)
- Yilin Liu
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junbai Li
- Beijing National Laboratory for Molecular Sciences (BNLMS), CAS Key Lab of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
19
|
Yan Y, Li Y, Li H, Ma X, Tang Y, Yi K, Lin X, Li J, Liu Z. Antimicrobial Zeolitic Imidazolate Frameworks with Dual Mechanisms of Action. ACS Infect Dis 2023; 9:507-517. [PMID: 36815744 DOI: 10.1021/acsinfecdis.2c00496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
The horizontal transfer of drug-resistant genes and the formation of biofilm barriers have threatened the therapeutic efficacy of conventional antibiotic drugs. Development of non-antibiotic agents with high delivery efficiency through bacterial biofilms is urgently required. A pyrithione (PT)-loading zeolitic imidazolate framework (ZIF-8@PT) is synthesized to destroy biofilms and improve the sensitivity of bacteria to PT. ZIF-8@PT can target and destroy the biofilm as well as the cell membrane, promoting the intracellular delivery of PT and possibly its interaction with SmpB, a protein that could regulate the drug resistance of bacteria. ZIF-8@PT effectively suppresses abdominal infections induced by multiresistant Aeromonas veronii C4 in rodent models without systemic toxicity. ZIF-8@PT promises wide applications in treating infections caused by multidrug-resistant bacteria through a dual mechanism of action.
Collapse
Affiliation(s)
- Yunxiang Yan
- School of Life Sciences, Hainan University, Haikou 570228, China.,One Health Institute, Hainan University, Haikou 570228, China
| | - Ye Li
- Environment and Plant Protection Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Hong Li
- School of Life Sciences, Hainan University, Haikou 570228, China.,One Health Institute, Hainan University, Haikou 570228, China
| | - Xiang Ma
- School of Life Sciences, Hainan University, Haikou 570228, China.,One Health Institute, Hainan University, Haikou 570228, China
| | - Yanqiong Tang
- School of Life Sciences, Hainan University, Haikou 570228, China.,One Health Institute, Hainan University, Haikou 570228, China
| | - Kexian Yi
- Environment and Plant Protection Institute, Chinese Academy of Tropical Agricultural Sciences, Haikou 571101, China
| | - Xiangmin Lin
- Fujian Provincial Key Laboratory of Agroecological Processing and Safety Monitoring, School of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China
| | - Juanjuan Li
- School of Life Sciences, Hainan University, Haikou 570228, China.,One Health Institute, Hainan University, Haikou 570228, China
| | - Zhu Liu
- School of Life Sciences, Hainan University, Haikou 570228, China.,One Health Institute, Hainan University, Haikou 570228, China
| |
Collapse
|
20
|
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: 8] [Impact Index Per Article: 8.0] [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
|
21
|
Liu F, Cheng Z, Yi H. NIR light-activatable dissolving microneedle system for melanoma ablation enabled by a combination of ROS-responsive chemotherapy and phototherapy. J Nanobiotechnology 2023; 21:61. [PMID: 36814244 PMCID: PMC9948357 DOI: 10.1186/s12951-023-01815-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 02/14/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND As a consequence of the aggressive and recurrent nature of melanoma, repeated, multimodal treatments are often necessary to cure the disease. While microneedle (MN)-based transdermal drug delivery methods can allow drugs to avoid first-pass metabolism and overcome the stratum corneum barrier, the main challenges of these delivery methods entail the lack of controlled drug release/activation and effective imaging methods to guide the entire treatment process. METHODS To enable a transdermal delivery method with controllable drug release/activation and effective imaging guidance, we designed a near-infrared (NIR) photoactivatable, dissolving MN system comprising dissolvable polyvinylpyrrolidone MNs arrays (MN-pB/I) containing liposomes that were co-loaded with the photosensitizer indocyanine green (ICG) and the reactive oxygen species (ROS)-activatable prodrug of doxorubicin (pB-DOX). RESULTS After applying the MN patch to the tumor site, the liposomes concentrated in the needle tips were released into the tumor tissue and distributed evenly upon dissolution of the matrix to enable targeted delivery. Then, the ROS produced by ICG after exposure to NIR light performed photodynamic therapy and activated the pB-DOX for chemotherapy by cleaving the prodrug moiety and converting it to DOX. As a dye, ICG was also used to guide the treatment regimens and monitor the efficacy by fluorescence and photoacoustic imaging. The growth of the tumors in the MN-pB/I group were inhibited by 93.5%, while those were only partially inhibited in the control groups. Negligible treatment-induced side effects and cardiotoxicity were observed. CONCLUSION The MN-pB/I represents a multimodal, biocompatible theragnostic system with spatiotemporal control that was capable of ablating melanoma tumors after a single dose, providing a promising candidate for clinical melanoma therapy.
Collapse
Affiliation(s)
- Fan Liu
- grid.216417.70000 0001 0379 7164Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Zeneng Cheng
- grid.216417.70000 0001 0379 7164Division of Biopharmaceutics and Pharmacokinetics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Hanxi Yi
- Department of Pathology, School of Basic Medical Science, Central South University, Tongzipo Road 172, Changsha, 410000, China. .,Department of Pathology, Xiangya Hospital, Ultrapathology (Biomedical Electron Microscopy) Center, Central South University, Changsha, China.
| |
Collapse
|
22
|
Hu H, Song Q, Yang W, Zeng Q, Liang Z, Liu W, Shao Z, Zhang Y, Chen C, Wang B. Oxidative stress induced by berberine-based mitochondria-targeted low temperature photothermal therapy. Front Chem 2023; 11:1114434. [PMID: 36817173 PMCID: PMC9932336 DOI: 10.3389/fchem.2023.1114434] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/17/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction: Mitochondria-targeted low-temperature photothermal therapy (LPTT) is a promising strategy that could maximize anticancer effects and overcome tumor thermal resistance. However, the successful synthesis of mitochondria-targeted nanodrug delivery system for LPTT still faces diverse challenges, such as laborious preparations processes, low drug-loading, and significant systemic toxicity from the carriers. Methods: In this study, we used the tumor-targeting folic acid (FA) and mitochondria-targeting berberine (BBR) derivatives (BD) co-modified polyethylene glycol (PEG)-decorated graphene oxide (GO) to synthesize a novel mitochondria-targeting nanocomposite (GO-PEG-FA/BD), which can effectively accumulate in mitochondria of the osteosarcoma (OS) cells and achieve enhanced mitochondria-targeted LPTT effects with minimal cell toxicity. The mitochondria-targeted LPTT effects were validated both in vitro and vivo. Results: In vitro experiments, the nanocomposites (GO-PEG-FA/BD) could eliminate membrane potential (ΔΨm), deprive the ATP of cancer cells, and increase the levels of reactive oxygen species (ROS), which ultimately induce oxidative stress damage. Furthermore, in vivo results showed that the enhanced mitochondria-targeted LPTT could exert an excellent anti-cancer effect with minimal toxicity. Discussion: Taken together, this study provides a practicable strategy to develop an ingenious nanoplatform for cancer synergetic therapy via mitochondria-targeted LPTT, which hold enormous potential for future clinical translation.
