1
|
Sun J, An X, Wang Y, Duan X, Pei Z, Lu Y, Pei Y. A hyaluronic acid modified copper-based metal-organic framework overcomes multidrug resistance via two-way redox dyshomeostasis under hypoxia. Int J Biol Macromol 2025; 300:140148. [PMID: 39848376 DOI: 10.1016/j.ijbiomac.2025.140148] [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: 08/04/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
Multidrug resistance (MDR) has become a major challenge in tumor chemotherapy, primarily associated with the overexpression of P-glycoprotein (P-gp). Inhibiting P-gp expression and function through redox dyshomeostasis has shown great potential for reversing MDR. Here, a nanoscale system of copper-based metal-organic framework (HA-CuMOF@DOX) modified with hyaluronic acid (HA) was constructed to overcome MDR via two-way regulation of redox homeostasis under hypoxia. HA-CuMOF@DOX is a spherical glutathione (GSH) responsive nanoparticle with a drug loading capacity of 20.69 %, which could deplete GSH through Cu2+ and electrophilic ligands, and generate •OH via a Fenton-like reaction. In vitro experiments suggested that the nanoparticles had good targetability to cancer cells and biocompatibility to normal cells. HA-CuMOF@DOX was successfully internalized by drug-resistant human hepatoma carcinoma cell line (HepG2-ADR). It aggravated redox dyshomeostasis via dual regulation, inducing mitochondrial damage, reducing intracellular adenosine triphosphate (ATP) levels, and downregulating P-gp to overcome HepG2-ADR drug resistance. More importantly, in vivo experiments demonstrated an 80.69 % tumor growth inhibition in nude mice bearing HepG2-ADR cells. This work represents a significant advancement in the development of effective treatments for drug-resistant tumors.
Collapse
Affiliation(s)
- Jiajia Sun
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xingwang An
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Yi Wang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Xiao Duan
- School of Pharmacy, Changzhi Medical College, Shanxi Provincial Department-Municipal Key Laboratory Cultivation Base for Quality Enhancement and Utilization of Shangdang Chinese Medicinal Materials, Changzhi, Shanxi 046000, PR China
| | - Zhichao Pei
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China
| | - Yuchao Lu
- School of Pharmacy, Changzhi Medical College, Shanxi Provincial Department-Municipal Key Laboratory Cultivation Base for Quality Enhancement and Utilization of Shangdang Chinese Medicinal Materials, Changzhi, Shanxi 046000, PR China.
| | - Yuxin Pei
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, PR China.
| |
Collapse
|
2
|
Barman S, Roy SM, Kishore P, Ghosh M, Bag P, Sarkar AK, Ghatak T, Maji PS, Basu A, Mukherjee R, Ghosh SK, Chowdhury AD, Maity AR. Concurrent targeted delivery of doxorubicin and curcumin to the cancer cells using simple and versatile ligand-installed multifaceted chitosan-based nanoconjugates. J Mater Chem B 2025; 13:2490-2503. [PMID: 39834258 DOI: 10.1039/d4tb01809e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Existing chemotherapeutic approaches against refractory cancers are ineffective due to off-target effects, inefficient delivery, and inadequate accumulation of anticancer drugs at the tumor site, which causes limited efficiency of drug treatment and toxicity to neighboring healthy cells. The development of nano-based drug delivery systems (DDSs) with the goal of delivering desired therapeutic doses to the diseased cells and has already proven to be a promising strategy to address these challenges. Our study focuses on achieving an efficient tumor-targeted delivery of a combination of drugs for therapeutic benefits by developing a versatile DDS by following a simple one-step chemical approach. We used low-molecular-weight chitosan and modified its primary amine groups with reactive forms of cholesterol and folic acid by simple chemical tools and thus prepared folic acid-chitosan-cholesterol graft copolymer. The polymer contains numerous residual primary amine groups, which offer enough water solubility and positive charge to its polymeric backbone to foster the interaction of negatively charged and/or hydrophobic drugs to load and encapsulate a wide variety of drugs within it via various non-bonding interactions. We used curcumin and doxorubicin as the combination of drugs and thus finally prepared targeted nanoconjugates (targeted NCs). In vitro cellular experiments show that our developed targeted NCs demonstrate 3-5 times higher cellular uptake than non-targeted NCs at various incubation times (2 h, 8 h, and 12 h) in KB cells where folate receptors are overexpressed. This enhanced cellular uptake of targeted NCs and the following delivery of drugs in the cytosol and its disposition to the nucleus exhibit a substantial amount of toxicity to KB cells towards an effective therapeutic strategy for treatment.