Collapse
Affiliation(s)
- Hongzhi Hu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijazhuang, China
| | - Qingcheng Song
- Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijazhuang, China
| | - Wenbo Yang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianwen Zeng
- School of Nursing, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zihui Liang
- Ministry of Education Key Laboratory for the Green Preparation and Application of Functional Materials, Faculty of Materials Science and Engineering, Hubei University, Wuhan, China
| | - Weijian Liu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,Department of Orthopaedic Surgery, The Third Hospital of Hebei Medical University, Shijazhuang, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiran Zhang
- School of Medicine, Nankai University, Tianjin, China,*Correspondence: Yiran Zhang, ; Chao Chen, ; Baichuan Wang,
| | - Chao Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Yiran Zhang, ; Chao Chen, ; Baichuan Wang,
| | - Baichuan Wang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Yiran Zhang, ; Chao Chen, ; Baichuan Wang,
| |
Collapse
|
23
|
Ma Z, Han H, Zhao Y. Mitochondrial dysfunction-targeted nanosystems for precise tumor therapeutics. Biomaterials 2023; 293:121947. [PMID: 36512861 DOI: 10.1016/j.biomaterials.2022.121947] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/16/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022]
Abstract
Mitochondria play critical roles in the regulation of the proliferation and apoptosis of cancerous cells. Targeted induction of mitochondrial dysfunction in cancer cells by multifunctional nanosystems for cancer treatment has attracted increasing attention in the past few years. Numerous therapeutic nanosystems have been designed for precise tumor therapy by inducing mitochondrial dysfunction, including reducing adenosine triphosphate, breaking redox homeostasis, inhibiting glycolysis, regulating proteins, membrane potential depolarization, mtDNA damage, mitophagy dysregulation and so on. Understanding the mechanisms of mitochondrial dysfunction would be helpful for efficient treatment of diseases and accelerating the translation of these therapeutic strategies into the clinic. Then, various strategies to construct mitochondria-targeted nanosystems and induce mitochondrial dysfunction are summarized, and the recent research progress regarding precise tumor therapeutics is highlighted. Finally, the major challenges and an outlook in this rapidly developing field are discussed. This review is expected to inspire further development of novel mitochondrial dysfunction-based strategies for precise treatments of cancer and other human diseases.
Collapse
Affiliation(s)
- Zhaoyu Ma
- The State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Science, Huazhong Agricultural University, Wuhan 430070, PR China; School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Heyou Han
- The State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, College of Science, Huazhong Agricultural University, Wuhan 430070, PR China.
| | - Yanli Zhao
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore.
| |
Collapse
|
24
|
Chen L, Jiang X, Liu Q, Tang Z, Wang D, Xiang Z, Liu S, Tang H. A dual-targeting near-infrared biomimetic drug delivery system for HBV treatment. J Med Virol 2023; 95:e28312. [PMID: 36404678 DOI: 10.1002/jmv.28312] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 09/23/2022] [Accepted: 11/12/2022] [Indexed: 11/22/2022]
Abstract
Hepatitis B virus (HBV) infection is a serious global public health threat. It remains elusive to achieve a functional HBV cure with currently available antivirals. Herein, a photo-responsive delivery vehicle composed of Nd3+ -sensitized core-shell upconversion nanoparticle (UCNP), mesoporous silica nanoparticle (MSN), antisense oligonucleotides (ASOs), and capsid-binding inhibitor C39 was established, which was named UMAC according to the initials of its components. Subsequently, the as-synthesized delivery vehicle was encapsulated by β- D-galactopyranoside (Gal) modified red blood cell (RBC) membrane vesicles, which enabled precise targeting of the liver cells (UMAC-M-Gal). Both in vitro and in vivo experiments demonstrated that this biomimetic system could successfully achieve controlled drug release under light conditions at 808 nm, leading to effective suppression of HBV replication in this dual-targeted therapeutic approach. Together, these results substantiate the system has huge prospects for application to achieve functional HBV cure, and provides a promising novel strategy for drug delivery.
Collapse
Affiliation(s)
- Liuxian Chen
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Xinyun Jiang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, China
| | - Qiang Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenrong Tang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Wang
- The People's Hospital of Rongchang District, Chongqing, China
| | - Zheng Xiang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengchun Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hua Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Chongqing Medical University, Chongqing, China
| |
Collapse
|
25
|
Chaudhuri A, Ramesh K, Kumar DN, Dehari D, Singh S, Kumar D, Agrawal AK. Polymeric micelles: A novel drug delivery system for the treatment of breast cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
26
|
Dou R, Cai X, Ruan L, Zhang J, Rouzi A, Chen J, Chai Z, Hu Y. Precision Nanomedicines: Targeting Hot Mitochondria in Cancer Cells. ACS APPLIED BIO MATERIALS 2022; 5:4103-4117. [PMID: 36066886 DOI: 10.1021/acsabm.2c00641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Mitochondrion is a multifunctional organelle in a cell, and it is one of the important targets of antitumor therapy. Conventional mitochondrial targeting strategies can hardly distinguish the mitochondria in cancer cells from those in normal cells, which might raise a concern about the biosafety. Recent studies suggest that a relatively high temperature of mitochondria exists in cancer cells. We named it tumor intrinsic mitochondrial overheating (TIMO). By taking advantage of the difference in mitochondrial temperatures between cancer cells and normal cells, therapeutic agents can be specifically delivered to the mitochondria in cancer cells. Here we will briefly overview the mitochondria-targeted delivery strategies. In addition, the recent discovery of hot mitochondria in cancer cells and the development of mitochondrial temperature-responsive delivery systems for antitumor therapy will be reviewed.
Collapse
Affiliation(s)
- Rui Dou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Beijing 100049, P. R. China
| | - Xiaomeng Cai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Beijing 100049, P. R. China
| | - Lifo Ruan
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Beijing 100049, P. R. China
| | - Jiayu Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Beijing 100049, P. R. China
| | - Aisha Rouzi
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Beijing 100049, P. R. China
| | - Jun Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Beijing 100049, P. R. China
| | - Zhifang Chai
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Beijing 100049, P. R. China
| | - Yi Hu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Multi-disciplinary Research Division, Institute of High Energy Physics and University of Chinese Academy of Sciences (UCAS), Chinese Academy of Sciences (CAS), Beijing 100049, P. R. China
| |
Collapse
|
27
|
Shi Y, Luo Z, You J. Subcellular delivery of lipid nanoparticles to endoplasmic reticulum and mitochondria. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1803. [PMID: 35441489 DOI: 10.1002/wnan.1803] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/23/2022] [Accepted: 03/27/2022] [Indexed: 06/14/2023]
Abstract
Primarily responsible for the biogenesis and metabolism of biomolecules, endoplasmic reticulum (ER) and mitochondria are gradually becoming the targets of therapeutic modulation, whose physiological activities and pathological manifestations determine the functional capacity and even the survival of cells. Drug delivery systems with specific physicochemical properties (passive targeting), or modified by small molecular compounds, polypeptides, and biomembranes demonstrating tropism for ER and mitochondria (active targeting) are able to reduce the nonselective accumulation of drugs, enhancing efficacy while reducing side effects. Lipid nanoparticles feature high biocompatibility, diverse cargo loading, and flexible structure modification, which are frequently used for subcellular organelle-targeted delivery of therapeutics. However, there is still a lack of systematic understanding of lipid nanoparticle-based ER and mitochondria targeting. Herein, we review the pathological significance of drug selectively delivered to the ER and mitochondria. We also summarize the molecular basis and application prospects of lipid nanoparticle-based ER and mitochondria targeting strategies, which may provide guidance for the prevention and treatment of associated diseases and disorders. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease Biology-Inspired Nanomaterials > Lipid-Based Structures Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Diagnostic Tools > In Vivo Nanodiagnostics and Imaging.