Collapse
Affiliation(s)
- Sourav Barman
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, 700135, India.
| | - Sayoni Maitra Roy
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, 700135, India.
| | - Purvi Kishore
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, 700135, India.
| | - Malabika Ghosh
- Amity Institute of Nanotechnology, Amity University, Kolkata, West Bengal, 700135, India
| | - Pousali Bag
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, 700135, India.
| | - Ankan Kumar Sarkar
- School of Materials Sciences, Indian Association for the Cultivation of Science, Kolkata, West Bengal, 700032, India
| | - Tapas Ghatak
- Department of Chemistry, School of Advanced Sciences, Vellore Institute of Technology, Vellore 632014, Tamil Nadu, India
| | - Partha Sona Maji
- Department of Physics, Amity Institute of Applied Sciences, Amity University, Kolkata, West Bengal, 700135, India
| | - Arnab Basu
- Department of Biomedical Science and Technology, The School of Biological Sciences, Ramakrishna Mission Vivekananda Educational Research Institute, Belur Math, Howrah, West Bengal, 711202, India
| | - Rupam Mukherjee
- Department of Physics, Presidency University, Bangalore, Karnataka, 560064, India
| | - Surya K Ghosh
- Department of Physics, National Institute of Technology, Warangal, Telangana, 506004, India
| | - Ankan Dutta Chowdhury
- Amity Institute of Nanotechnology, Amity University, Kolkata, West Bengal, 700135, India
| | - Amit Ranjan Maity
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, 700135, India.
| |
Collapse
|
3
|
Zhang J, Pan T, Lee J, Goldberg S, King SA, Tang E, Hu Y, Chen L, Hoover A, Zhu L, Eng OS, Dekel B, Huang J, Wu X. Enabling tumor-specific drug delivery by targeting the Warburg effect of cancer. Cell Rep Med 2025; 6:101920. [PMID: 39809265 PMCID: PMC11866520 DOI: 10.1016/j.xcrm.2024.101920] [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: 08/31/2023] [Revised: 02/14/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025]
Abstract
Metabolic reprogramming of tumor cells is an emerging hallmark of cancer. Among all the changes in cancer metabolism, increased glucose uptake and the accumulation of lactate under normoxic conditions (the "Warburg effect") is a common feature of cancer cells. In this study, we develop a lactate-responsive drug delivery platform by targeting the Warburg effect. We design and test a gold/mesoporous silica Janus nanoparticle system as a gated drug carrier, in which the gold particles are functionalized with lactate oxidase and the silica particles are capped with α-cyclodextrin through surface arylboronate modification. In the presence of lactate, the lactate oxidase generates hydrogen peroxide, which induces the self-immolation reaction of arylboronate, leading to uncapping and drug release. Our results demonstrate greatly improved drug delivery specificity and therapeutic efficacy with this platform for the treatment of different cancers. Our findings present an effective approach for drug delivery by metabolic targeting of tumors.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27606, USA; Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Tony Pan
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Jimmy Lee
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA; Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Sanja Goldberg
- Pediatric Stem Cell Research Institute, Safra Children's Hospital, Sheba Medical Center, Tel Aviv, Israel
| | - Sarah Ann King
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Erting Tang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Yifei Hu
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Lifeng Chen
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA
| | - Alex Hoover
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Linyong Zhu
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science & Technology, Shanghai 200237, China
| | - Oliver S Eng
- Department of Surgery, University of California, Irvine, Orange, CA 92868, USA
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Safra Children's Hospital, Sheba Medical Center, Tel Aviv, Israel; Division of Pediatric Nephrology and Pediatric Stem Cell Research Institute, Safra Children's Hospital, Sheba Medical Center, Tel Hasomer, Sago Center for Regenerative Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jun Huang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA.
| | - Xiaoyang Wu
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
4
|
Man X, Li W, Zhu M, Li S, Xu G, Zhang Z, Liang H, Yang F. Rational Design of a Hetero-multinuclear Gadolinium(III)-Copper(II) Complex: Integrating Magnetic Resonance Imaging, Photoacoustic Imaging, Mild Photothermal Therapy, Chemotherapy and Immunotherapy of Cancer. J Med Chem 2024; 67:15606-15619. [PMID: 39143701 DOI: 10.1021/acs.jmedchem.4c01273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
For more accurate diagnosis and effective treatment of cancer, we proposed to develop a hetero-multinuclear metal complex based on the property of apoferritin (AFt) for targeting tumor theranostics by integrating dual-modality imaging diagnosis and multimodality therapy. To this end, we rational designed and synthesized a trinuclear Gd(III)-Cu(II) thiosemicarbazone complex (Gd-2Cu) and then constructed a Gd-2Cu@AFt nanoparticle (NP) delivery system. Gd-2Cu/Gd-2Cu@AFt NPs not only had significant T1-weighted magnetic resonance imaging and photoacoustic imaging of the tumor but also effectively inhibited tumor growth through a combination of mild photothermal therapy, chemotherapy, and immunotherapy. Gd-2Cu@AFt NPs optimized the behavior of imaging diagnosis and therapy of Gd-2Cu, improved its targeting ability, and reduced the side effects in vivo. Besides, we revealed and clarified the anticancer mechanism of Gd-2Cu: interrupting energy metabolism of the tumor cell, inducing apoptosis of the tumor cell, and activating a systemic immune response by inducing immunogenic cell death of cancer cells.
Collapse
Affiliation(s)
- Xueyu Man
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Wenjuan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Minghui Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), Collaborative Innovation Center for Guangxi Ethnic Medicine, School of Chemistry and Pharmaceutical Sciences, Guangxi Normal University, Guilin, Guangxi 541004, China
| |
Collapse
|
5
|
Zhang J, Zhou J, Tang L, Ma J, Wang Y, Yang H, Wang X, Fan W. Custom-Design of Multi-Stimuli-Responsive Degradable Silica Nanoparticles for Advanced Cancer-Specific Chemotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2400353. [PMID: 38651235 DOI: 10.1002/smll.202400353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/24/2024] [Indexed: 04/25/2024]
Abstract
Chemotherapy is crucial in oncology for combating malignant tumors but often encounters obatacles such as severe adverse effects, drug resistance, and biocompatibility issues. The advantages of degradable silica nanoparticles in tumor diagnosis and treatment lie in their ability to target drug delivery, minimizing toxicity to normal tissues while enhancing therapeutic efficacy. Moreover, their responsiveness to both endogenous and exogenous stimuli opens up new possibilities for integrating multiple treatment modalities. This review scrutinizes the burgeoning utility of degradable silica nanoparticles in combination with chemotherapy and other treatment modalities. Commencing the elucidation of degradable silica synthesis and degradation mechanisms, emphasis is placed on the responsiveness of these materials to endogenous (e.g., pH, redox reactions, hypoxia, and enzymes) and exogenous stimuli (e.g., light and high-intensity focused ultrasound). Moreover, this exploration delves into strategies harnessing degradable silica nanoparticles in chemotherapy alone, coupled with radiotherapy, photothermal therapy, photodynamic therapy, gas therapy, immunotherapy, starvation therapy, and chemodynamic therapy, elucidating multimodal synergies. Concluding with an assessment of advances, challenges, and constraints in oncology, despite hurdles, future investigations are anticipated to augment the role of degradable silica in cancer therapy. These insights can serve as a compass for devising more efficacious combined tumor treatment strategies.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing, 210023, P. R. China
| | - Jiani Zhou
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | | | - Jiayi Ma
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Ying Wang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Hui Yang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, 233030, P. R. China
| | - Xiaoxiao Wang
- Biochemical Engineering Research Center, School of Chemistry and Chemical Engineering, Anhui University of Technology, Ma'anshan, 243032, P. R. China
| | - Wenpei Fan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases, Center of Advanced Pharmaceuticals and Biomaterials, China Pharmaceutical University, Nanjing, 211198, P. R. China
| |
Collapse
|
6
|
Zheng C, Zhang W, Gong X, Xiong F, Jiang L, Zhou L, Zhang Y, Zhu HH, Wang H, Li Y, Zhang P. Chemical conjugation mitigates immunotoxicity of chemotherapy via reducing receptor-mediated drug leakage from lipid nanoparticles. SCIENCE ADVANCES 2024; 10:eadk9996. [PMID: 38838152 DOI: 10.1126/sciadv.adk9996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 05/02/2024] [Indexed: 06/07/2024]
Abstract
Immunotoxicity remains a major hindrance to chemotherapy in cancer therapy. Nanocarriers may alleviate the immunotoxicity, but the optimal design remains unclear. Here, we created two variants of maytansine (DM1)-loaded synthetic high-density lipoproteins (D-sHDL) with either physically entrapped (ED-sHDL) or chemically conjugated (CD-sHDL) DM1. We found that CD-sHDL showed less accumulation in the tumor draining lymph nodes (DLNs) and femur, resulting in a lower toxicity against myeloid cells than ED-sHDL via avoiding scavenger receptor class B type 1 (SR-B1)-mediated DM1 transportation into the granulocyte-monocyte progenitors and dendritic cells. Therefore, higher densities of lymphocytes in the tumors, DLNs, and blood were recorded in mice receiving CD-sHDL, leading to a better efficacy and immune memory of CD-sHDL against colon cancer. Furthermore, liposomes with conjugated DM1 (CD-Lipo) showed lower immunotoxicity than those with entrapped drug (ED-Lipo) through the same mechanism after apolipoprotein opsonization. Our findings highlight the critical role of drug loading patterns in dictating the biological fate and activity of nanomedicine.