Collapse
Affiliation(s)
- Yingying Shi
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhenyu Luo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jian You
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Mitochondrial targeting theranostic nanomedicine and molecular biomarkers for efficient cancer diagnosis and therapy. Biomed Pharmacother 2022; 153:113451. [DOI: 10.1016/j.biopha.2022.113451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/12/2022] [Accepted: 07/18/2022] [Indexed: 01/10/2023] Open
|
29
|
Ren L, Xu P, Yao J, Wang Z, Shi K, Han W, Wang H. Targeting the Mitochondria with Pseudo-Stealthy Nanotaxanes to Impair Mitochondrial Biogenesis for Effective Cancer Treatment. ACS NANO 2022; 16:10242-10259. [PMID: 35820199 DOI: 10.1021/acsnano.1c08008] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The clinical success of anticancer therapy is usually limited by drug resistance and the metastatic dissemination of cancer cells. Mitochondria are essential generators of cellular energy and play a crucial role in sustaining cell survival and metastatic escape. Selective drug strategies targeting mitochondria are able to rewire mitochondrial metabolism and may provide an alternative paradigm to treat many aggressive cancers with high efficiency and low toxicity. Here, we present a pseudo-stealthy mitochondria-targeted pro-nanotaxane and test it against recurrent and metastatic tumor xenografts. The nanoparticle encapsulates a mitochondria-targetable pro-taxane agent, which can be converted into the chemically unmodified cabazitaxel drug, with further surface cloaking with a low-density lipophilic triphenylphosphonium cation. The resultant nanotaxane could be effectively taken up by cells and consequently specifically localized to the mitochondria. The in situ activated cabazitaxel causes mitochondrial dysfunction and ultimately results in potent cell apoptosis. After intravenous administration to animals, pro-nanotaxane mimics the stealthy behavior of polyethylene glycol-cloaked nanoparticles to provide a long circulation time. The antitumor efficacy of this mitochondria-targeted system was validated in multiple preclinical drug-resistant tumor models. Notably, in a patient-derived metastatic melanoma model that was initially pretreated with cabazitaxel, nanotaxane administration not only produced durable tumor reduction but also substantially suppressed metastatic recurrence. Taken together, these results demonstrate that this combination of a pseudo-stealthy platform with a rationally designed pro-drug is an attractive approach to target mitochondria and enhance drug efficacy.
Collapse
Affiliation(s)
- Lulu Ren
- NHC Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, People's Republic of China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, People's Republic of China
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People's Republic of China
| | - Peirong Xu
- NHC Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, People's Republic of China
- Department of Chemical Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, People's Republic of China
| | - Jie Yao
- NHC Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, People's Republic of China
- Department of Chemical Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, People's Republic of China
| | - Zihan Wang
- College of Animal Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Kewei Shi
- NHC Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, People's Republic of China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310016, People's Republic of China
| | - Hangxiang Wang
- NHC Key Laboratory of Combined Multi-Organ Transplantation, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, People's Republic of China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong 250117, People's Republic of China
| |
Collapse
|
30
|
Zeng Y, Yu T, Zhang S, Song G, Meng T, Yuan H, Hu F. Combination of tumor vessel normalization and immune checkpoint blockade for breast cancer treatment via multifunctional nanocomplexes. Biomater Sci 2022; 10:4140-4155. [PMID: 35726757 DOI: 10.1039/d2bm00600f] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumor vessel normalization can alleviate hypoxia, reduce the intratumoral infiltration of immunosuppressive cells and increase the intratumoral infiltration of immune effector cells (CD8+ T cells), further reversing the immunosuppressive microenvironment. Here, nanocomplexes (lipo/St@FA-COSA/BMS-202) which can accurately deliver drugs to tumor tissues and release different drugs at different sites with different rates were prepared to combine tumor vessel normalization with immune checkpoint blockade. The results of drug release in vitro showed that in a pH 6.5 release medium, lipo/St@FA-COSA/BMS-202 rapidly released the vascular normalizing drug (sunitinib, St) and slowly released the PD-1/PD-L1-blocking drug (BMS-202). The results of in vivo experiments showed that the rapidly released St normalized tumor vessels and formed an immunosupportive microenvironment which improved the anti-tumor efficacy of BMS-202. In conclusion, the drug delivery strategy significantly inhibited tumor growth and had excellent anti-tumor efficacy, which can provide a potential approach for effective tumor treatment.
Collapse
Affiliation(s)
- Yingping Zeng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Tong Yu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Shufen Zhang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Guangtao Song
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Tingting Meng
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
31
|
González-Rubio S, Salgado C, Manzaneda-González V, Muñoz-Úbeda M, Ahijado-Guzmán R, Natale P, Almendro-Vedia VG, Junquera E, Barcina JO, Ferrer I, Guerrero-Martínez A, Paz-Ares L, López-Montero I. Tunable gold nanorod/NAO conjugates for selective drug delivery in mitochondria-targeted cancer therapy. NANOSCALE 2022; 14:8028-8040. [PMID: 35616261 DOI: 10.1039/d2nr02353a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Nonyl acridine orange (NAO) is a lipophilic and positively charged molecule widely used as a mitochondrial fluorescent probe. NAO is cytotoxic at micromolar concentration and might be potentially used as a mitochondria-targeted drug for cancer therapy. However, the use of NAO under in vivo conditions would be compromised by the unspecific interactions with off-target cells and negatively charged proteins present in the bloodstream. To tackle this limitation, we have synthesized NAO analogues carrying an imidazole group for their specific binding to nitrilotriacetic (NTA) functionalized gold nanorods (AuNRs). We demonstrate that AuNRs provide 104 binding sites and a controlled delivery under acidic conditions. Upon incubation with mouse embryonic fibroblasts, the endosomal acidic environment releases the NAO analogues from AuNRs, as visualized through the staining of the mitochondrial network. The addition of the monoclonal antibody Cetuximab to the conjugates enhanced their uptake within lung cancer cells and the conjugates were cytotoxic at subnanomolar concentrations (c50 ≈ 0.06 nM). Moreover, the specific interactions of Cetuximab with the epidermal growth factor receptor (EGFR) provided a specific targeting of EGFR-expressing lung cancer cells. After intravenous administration in patient-derived xenografts (PDX) mouse models, the conjugates reduced the progression of EGFR-positive tumors. Overall, the NAO-AuNRs provide a promising strategy to realize membrane mitochondria-targeted conjugates for lung cancer therapy.
Collapse
Affiliation(s)
- Sergio González-Rubio
- Departamento Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain.
- Instituto de Investigación Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain.
| | - Cástor Salgado
- Departamento Química Orgánica, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain
| | - Vanesa Manzaneda-González
- Departamento Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain.
| | - Mónica Muñoz-Úbeda
- Departamento Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain.
- Instituto de Investigación Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain.
| | - Rubén Ahijado-Guzmán
- Departamento Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain.
| | - Paolo Natale
- Departamento Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain.
- Instituto de Investigación Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain.
| | - Víctor G Almendro-Vedia
- Departamento Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain.
- Instituto de Investigación Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain.
| | - Elena Junquera
- Departamento Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain.
| | - José Osío Barcina
- Departamento Química Orgánica, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain
| | - Irene Ferrer
- Instituto de Investigación Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain.
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Ciberonc, Madrid, Spain
| | - Andrés Guerrero-Martínez
- Departamento Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain.
| | - Luis Paz-Ares
- Instituto de Investigación Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain.
- Centro Nacional de Investigaciones Oncológicas (CNIO), Madrid, Spain
- Ciberonc, Madrid, Spain
- Departamento de Medicina, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain
| | - Iván López-Montero
- Departamento Química Física, Universidad Complutense de Madrid, Avda. Complutense s/n, 28040 Madrid, Spain.
- Instituto de Investigación Hospital Doce de Octubre (imas12), Avenida de Córdoba s/n, 28041 Madrid, Spain.
- Instituto Pluridisciplinar, Ps. Juan XXIII 1, 28040 Madrid, Spain
| |
Collapse
|
32
|
Xiao Y, Li X, Mao J, Zheng H, Ji R, Wang Z, Guo M, Yuan H, Anwar A, Chen C, Liu X, Liu J. Reverse anti-breast cancer drug resistance effects by a novel two-step assembled nano-celastrol medicine. NANOSCALE 2022; 14:7856-7863. [PMID: 35583119 DOI: 10.1039/d2nr02064e] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Multidrug resistance (MDR) has become one of the most intractable problems in clinics as it would cause failure in chemotherapy. In this study, we demonstrated that a nanoscale self-assembled nanomedicine, which almost consisted of a pure chemo-drug, could efficiently overcome MDR. Celastrol (CST) was directly assembled into a discrete nanomedicine by precipitation, and then CST nanoparticles (CNPs) inhibited drug efflux pumps by activating HSF-1 expression and promoting HSF-1 translocation into nucleus to suppress the Pgp expression. The more drug accumulated in cells could activate apoptosis signals simultaneously and realize drug resistance reversal. CNPs significantly increased the level of ROS to regulate ERK/JNK signaling, which would further induce resistant cell apoptosis. The tandem apoptosis strategy used the same concentration of CST but achieved a higher antitumor effect. Overall, our study provides a new translational and alternative strategy using conventional natural products to overcome MDR with high efficacy.
Collapse
Affiliation(s)
- Yating Xiao
- School of Molecular Medicine, Hangzhou Institute for Advanced Study, UCAS, Hangzhou 310024, China
| | - Xun Li
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiahui Mao
- Key Laboratory of Resource Biology and Biotechnology in Western China Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, P. R. China.
| | - Hong Zheng
- Key Laboratory of Resource Biology and Biotechnology in Western China Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, P. R. China.
| | - Rui Ji
- Key Laboratory of Resource Biology and Biotechnology in Western China Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, P. R. China.
| | - Zhice Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, P. R. China.
| | - Mengyu Guo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Hui Yuan
- Department of Etiology and Carcinogenesis, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Arfidin Anwar
- School of Chemical Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| | - Xingjun Liu
- Department of Endocrinology, First Hospital Affiliated to Medical College of Xi'an Jiaotong University, Xi'an, P.R. China.
| | - Jing Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China Ministry of Education, School of Medicine, Northwest University, Xi'an 710069, P. R. China.
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellent in Nanoscience, National Center for Nanoscience and Technology of China, University of Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
33
|
Castillo SR, Rickeard BW, DiPasquale M, Nguyen MHL, Lewis-Laurent A, Doktorova M, Kav B, Miettinen MS, Nagao M, Kelley EG, Marquardt D. Probing the Link between Pancratistatin and Mitochondrial Apoptosis through Changes in the Membrane Dynamics on the Nanoscale. Mol Pharm 2022; 19:1839-1852. [PMID: 35559658 DOI: 10.1021/acs.molpharmaceut.1c00926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Pancratistatin (PST) is a natural antiviral alkaloid that has demonstrated specificity toward cancerous cells and explicitly targets the mitochondria. PST initiates apoptosis while leaving healthy, noncancerous cells unscathed. However, the manner by which PST induces apoptosis remains elusive and impedes the advancement of PST as a natural anticancer therapeutic agent. Herein, we use neutron spin-echo (NSE) spectroscopy, molecular dynamics (MD) simulations, and supporting small angle scattering techniques to study PST's effect on membrane dynamics using biologically representative model membranes. Our data suggests that PST stiffens the inner mitochondrial membrane (IMM) by being preferentially associated with cardiolipin, which would lead to the relocation and release of cytochrome c. Second, PST has an ordering effect on the lipids and disrupts their distribution within the IMM, which would interfere with the maintenance and functionality of the active forms of proteins in the electron transport chain. These previously unreported findings implicate PST's effect on mitochondrial apoptosis.
Collapse
Affiliation(s)
- Stuart R Castillo
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| | - Brett W Rickeard
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| | - Mitchell DiPasquale
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| | - Michael H L Nguyen
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| | - Aislyn Lewis-Laurent
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| | - Milka Doktorova
- Department of Molecular Physiology and Biological Physics, University of Virginia School of Medicine, Charlottesville, Virginia 22903, United States
| | - Batuhan Kav
- Max-Planck Institute of Colloids and Interfaces, Potsdam 14476, Germany.,Institute of Biological Information Processing: Structural Biochemistry (IBI-7), Forschungszentrum Julich, Julich 52428, Germany
| | | | - Michihiro Nagao
- National Institute of Standards and Technology, Center for Neutron Research, Gaithersburg, Maryland 20899, United States.,Department of Materials Science and Engineering, University of Maryland, College Park, Maryland 20742, United States.,Department of Physics and Astronomy, University of Delaware, Newark, Delaware 19716, United States
| | - Elizabeth G Kelley
- National Institute of Standards and Technology, Center for Neutron Research, Gaithersburg, Maryland 20899, United States
| | - Drew Marquardt
- Department of Chemistry and Biochemistry, University of Windsor, Windsor, Ontario N9B 3P4, Canada.,Department of Physics, University of Windsor, Windsor, Ontario N9B 3P4, Canada
| |
Collapse
|
34
|
Luo W, Zhang S, Ye J, Jiang B, Meng Q, Zhang G, Li J, Tang Y. A multimodal fluorescent probe for portable colorimetric detection of pH and it's application in mitochondrial bioimaging. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2022; 267:120554. [PMID: 34749111 DOI: 10.1016/j.saa.2021.120554] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 06/13/2023]
Abstract
Mitochondria, as vital energy supplying organelles, play important roles in cellular metabolism, which are closely related with mitochondrial pH (∼8.0). In this work, a novel multimodal fluorescent probe was employed for ratiometric and colorimetric detection of pH. The probe is designed to work by controlling benzothiazole phenol-hemicyanine system as the interaction site and hemicyanine connected by conjugate bonds as the mitochondrial targeting, which also could make the fluorescence of probe red-shifted. This system results in a perfect ratiometric fluorescent response, whose emission changed from red to blue under pH 2.0-10.0, having a broad linear range (pH = 3.0-10.0). And the marked colour change (light yellow to deep purple via naked eye under pH 2.0-11.0) could be used to construct the test strip colorimetry and smartphone APP detection method, realizing the fast, portable, and accurate detection of pH in vitro and environment. Besides, the probe owns the characteristics of easy loading, high selectivity and staining ability of mitochondria, and low cytotoxicity, thereby allowing imaging of pH values and real-time monitor the subcellular mitochondria pH changes caused by drugs in living cells. It thus could be used to monitor the organ-specific dynamics related to transitions between pathological and physiological states.