Collapse
Affiliation(s)
- Chao Zheng
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Wen Zhang
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Xiang Gong
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Fengqin Xiong
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Linyang Jiang
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Lingli Zhou
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Yuan Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Helen He Zhu
- State Key Laboratory of Oncogenes and Related Genes, Renji-Med-X Stem Cell Research Center, Department of Urology, Ren Ji Hospital, School of Medicine and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Hao Wang
- China State Institute of Pharmaceutical Industry, Shanghai 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- Yantai Key Laboratory of Nanomedicine and Advanced Preparations, Yantai Institute of Materia Medica, Yantai 264000, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Pengcheng Zhang
- State Key Laboratory of Drug Research and Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Biomedical Engineering, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
7
|
Wu Y, Li Y, Hu Z, Li Y, Zhang S, Bao X, Zhou Y, Gao Y, Li Y, Zhang Z. Extracellular Matrix-Trapped Bioinspired Lipoprotein Prolongs Tumor Retention to Potentiate Antitumor Immunity. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2310982. [PMID: 38216153 DOI: 10.1002/adma.202310982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/28/2023] [Indexed: 01/14/2024]
Abstract
The immunomodulatory effects of many therapeutic agents are significantly challenged by their insufficient delivery efficiency and short retention time in tumors. Regarding the distinctively upregulated fibronectin (FN1) and tenascin C (TNC) in tumor stroma, herein a protease-activated FN1 and/or TNC binding peptide (FTF) is designed and an extracellular matrix (ECM)-trapped bioinspired lipoprotein (BL) (FTF-BL-CP) is proposed that can be preferentially captured by the TNC and/or FN1 for tumor retention, and then be responsively dissociated from the matrix to potentiate the antitumor immunity. The FTF-BL-CP treatment produces a 6.96-, 9.24-, 6.72-, 7.32-, and 6.73-fold increase of CD3+CD8+ T cells and their interferon-γ-, granzyme B-, perforin-, and Ki67-expressing subtypes versus the negative control, thereby profoundly eliciting the antitumor immunity. In orthotopic and lung metastatic breast cancer models, FTF-BL-CP produces notable therapeutic benefits of retarding tumor growth, extending survivals, and inhibiting lung metastasis. Therefore, this ECM-trapping strategy provides an encouraging possibility of prolonging tumor retention to potentiate the antitumor immunity for anticancer immunotherapy.
Collapse
Affiliation(s)
- Yao Wu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yongping Li
- Department of Breast Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, 201399, China
| | - Zixin Hu
- Artificial Intelligence Innovation and Incubation Institute, Fudan University, Shanghai, 200433, China
| | - Yuan Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Shixuan Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences & Human Phenome Institute, Fudan University, Shanghai, 200433, China
| | - Xinyue Bao
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yu Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yuan Gao
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| | - Yaping Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264005, China
| | - Zhiwen Zhang
- School of Pharmacy, Fudan University, Shanghai, 201203, China
| |
Collapse
|
8
|
Yang X, Li C, Yang H, Li T, Ling S, Zhang Y, Wu F, Liu X, Liu S, Fan C, Wang Q. Programmed Remodeling of the Tumor Milieu to Enhance NK Cell Immunotherapy Combined with Chemotherapy for Pancreatic Cancer. NANO LETTERS 2024; 24:3421-3431. [PMID: 38377170 DOI: 10.1021/acs.nanolett.4c00002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Natural killer (NK) cell-based adoptive immunotherapy has demonstrated encouraging therapeutic effects in clinical trials for hematological cancers. However, the effectiveness of treatment for solid tumors remains a challenge due to insufficient recruitment and infiltration of NK cells into tumor tissues. Herein, a programmed nanoremodeler (DAS@P/H/pp) is designed to remodel dense physical stromal barriers and for dysregulation of the chemokine of the tumor environment to enhance the recruitment and infiltration of NK cells in tumors. The DAS@P/H/pp is triggered by the acidic tumor environment, resulting in charge reversal and subsequent hyaluronidase (HAase) release. HAase effectively degrades the extracellular matrix, promoting the delivery of immunoregulatory molecules and chemotherapy drugs into deep tumor tissues. In mouse models of pancreatic cancer, this nanomediated strategy for the programmed remodeling of the tumor microenvironment significantly boosts the recruitment of NK92 cells and their tumor cell-killing capabilities under the supervision of multiplexed near-infrared-II fluorescence.