Collapse
Affiliation(s)
- Weifang Luo
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China.
| | - Shuan Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China
| | - Jing Ye
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China
| | - Bohong Jiang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China
| | - Qinghua Meng
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China
| | - Guanghui Zhang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China
| | - Jiayi Li
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China
| | - Yuping Tang
- State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an 712046, Shaanxi Province, China
| |
Collapse
|
35
|
Bajpai A, Desai NN, Pandey S, Shukla C, Datta B, Basu S. Chimeric nanoparticles for targeting mitochondria in cancer cells. NANOSCALE ADVANCES 2022; 4:1112-1118. [PMID: 36131756 PMCID: PMC9419202 DOI: 10.1039/d1na00644d] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 01/07/2022] [Indexed: 06/15/2023]
Abstract
Mitochondrial dysfunction is implicated in myriad diseases, including cancer. Subsequently, targeting mitochondrial DNA (mt-DNA) in cancer cells has emerged as an unorthodox strategy for anti-cancer therapy. However, approaches targeting only one component of the mitochondrial "central dogma" can be evaded by cancer cells through various mechanisms. To address this, herein, we have engineered mitochondria-targeting cholesterol-based chimeric nanoparticles (mt-CNPs) consisting of cisplatin, camptothecin, and tigecycline, which can simultaneously impair mt-DNA, mitochondrial topoisomerase I (mt-Top1), and mitochondrial ribosomes. mt-CNPs were characterized as being positively charged, spherical in shape, and 187 nm in diameter. Confocal microscopy confirmed that mt-CNPs efficiently localized into the mitochondria of A549 lung cancer cells within 6 h, followed by mitochondrial morphology damage and the subsequent generation of reactive oxygen species (ROS). mt-CNPs showed remarkable cancer-cell killing abilities compared to free-drug combinations in A549 (lung), HeLa (cervical), and MCF7 (breast) cancer cells. These mitochondria-targeting lipidic chimeric nanoparticles could be explored further to impair multiple targets in mitochondria, helping researchers to gain an understanding of mitochondrial translational and transcriptional machinery and to develop new strategies for cancer therapy.
Collapse
Affiliation(s)
- Aman Bajpai
- Discipline of Chemistry, Indian Institute of Technology (IIT) Gandhinagar Palaj Gandhinagar Gujarat 382355 India
| | - Nakshi Nayan Desai
- Discipline of Biological Engineering, Indian Institute of Technology (IIT) Gandhinagar Palaj Gandhinagar Gujarat 382355 India
| | - Shalini Pandey
- Discipline of Chemistry, Indian Institute of Technology (IIT) Gandhinagar Palaj Gandhinagar Gujarat 382355 India
| | - Chinmayee Shukla
- Discipline of Biological Engineering, Indian Institute of Technology (IIT) Gandhinagar Palaj Gandhinagar Gujarat 382355 India
| | - Bhaskar Datta
- Discipline of Chemistry, Indian Institute of Technology (IIT) Gandhinagar Palaj Gandhinagar Gujarat 382355 India
- Discipline of Biological Engineering, Indian Institute of Technology (IIT) Gandhinagar Palaj Gandhinagar Gujarat 382355 India
| | - Sudipta Basu
- Discipline of Chemistry, Indian Institute of Technology (IIT) Gandhinagar Palaj Gandhinagar Gujarat 382355 India
| |
Collapse
|
36
|
Regulated preparation of celastrol-loaded nanoparticle by flash nanoprecipitation. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
37
|
Pan Z, Yang G, Yuan J, Pan M, Li J, Tan H. Effect of the disulfide bond and polyethylene glycol on the degradation and biophysicochemical properties of polyurethane micelles. Biomater Sci 2022; 10:794-807. [PMID: 34988575 DOI: 10.1039/d1bm01422f] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The disulfide bond has emerged as a promising redox-sensitive switch for smart polymeric micelles, due to its properties of rapid response to the reductive environment and spatiotemporally-controlled therapeutic agent delivery. However, the dilemma of multifunctional nanomedicine is that the more intelligent the functionalities integrated into a system, the vaguer the understanding of the structure and interaction between the multi-functional moieties becomes. To better understand the interaction between the disulfide bond and methoxy polyethylene glycol (mPEG), and their effects on the biophysicochemical characterization of micelles, we developed a series of polyurethane micelles containing various densities of disulfide bonds and bearing different molecular weights of mPEG. In this work, we found that the critical factor determining the degradation rate of polymer micelles was the hydrophobic/hydrophilic ratio of broken polymer segments triggered by disulfide bond breaking. The higher density of the disulfide bond and longer mPEG chain accelerate the degradation process due to the disproportionate hydrophobic/hydrophilic ratio of the broken chain, which is the key factor to determine the micellization and stabilization of polymer micelles. This work provides a fundamental understanding of the interaction between the complex functional groups and a new insight into the mechanism of the micelle degradation process, offering guidance on the rational design and fabrication of multifunctional nanoformulations.
Collapse
Affiliation(s)
- Zhicheng Pan
- Hebei Key Laboratory of Functional Polymers, Department of Polymer Materials and Engineering, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Guangxuan Yang
- Hebei Key Laboratory of Functional Polymers, Department of Polymer Materials and Engineering, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Jinfeng Yuan
- Hebei Key Laboratory of Functional Polymers, Department of Polymer Materials and Engineering, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Mingwang Pan
- Hebei Key Laboratory of Functional Polymers, Department of Polymer Materials and Engineering, School of Chemical Engineering and Technology, Hebei University of Technology, Tianjin 300130, China
| | - Jiehua Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
38
|
Hu T, Qin Z, Shen C, Gong HL, He ZY. Multifunctional Mitochondria-Targeting Nanosystems for Enhanced Anticancer Efficacy. Front Bioeng Biotechnol 2021; 9:786621. [PMID: 34900973 PMCID: PMC8652136 DOI: 10.3389/fbioe.2021.786621] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/01/2021] [Indexed: 02/05/2023] Open
Abstract
Mitochondria, a kind of subcellular organelle, play crucial roles in cancer cells as an energy source and as a generator of reactive substrates, which concern the generation, proliferation, drug resistance, and other functions of cancer. Therefore, precise delivery of anticancer agents to mitochondria can be a novel strategy for enhanced cancer treatment. Mitochondria have a four-layer structure with a high negative potential, which thereby prevents many molecules from reaching the mitochondria. Luckily, the advances in nanosystems have provided enormous hope to overcome this challenge. These nanosystems include liposomes, nanoparticles, and nanomicelles. Here, we summarize the very latest developments in mitochondria-targeting nanomedicines in cancer treatment as well as focus on designing multifunctional mitochondria-targeting nanosystems based on the latest nanotechnology.
Collapse
Affiliation(s)
- Tingting Hu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zhou Qin
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Shen
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Han-Lin Gong
- Department of Integrated Traditional Chinese and Western Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Zhi-Yao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.,Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Yi X, Yan Y, Li L, Zhou R, Shen X, Huang Y. Combination of mitochondria impairment and inflammation blockade to combat metastasis. J Control Release 2021; 341:753-768. [PMID: 34915072 DOI: 10.1016/j.jconrel.2021.12.015] [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] [Received: 08/15/2021] [Revised: 12/09/2021] [Accepted: 12/09/2021] [Indexed: 12/16/2022]
Abstract
Targeted induction of mitochondria impairment has emerged as a promising strategy for anti-metastasis therapy. However, problems such as limited mitochondria targeting efficiency, undesired drug leakage and insufficient drug release inside mitochondria remain crucial challenges for mitochondria-targeting therapy. Here, we constructed an N-(2-hydroxypropyl) methacrylamide (HPMA) polymer based cationic system that could target to mitochondria and facilitate on demand drug release in response to excessive mitochondrial reactive oxygen species. Whereas, this drug delivery system is still challenged by limitations of (1) in vivo application, and (2) inflammatory tumor microenvironment (TME). On one aspect, to prolong blood circulation and increase tumor targeting, we designed a nanocomposite (PDT-NCs) that assembled from the cationic HPMA polymer and anionic hyaluronic acid via electrostatic interaction. On another aspect, a celecoxib loaded liposome (Lip-Cel) was further fabricated to alleviate inflammation in TME by downregulating various metastasis-associated factors. Ultimately, PDT-NCs and Lip-Cel led to a drastic improvement in the suppression of primary tumor growth and distant lung metastasis. Our work provided a generalizable approach of mitochondria dysfunction and inflammation blockade to combat metastatic tumors.
Collapse
Affiliation(s)
- Xiaoli Yi
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, China
| | - Yue Yan
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, China
| | - Rui Zhou
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, China
| | - Xinran Shen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, China.
| |
Collapse
|
40
|
Tan YN, Li YP, Huang JD, Luo M, Li SS, Lee AWM, Hu FQ, Guan XY. Thermal-sensitive lipid nanoparticles potentiate anti-PD therapy through enhancing drug penetration and T lymphocytes infiltration in metastatic tumor. Cancer Lett 2021; 522:238-254. [PMID: 34571084 DOI: 10.1016/j.canlet.2021.09.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/06/2021] [Accepted: 09/21/2021] [Indexed: 12/12/2022]
Abstract
The response rate of anti-PD therapy in most cancer patients remains low. Therapeutic drug and tumor-infiltrating lymphocytes (TILs) are usually obstructed by the stromal region within tumor microenvironment (TME) rather than distributed around tumor cells, thus unable to induce the immune response of cytotoxic T cells. Here, we constructed the cationic thermosensitive lipid nanoparticles IR780/DPPC/BMS by introducing cationic NIR photosensitizer IR-780 iodide (IR780) modified lipid components, thermosensitive lipid DPPC and PD-1/PD-L1 inhibitor BMS202 (BMS). Upon laser irradiation, IR780/DPPC/BMS penetrated into deep tumor, and reduced cancer-associated fibroblasts (CAFs) around tumor cells to remodel the spatial distribution of TILs in TME. Interestingly, the cationic IR780/DPPC/BMS could capture released tumor-associated antigens (TAAs), thereby enhancing the antigen-presenting ability of DCs to activate cytotoxic T lymphocytes. Moreover, IR780/DPPC/BMS initiated gel-liquid crystal phase transition under laser irradiation, accelerating the disintegration of lipid bilayer structure and leading to the responsive release of BMS, which would reverse the tumor immunosuppression state by blocking PD-1/PD-L1 pathway for a long term. This combination treatment can synergistically exert the antitumor immune response and inhibit the tumor growth and metastasis.
Collapse
Affiliation(s)
- Ya-Nan Tan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China; CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Yong-Peng Li
- Department of Urology, The Third Affiliated Hospital of Shenzhen University (Luohu Hospital Group), Shenzhen, 518000, China
| | - Jian-Dong Huang
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Min Luo
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Shan-Shan Li
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Anne Wing-Mui Lee
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China; Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 00852, China
| | - Fu-Qiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China
| | - Xin-Yuan Guan
- Department of Clinical Oncology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China; Department of Clinical Oncology, The University of Hong Kong, Hong Kong, 00852, China.
| |
Collapse
|
41
|
Wei G, Chen J, Jing Z, Li Y, Li Z, Zheng W, Sun X, Zhao W, Zhang Z, Wang X, Han H, Li C, Zhang Y, Ma P. Glucose transporter 1 (GLUT1)-targeting and hypoxia-activated mitochondria-specific chemo-thermal therapy via a glycosylated poly(amido amine)/celastrol (PAMAM/Cel) complex. J Colloid Interface Sci 2021; 608:1355-1365. [PMID: 34742058 DOI: 10.1016/j.jcis.2021.10.129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/16/2022]
Abstract
Mitochondria are appealing targets in cancer therapy for providing a suitable microenvironment and energy supply. Herein, we constructed a glycosylated poly(amido amine)/celastrol (PAMAM/Cel) complex for hypoxia-activated mitochondria-specific drug delivery and chemothermal therapy to inhibit tumor growth and metastasis. The complex was characterized by high photothermal conversion efficiency, hypoxia-sensitive polyethylene glycol (PEG) outer layer detachment, and alkaline-sensitive drug release. The complex showed specific cellular uptake in glucose transporter 1 (GLUT1)-overexpressing tumor cells and mitochondrial accumulation in a hypoxic environment. Combined with near-infrared (NIR) laser irradiation, the complex exhibited higher cytotoxicity, apoptosis induction, and metastasis inhibition rates due to the synergistic chemothermal effect. Similarly, the complex also targeted tumors and accumulated in mitochondria in tumor-bearing nude mice, resulting in superior inhibitory effects on tumor growth and metastasis as well as low systematic toxicity. Further mechanistic studies discovered that the complex impaired the mitochondrial membrane, reduced adenosine triphosphate (ATP) content, and regulated metastasis-related protein expression. Thus, the present study provides a promising nanomedicine for tumor therapy.
Collapse
Affiliation(s)
- Guijie Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Jianhua Chen
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ziqi Jing
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yanyi Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhihui Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Zheng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiurui Sun
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Wenwen Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Zhe Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xue Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Hongcui Han
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Chu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yujie Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| | - Pengkai Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
42
|
Concurrent impairment of nucleus and mitochondria for synergistic inhibition of cancer metastasis. Int J Pharm 2021; 608:121077. [PMID: 34487811 DOI: 10.1016/j.ijpharm.2021.121077] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 08/23/2021] [Accepted: 09/01/2021] [Indexed: 12/22/2022]
Abstract
Cancer metastasis, which increases the mortality in a short period of time, has been considered as the main challenge in tumor treatment. However, tumor growth suppression also should not be ignored in cancer metastasis treatment. Recently, accumulating evidences have suggested that mitochondria play an important role in mitigating caner metastasis. Nucleus, as the repository of genetic information, plays a key role in cell proliferation. However, it remains elusive that the concurrent impairment of nucleus and mitochondria may achieve better anti-tumor and anti-metastatic effects. Here, we designed a mitochondria-penetrating peptide modified doxorubicin (MPP-Dox) loaded N-(2-hydroxypropyl) methacrylamide (HPMA) copolymer conjugates (PM), as well as a nuclear accumulating HPMA copolymer Dox conjugates (PN) by the nuclear tendency of Dox. After co-delivering the two copolymers (abbreviation for PMN), PM promoted cell apoptosis and inhibited tumor metastasis by damaging mitochondria, whereas PN suppressed cell proliferation and promoted apoptosis by destroying nucleus. Importantly, PM and PN complemented each other as expected. The mitochondrial dysfunction and tumor metastasis inhibition of PM was improved by PN, while cell proliferation suppression and apoptosis by nucleus destroying of PN was enhanced by PM. As a result, tumor growth of breast cancer 4T1 cells in vivo was significantly restrained and lung metastasis was potently decreased and almost eradicated, fully reflecting the advantages of organelle targeting combination therapy. As a consequence, our work showed that concurrent impairment of nucleus and mitochondria was feasible and beneficial to metastatic cancer treatment.