Collapse
Affiliation(s)
- Xiaohu Yang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Chunyan Li
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Hongchao Yang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Tuanwei Li
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Sisi Ling
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yejun Zhang
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Feng Wu
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Xiaoguo Liu
- School of Chemistry and Chemical Engineering, Frontiers Science Centre for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Shaoqin Liu
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Chunhai Fan
- School of Chemistry and Chemical Engineering, Frontiers Science Centre for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Qiangbin Wang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
- College of Materials Sciences and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
9
|
Zhou H, Yu CY, Wei H. Liposome-based nanomedicine for immune checkpoint blocking therapy and combinatory cancer therapy. Int J Pharm 2024; 652:123818. [PMID: 38253269 DOI: 10.1016/j.ijpharm.2024.123818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/06/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024]
Abstract
The discovery of immune checkpoint (IC) has led to a wave of leap forward in cancer immunotherapy that represents probably the most promising strategy for cancer therapy. However, the clinical use of immune checkpoint block (ICB) therapy is limited by response rates and side effects. A strategy that addresses the limitations of ICB therapies through combination therapies, using nanocarriers as mediators, has been mentioned in numerous research papers. Liposomes have been probably one of the most extensively used nanocarriers for clinical applications, with broad drug delivery and high safety. A timely review on this hot subject of research, i.e., the application of liposomes for ICB, is thus highly desirable for both fundamental and clinical translatable studies, but remains, to our knowledge, unexplored so far. For this purpose, this review is composed to address the dilemma of ICB therapy and the reasons for this dilemma. We later describe how other cancer treatments have broken this dilemma. Finally, we focus on the role of liposomes in various combinatory cancer therapy. This review is believed to serve as a guidance for the rational design and development of liposome for immunotherapy with enhanced therapeutic efficiency.
Collapse
Affiliation(s)
- Haoyuan Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China.
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China.
| |
Collapse
|
10
|
Ouyang C, Zhang W, Nie J, Yu L, Liu J, Ren L, Chen G. Nanoparticles with Active Targeting Ability and Acid Responsiveness for an Enhanced Antitumor Effect of Docetaxel. Biomacromolecules 2024; 25:213-221. [PMID: 38116982 DOI: 10.1021/acs.biomac.3c00922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Docetaxel (DOC) is commonly used in cancer treatment, especially for breast cancer. However, there are severe side effects in clinical application. In order to deliver docetaxel more effectively, a novel, active targeting acid-responsive polymer called cRGD-PAE-PEG-DSPE was developed. The polymer structure incorporated poly(ethylene glycol) (PEG) as the hydrophilic segment, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine (DSPE) as the hydrophobic segment, and poly(β-amino ester) (PAE) as the acid-responsive group, which was grafted onto the PEG. Furthermore, c(RGDyC) was grafted onto PAE to confer active targeting capability. Through self-assembly, docetaxel was encapsulated in RAED@DOC. Through in vitro experiments, it was confirmed that RAED@DOC had good serum stability and acid responsiveness, as well as enhanced uptake by MDA-MB-231 cells. Additionally, the antitumor efficiency in vivo and histopathological analysis showed that RAED@DOC exhibited higher antitumor activity and lower systemic toxicity in comparison to free docetaxel. These results suggested that RAED@DOC had considerable potential clinical use.
Collapse
Affiliation(s)
| | - Wei Zhang
- Nanjing Tech University, Nanjing 211816, China
| | - Junfang Nie
- Nanjing Tech University, Nanjing 211816, China
| | - Luting Yu
- Nanjing Tech University, Nanjing 211816, China
| | - Jia Liu
- Nanjing Tech University, Nanjing 211816, China
| | - Lili Ren
- Nanjing Tech University, Nanjing 211816, China
| | | |
Collapse
|
11
|
Jackman RP, Darst O, Gaillard B, Tran JQ, Tomayko MM, Muench MO. Enhanced alloresponse to platelet transfusion due to immune dysregulation following ablative chemotherapy in mice. Front Immunol 2023; 14:1281123. [PMID: 38090570 PMCID: PMC10711281 DOI: 10.3389/fimmu.2023.1281123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/07/2023] [Indexed: 12/18/2023] Open
Abstract
Introduction Alloimmunization is common following platelet transfusion and can result in negative outcomes for recipients such as refractoriness to subsequent transfusions and rejection of transplants. Healthy people do not receive blood transfusions, and the diseases and therapies that result in a need to transfuse have significant impacts on the immunological environment to which these alloantigens are introduced. Ablative chemotherapies are common among platelet recipients and have potent immunological effects. In this study, we modeled the impact of chemotherapy on the alloresponse to platelet transfusion. As chemotherapies are generally regarded as immunosuppressive, we hypothesized that that they would result in a diminished alloresponse. Methods Mice were given a combination chemotherapeutic treatment of cytarabine and doxorubicin followed by transfusion of allogeneic platelets, and compared to controls given no treatment, chemotherapy alone, or transfusion alone. Alloantibody responses were measured 2 weeks after transfusion, and cellular responses and growth factors were monitored over time. Results Contrary to our hypothesis, we found that chemotherapy led to increased alloantibody responses to allogeneic platelet transfusion. This enhanced response was antigen-specific and was associated with increased CD4+ and CD8+ T cell responses. Chemotherapy led to rapid lymphocyte depletion followed by reconstitution, non-specific activation of transitional B cells with the highest levels of activation in the least mature subsets, and increased serum levels of B cell activating factor (BAFF). Conclusion These data suggest that ablative chemotherapy can increase the risk of alloimmunization and, if confirmed clinically, that additional measures to protect these patient populations may be warranted.