Collapse
|
43
|
Guo L, Zhang Y, Al-Jamal KT. Recent progress in nanotechnology-based drug carriers for celastrol delivery. Biomater Sci 2021; 9:6355-6380. [PMID: 34582530 DOI: 10.1039/d1bm00639h] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Celastrol (CLT) is an active ingredient that was initially discovered and extracted from the root of Tripterygium wilfordii. The potential pharmacological activities of CLT in cancer, obesity, and inflammatory, auto-immune, and neurodegenerative diseases have been demonstrated in recent years. However, CLT's clinical application is extremely restricted by its low solubility/permeability, poor bioavailability, and potential off-target toxicity. The advent of nanotechnology provides a solution to improve the oral bioavailability, therapeutic effects or tissue-targeting ability of CLT. This review focuses on the most recent advances, improvements, inventions, and updated literature of various nanocarrier systems for CLT.
Collapse
Affiliation(s)
- Ling Guo
- Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Dongqing South Road, Huaxi University City, Guiyang, Guizhou 550025, P.R. China.,Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| | - Yongping Zhang
- Guizhou Engineering Technology Research Center for Processing and Preparation of Traditional Chinese Medicine and Ethnic Medicine, College of Pharmaceutical Sciences, Guizhou University of Traditional Chinese Medicine, Dongqing South Road, Huaxi University City, Guiyang, Guizhou 550025, P.R. China.,Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
44
|
Khan S, Vahdani Y, Hussain A, Haghighat S, Heidari F, Nouri M, Haj Bloukh S, Edis Z, Mahdi Nejadi Babadaei M, Ale-Ebrahim M, Hasan A, Sharifi M, Bai Q, Hassan M, Falahati M. Polymeric micelles functionalized with cell penetrating peptides as potential pH-sensitive platforms in drug delivery for cancer therapy: A review. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103264] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
45
|
Qian Y, Zhang J, Xu R, Li Q, Shen Q, Zhu G. Nanoparticles based on polymers modified with pH-sensitive molecular switch and low molecular weight heparin carrying Celastrol and ferrocene for breast cancer treatment. Int J Biol Macromol 2021; 183:2215-2226. [PMID: 34097964 DOI: 10.1016/j.ijbiomac.2021.05.204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 05/27/2021] [Accepted: 05/30/2021] [Indexed: 10/21/2022]
Abstract
Triple negative breast cancer (TNBC) metastasis is still one of the obstacles in clinical treatment, while highly-effective cancer drugs usually cannot be used for their hydrophobicity and comprehensive system toxicity. This study built a kind of pH-sensitive nanoparticles (PP/H NPs) constructed by poly (lactic-co-glycolic acid) modified with β-cyclodextrin (PLGA-β-CD), polyethyleneimine grafted with benzimidazole (PEI-BM) and low molecular weight heparin (LMWH) to delivery Celastrol (Cela) and ferrocene (Fc) for breast cancer therapy. PLGA-β-CD and PEI-BM were synthesized by amidation reaction, the amphipathic polymer nanoparticles with 108.37 ± 1.02 nm were self-assembled in water. After PP/H NPs treatment, the half maximal inhibitory concentration (IC50) decreased by 91% compared with Cela, and ROS level was also elevated. PP/H NPs led to substantial tumor inhibiting rate (TIR, 65.86%), utilized LMWH to strengthen the anti-metastasis effect of PP/H NPs. PP/H NPs took advantage of exogenous chemotherapeutics and endogenous ROS to inhibit tumor growth, and combined with LMWH to hinder breast cancer metastasis.
Collapse
Affiliation(s)
- Yun Qian
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Jun Zhang
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Rui Xu
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Qiang Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China
| | - Qi Shen
- School of Pharmacy, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai 200240, China
| | - Guofu Zhu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Shanghai 201203, China.
| |
Collapse
|
46
|
Choudhary D, Goykar H, Karanwad T, Kannaujia S, Gadekar V, Misra M. An understanding of mitochondria and its role in targeting nanocarriers for diagnosis and treatment of cancer. Asian J Pharm Sci 2021; 16:397-418. [PMID: 34703491 PMCID: PMC8520044 DOI: 10.1016/j.ajps.2020.10.002] [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: 04/26/2020] [Revised: 09/24/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
Nanotechnology has changed the entire paradigm of drug targeting and has shown tremendous potential in the area of cancer therapy due to its specificity. In cancer, several targets have been explored which could be utilized for the better treatment of disease. Mitochondria, the so-called powerhouse of cell, portrays significant role in the survival and death of cells, and has emerged as potential target for cancer therapy. Direct targeting and nanotechnology based approaches can be tailor-made to target mitochondria and thus improve the survival rate of patients suffering from cancer. With this backdrop, in present review, we have reemphasized the role of mitochondria in cancer progression and inhibition, highlighting the different targets that can be explored for targeting of disease. Moreover, we have also summarized different nanoparticulate systems that have been used for treatment of cancer via mitochondrial targeting.
Collapse
Affiliation(s)
- Devendra Choudhary
- National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Palaj, Opp. Air force station headqtrs, Gandhinagar 382355, India
| | - Hanmant Goykar
- National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Palaj, Opp. Air force station headqtrs, Gandhinagar 382355, India
| | - Tukaram Karanwad
- National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Palaj, Opp. Air force station headqtrs, Gandhinagar 382355, India
| | - Suraj Kannaujia
- National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Palaj, Opp. Air force station headqtrs, Gandhinagar 382355, India
| | - Vedant Gadekar
- National Institute of Pharmaceutical Education and Research (NIPER) Ahmedabad, Palaj, Opp. Air force station headqtrs, Gandhinagar 382355, India
| | | |
Collapse
|
47
|
Huang Y, Wang T, Tan Q, He D, Wu M, Fan J, Yang J, Zhong C, Li K, Zhang J. Smart Stimuli-Responsive and Mitochondria Targeting Delivery in Cancer Therapy. Int J Nanomedicine 2021; 16:4117-4146. [PMID: 34163163 PMCID: PMC8214531 DOI: 10.2147/ijn.s315368] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 05/22/2021] [Indexed: 01/02/2023] Open
Abstract
Dysfunction in the mitochondria (Mc) contributes to tumor progression. It is a major challenge to deliver therapeutic agents specifically to the Mc for precise treatment. Smart drug delivery systems are based on stimuli-responsiveness and active targeting. Here, we give a whole list of documented pathways to achieve smart stimuli-responsive (St-) and Mc-targeted DDSs (St-Mc-DDSs) by combining St and Mc targeting strategies. We present the formulations, targeting characteristics of St-Mc-DDSs and clarify their anti-cancer mechanisms as well as improvement in efficacy and safety. St-Mc-DDSs usually not only have Mc-targeting groups, molecules (lipophilic cations, peptides, and aptamers) or materials but also sense the surrounding environment and correspondingly respond to internal biostimulators such as pH, redox changes, enzyme and glucose, and/or externally applied triggers such as light, magnet, temperature and ultrasound. St-Mc-DDSs exquisitely control the action site, increase therapeutic efficacy and decrease side effects of the drug. We summarize the clinical research progress and propose suggestions for follow-up research. St-Mc-DDSs may be an innovative and sensitive precision medicine for cancer treatment.
Collapse
Affiliation(s)
- Yongjia Huang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, People's Republic of China
| | - Tingting Wang
- Biochemistry and Molecular Biology Laboratory, Experimental Teaching and Management Center, Chongqing Medical University, Chongqing, People's Republic of China
| | - Qunyou Tan
- Department of Thoracic Surgery, Daping Hospital of Army Medical University, PLA, Chongqing, People's Republic of China
| | - Dan He
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, People's Republic of China
| | - Mingjun Wu
- Institute of Life Science, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jingchuan Fan
- Institute of Life Science, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jie Yang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, People's Republic of China
| | - Cailing Zhong
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, People's Republic of China
| | - Kailing Li
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jingqing Zhang
- Chongqing Research Center for Pharmaceutical Engineering, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
48
|
Wagh PR, Desai P, Prabhu S, Wang J. Nanotechnology-Based Celastrol Formulations and Their Therapeutic Applications. Front Pharmacol 2021; 12:673209. [PMID: 34177584 PMCID: PMC8226115 DOI: 10.3389/fphar.2021.673209] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 05/10/2021] [Indexed: 12/23/2022] Open
Abstract
Celastrol (also called tripterine) is a quinone methide triterpene isolated from the root extract of Tripterygium wilfordii (thunder god vine in traditional Chinese medicine). Over the past two decades, celastrol has gained wide attention as a potent anti-inflammatory, anti-autoimmune, anti-cancer, anti-oxidant, and neuroprotective agent. However, its clinical translation is very challenging due to its lower aqueous solubility, poor oral bioavailability, and high organ toxicity. To deal with these issues, various formulation strategies have been investigated to augment the overall celastrol efficacy in vivo by attempting to increase the bioavailability and/or reduce the toxicity. Among these, nanotechnology-based celastrol formulations are most widely explored by pharmaceutical scientists worldwide. Based on the survey of literature over the past 15 years, this mini-review is aimed at summarizing a multitude of celastrol nanoformulations that have been developed and tested for various therapeutic applications. In addition, the review highlights the unmet need in the clinical translation of celastrol nanoformulations and the path forward.
Collapse
Affiliation(s)
- Pushkaraj Rajendra Wagh
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - Preshita Desai
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - Sunil Prabhu
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| | - Jeffrey Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, CA, United States
| |
Collapse
|
49
|
S Allemailem K, Almatroudi A, Alsahli MA, Aljaghwani A, M El-Kady A, Rahmani AH, Khan AA. Novel Strategies for Disrupting Cancer-Cell Functions with Mitochondria-Targeted Antitumor Drug-Loaded Nanoformulations. Int J Nanomedicine 2021; 16:3907-3936. [PMID: 34135584 PMCID: PMC8200140 DOI: 10.2147/ijn.s303832] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/24/2021] [Indexed: 12/16/2022] Open
Abstract
Any variation in normal cellular function results in mitochondrial dysregulation that occurs in several diseases, including cancer. Such processes as oxidative stress, metabolism, signaling, and biogenesis play significant roles in cancer initiation and progression. Due to their central role in cellular metabolism, mitochondria are favorable therapeutic targets for the prevention and treatment of conditions like neurodegenerative diseases, diabetes, and cancer. Subcellular mitochondria-specific theranostic nanoformulations for simultaneous targeting, drug delivery, and imaging of these organelles are of immense interest in cancer therapy. It is a challenging task to cross multiple barriers to target mitochondria in diseased cells. To overcome these multiple barriers, several mitochondriotropic nanoformulations have been engineered for the transportation of mitochondria-specific drugs. These nanoformulations include liposomes, dendrimers, carbon nanotubes, polymeric nanoparticles (NPs), and inorganic NPs. These nanoformulations are made mitochondriotropic by conjugating them with moieties like dequalinium, Mito-Porter, triphenylphosphonium, and Mitochondria-penetrating peptides. Most of these nanoformulations are meticulously tailored to control their size, charge, shape, mitochondriotropic drug loading, and specific cell-membrane interactions. Recently, some novel mitochondria-selective antitumor compounds known as mitocans have shown high toxicity against cancer cells. These selective compounds form vicious oxidative stress and reactive oxygen species cycles within cancer cells and ultimately push them to cell death. Nanoformulations approved by the FDA and EMA for clinical applications in cancer patients include Doxil, NK105, and Abraxane. The novel use of these NPs still faces tremendous challenges and an immense amount of research is needed to understand the proper mechanisms of cancer progression and control by these NPs. Here in this review, we summarize current advancements and novel strategies of delivering different anticancer therapeutic agents to mitochondria with the help of various nanoformulations.
Collapse
Affiliation(s)
- Khaled S Allemailem
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Ahmad Almatroudi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Mohammed A Alsahli
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Aseel Aljaghwani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Asmaa M El-Kady
- Department of Medical Parasitology, Faculty of Medicine, South Valley University, Qena, Egypt
| | - Arshad Husain Rahmani
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Amjad Ali Khan
- Department of Basic Health Sciences, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
50
|
Zhou X, Liu X, Yang X, Wang L, Hong Y, Lian K, Qiu G, Shang X, Ma Z, Yuan H, Hu F. Tumor progress intercept by intervening in Caveolin-1 related intercellular communication via ROS-sensitive c-Myc targeting therapy. Biomaterials 2021; 275:120958. [PMID: 34130142 DOI: 10.1016/j.biomaterials.2021.120958] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 04/30/2021] [Accepted: 06/04/2021] [Indexed: 12/24/2022]
Abstract
Tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) play an important role in the development of tumors by secreting a variety of cytokines or directly communicating with tumor cells, making TAMs-targeted therapeutic strategies very attractive. It has been reported that oncogene c-Myc is related to every aspect of the oncogenic process of tumor cells and the alternative activation of macrophages. Hence, we constructed a glycolipid nanocarrier containing ROS-responsive peroxalate linkages (CSOPOSA) for ROS-triggered release of drugs and further modified it with Ex 26 (Ex 26-CSOPOSA), a selective sphingosine 1-phosphate receptor 1 (S1PR1) antagonist, to achieve the dual-targeted delivery of the c-Myc inhibitor JQ1 via S1PR1, which is overexpressed on both tumor cells and TAMs, thereby inducing apoptosis of tumor cells, and blocking M2 polarization of macrophages. More strikingly, our studies found that JQ1 could effectively inhibit the migration of tumor cells induced by M2 macrophages-derived exosomes via blocking Caveolin-1 related intercellular exosome exchange through lncRNA H19 and miR-107. The in vivo results revealed that this dual-targeted delivery strategy effectively inhibited tumor growth and metastasis with less systemic toxicity, providing a potential method for effective tumor treatment.
Collapse
Affiliation(s)
- Xueqing Zhou
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China
| | - Xuan Liu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China
| | - Xiqin Yang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China
| | - Li Wang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China
| | - Yiling Hong
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China
| | - Keke Lian
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China
| | - Guoxi Qiu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China
| | - Xuwei Shang
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China
| | - Zhongjun Ma
- Ocean College, Zhejiang University, Zhoushan, 316021, China
| | - Hong Yuan
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China
| | - Fuqiang Hu
- College of Pharmaceutical Science, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|