Collapse
Affiliation(s)
- Rachael P. Jackman
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Orsolya Darst
- Vitalant Research Institute, San Francisco, CA, United States
| | - Betty Gaillard
- Vitalant Research Institute, San Francisco, CA, United States
| | - Johnson Q. Tran
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| | - Mary M. Tomayko
- Department of Dermatology, Yale School of Medicine, New Haven, CT, United States
- Department of Pathology, Yale School of Medicine, New Haven, CT, United States
| | - Marcus O. Muench
- Vitalant Research Institute, San Francisco, CA, United States
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA, United States
| |
Collapse
|
12
|
Zhu M, Man X, Tongfu Y, Li W, Li S, Xu G, Zhang Z, Liang H, Yang F. Developing a Hetero-Trinuclear Erbium(III)-Copper(II) Complex Based on Apoferritin: Targeted Photoacoustic Imaging and Multimodality Therapy of Tumor. J Med Chem 2023; 66:15424-15436. [PMID: 37956097 DOI: 10.1021/acs.jmedchem.3c01583] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
For the integration of targeted diagnosis and treatment of tumor, we innovatively designed and synthesized a single-molecule hetero-multinuclear Er(III)-Cu(II) complex (ErCu2) and then constructed an ErCu2@apoferritin (AFt) nanoparticle (NP) delivery system. ErCu2 and ErCu2@AFt NPs not only provided an evident photoacoustic imaging (PAI) signal of the tumor but also effectively inhibited tumor growth by integrating photothermal therapy, chemotherapy, and immunotherapy. ErCu2@AFt NPs improved the targeting ability and decreased the systemic toxicity of ErCu2 in vivo. Furthermore, we confirmed that ErCu2 and ErCu2@AFt NPs inhibited tumor growth by inducing apoptosis and autophagy of tumor cells and activating the immune system. The study not only provides a novel strategy to develop therapeutic metal agents but also reveals their potential for targeted accurate diagnosis and multimodality therapy of cancer.
Collapse
Affiliation(s)
- Minghui Zhu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Xueyu Man
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
- School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou 563006, China
| | - Yang Tongfu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Wenjuan Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Shanhe Li
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Gang Xu
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Zhenlei Zhang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Hong Liang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| | - Feng Yang
- State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources/Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources (Ministry of Education of China), School of Chemistry and Pharmaceutical Sciences, Collaborative Innovation Center for Guangxi Ethnic Medicine, Guangxi Normal University, Guilin, Guangxi 541004, China
| |
Collapse
|
13
|
Li Y, Liang X, Shen C, Deng K, Zeng Z, Guo B, Xu X. Bio-Responsive Macromolecular Drug and Small-Molecular Drug Conjugates: Nanoparticulate Prodrugs for Tumor Microenvironment Heterogeneity Management and Therapeutic Response Enhancement. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301656. [PMID: 37144435 DOI: 10.1002/smll.202301656] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/07/2023] [Indexed: 05/06/2023]
Abstract
How to break through the poor response of current drug therapy, which often resulted from tumor microenvironment heterogeneity (TMH), remains an enormous challenge in the treatment of critical diseases. In this work, a practical solution on bio-responsive dual-drug conjugates for overcoming TMH and improving antitumor treatment, which integrates the advantages of macromolecular drugs and small-molecular drugs, is proposed. Nanoparticulate prodrugs based on small-molecular drug and macromolecular drug conjugates are designed as a robust weapon for programmable multidrug delivery at tumor-specific sites: the tumor microenvironment acid condition triggers delivery of macromolecular aptamer drugs (AX102) to manage TMH (including tumor stroma matrix, interstitial fluid pressure, vasculature network, blood perfusion, and oxygen distribution), and intracellular lysosomal acid condition activates rapid release of small-molecular drugs (doxorubicin and dactolisib) to enhance curative effects. As compared with doxorubicin chemotherapy, the tumor growth inhibition rate is enhanced by 47.94% after multiple tumor heterogeneity management. This work verifies that the nanoparticulate prodrugs facilitate TMH management and therapeutic response enhancements, as well as elucidates synergetic mechanisms for drug resistance reversal and metastasis inhibition. It is hoped that the nanoparticulate prodrugs will be an excellent demonstration of the co-delivery of small-molecular drugs and macromolecular drugs.
Collapse
Affiliation(s)
- Yachao Li
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Changsha, Hunan, 410082, China
| | - Xiaoyu Liang
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Cheng Shen
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Kefurong Deng
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Zenan Zeng
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Beiling Guo
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
| | - Xianghui Xu
- Department of Pharmacy, College of Biology, Hunan University, Changsha, Hunan, 410082, China
- State Key Laboratory of Chemo/Biosensing and Chemometrics, Changsha, Hunan, 410082, China
| |
Collapse
|
14
|
Liang D, Kuang G, Chen X, Lu J, Shang L, Sun W. Near-infrared light-responsive Nitric oxide microcarrier for multimodal tumor therapy. SMART MEDICINE 2023; 2:e20230016. [PMID: 39188343 PMCID: PMC11236066 DOI: 10.1002/smmd.20230016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/08/2023] [Indexed: 08/28/2024]
Abstract
Nitric oxide (NO) has shown great potential in tumor therapy, and the development of a platform for precise and controllable NO release still needs to be explored. Herein, a microfluidic electrospray strategy is proposed for the fabrication of hydrogel microspheres encapsulating NO donors (S-nitrosoglutathione, GSNO) together with black phosphorus (BP) and chemotherapeutic doxorubicin (DOX) as microcarriers for tumor therapy. Based on the excellent photothermal property of BP and thermal sensitivity of GSNO, the microcarriers exhibit a near-infrared light (NIR)-responsive NO release behavior. Besides, the photothermal performance of the microcarriers accelerates the release of DOX. All these contribute to the excellent tumor-killing effect of the microcarriers by combining multiple therapeutic strategies including NO therapy, photothermal therapy, and chemotherapy. Moreover, it was demonstrated that the NIR-responsive NO delivery microcarriers could significantly inhibit tumor growth without apparent side effects in vivo. Therefore, it is believed that the novel NIR-responsive NO microcarriers are promising candidates in clinical tumor therapy applications.
Collapse
Affiliation(s)
- Danna Liang
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Gaizhen Kuang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
| | - Xiang Chen
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Jianhua Lu
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| | - Luoran Shang
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health)Wenzhou InstituteUniversity of Chinese Academy of SciencesWenzhouZhejiangChina
- Zhongshan‐Xuhui Hospital and the Shanghai Key Laboratory of Medical Epigenetics, the International Co‐laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology)Institutes of Biomedical SciencesFudan UniversityShanghaiChina
| | - Weijian Sun
- Department of Gastrointestinal SurgeryThe Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical UniversityWenzhouZhejiangChina
| |
Collapse
|
15
|
Li Z, Li Z, Wang J. Visualization of Phototherapy Evolution by Optical Imaging. Molecules 2023; 28:molecules28103992. [PMID: 37241733 DOI: 10.3390/molecules28103992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Phototherapy, including photodynamic therapy (PDT) and photothermal therapy (PTT), is a non-invasive and effective approach used for cancer treatment, in which phototherapeutic agents are irradiated with an appropriate light source to produce cytotoxic reactive oxygen species (ROS) or heat to ablate cancer cells. Unfortunately, traditional phototherapy lacks a facile imaging method to monitor the therapeutic process and efficiency in real time, usually leading to severe side effects due to high levels of ROS and hyperthermia. To realize precise cancer treatment methods, it is highly desired to develop phototherapeutic agents possessing an imaging ability to evaluate the therapeutic process and efficacy in real time during cancer phototherapy. Recently, a series of self-reporting phototherapeutic agents were reported to monitor PDT and PTT processes by combining optical imaging technologies with phototherapy. Due to the real-time feedback provided by optical imaging technology, therapeutic responses or dynamic changes in the tumor microenvironment could be evaluated in a timely manner, thereby achieving personalized precision treatment and minimizing toxic side effects. In this review, we focus on the advances in the development of self-reporting phototherapeutic agents for a cancer phototherapy evaluation based on optical imaging technology to realize precision cancer treatments. Additionally, we propose the current challenges and future directions of self-reporting agents for precision medicine.
Collapse
Affiliation(s)
- Zhiheng Li
- College of Materials and Chemical Engineering, Zhengzhou University of Light Industry, Zhengzhou 450002, China
| | - Zheng Li
- Wuhan Academy of Agricultural Sciences, Wuhan 430072, China
| | - Jie Wang
- The Key Lab of Health Chemistry & Molecular Diagnosis of Suzhou, College of Chemistry, Chemical Engineering & Materials Science, Soochow University, Suzhou 215123, China
| |
Collapse
|
16
|
Liu J, Zhu H, Lin L, Zhao W, Zhu X, Pang DW, Liu AA. Redox Imbalance Triggered Intratumoral Cascade Reaction for Tumor "turn on" Imaging and Synergistic Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2206272. [PMID: 36683231 DOI: 10.1002/smll.202206272] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/19/2022] [Indexed: 06/17/2023]
Abstract
The redox homeostasis in tumors enhances their antioxidant defense ability, limiting reactive oxygen species mediated tumor therapy efficacy. The development of strategies for specific and continuous disruption of the redox homeostasis in tumor cells facilitates the improvement of the cancer therapeutic effect by promoting the apoptosis of tumor cells. Herein, a responsively biodegradable targeting multifunctional integrated nanosphere (HDMn-QDs/PEG-FA) is designed to enhance the anti-tumor efficacy by triggering intratumoral cascade reactions to effectively disrupt intracellular redox homeostasis. Once HDMn-QDs/PEG-FA enters tumor cells, manganese dioxide (MnO2 ) shell on the surface of nanosphere consumes glutathione (GSH) to produce Mn2+ , enabling enhanced chemodynamic therapy (CDT) via a Fenton-like reaction and T1 -weighted magnetic resonance imaging. Meanwhile, the degradation of MnO2 can also cause the fluorescence recovery of quantum dots conjugated on the surface of the shell, realizing "turn-on" fluorescence imaging. In addition, the doxorubicin is released because of the cleavage of the embedded SS bond in the hybrid core framework by GSH. A superior synergistic therapeutic efficiency combined CDT and chemotherapy is shown by HDMn-QDs/PEG-FA in vivo. The tumor-inhibition rate reaches to 94.8% and does not cause normal tissue damage due to the good targeting and tumor microenvironment-specific response.
Collapse
Affiliation(s)
- Juanzu Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Haihe Laboratory of Sustainable Chemical Transformations, Nankai University, Tianjin, 300071, P. R. China
| | - Han Zhu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Haihe Laboratory of Sustainable Chemical Transformations, Nankai University, Tianjin, 300071, P. R. China
| | - Leping Lin
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Haihe Laboratory of Sustainable Chemical Transformations, Nankai University, Tianjin, 300071, P. R. China
- Cannano Jiayuan (Guangzhou) Science & Technology Co., Ltd, Guangzhou, 510700, P. R. China
| | - Wei Zhao
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Haihe Laboratory of Sustainable Chemical Transformations, Nankai University, Tianjin, 300071, P. R. China
| | - Xiaobo Zhu
- Cannano Jiayuan (Guangzhou) Science & Technology Co., Ltd, Guangzhou, 510700, P. R. China
| | - Dai-Wen Pang
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Haihe Laboratory of Sustainable Chemical Transformations, Nankai University, Tianjin, 300071, P. R. China
| | - An-An Liu
- State Key Laboratory of Medicinal Chemical Biology, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Frontiers Science Center for New Organic Matter, Research Center for Analytical Sciences, College of Chemistry, Frontiers Science Center for Cell Responses, Haihe Laboratory of Sustainable Chemical Transformations, Nankai University, Tianjin, 300071, P. R. China
| |
Collapse
|
17
|
Zhang Y, Chen J, Shi L, Ma F. Polymeric nanoparticle-based nanovaccines for cancer immunotherapy. MATERIALS HORIZONS 2023; 10:361-392. [PMID: 36541078 DOI: 10.1039/d2mh01358d] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Therapeutic cancer vaccines, which are designed to amplify tumor-specific T cell responses, have been envisioned as one of the most powerful tools for effective cancer immunotherapy. However, increasing the potency, quality and durability of the vaccine response remains a big challenge. In recent years, materials-based delivery systems focusing on the co-delivery of antigens and adjuvants to enhance cancer vaccination therapy have attracted increasing interest. Among various materials, polymeric nanoparticles (NPs) with different physicochemical properties which can incorporate multiple immunological cues are of great interest. In this review, the recent progress in the design and construction of both ex vivo subunit and in situ cancer vaccines using polymeric NPs is summarized. Especially, we will focus on how these NPs improve the adjuvanticity of vaccines. The design principles of polymeric NPs for ex vivo subunit cancer vaccines and in situ cancer vaccination are also discussed. Finally, we want to briefly discuss molecular chaperones in cancer immunity and the applications of our unique self-assembly mixed shell polymeric micelle-based nanochaperones for cancer vaccines.
Collapse
Affiliation(s)
- Yongxin Zhang
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
| | - Jiajing Chen
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
| | - Linqi Shi
- Key Laboratory of Functional Polymer Materials of Ministry of Education, State Key Laboratory of Medicinal Chemical Biology, Institute of Polymer Chemistry and College of Chemistry, Nankai University, Tianjin, 300071, P. R. China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin, 300192, P. R. China
| | - Feihe Ma
- State Key Laboratory of Separation Membranes and Membrane Processes, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, P. R. China.
| |
Collapse
|
18
|
The combination of in situ photodynamic promotion and ion-interference to improve the efficacy of cancer therapy. J Colloid Interface Sci 2023; 629:522-533. [PMID: 36088697 DOI: 10.1016/j.jcis.2022.08.125] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/22/2022] [Accepted: 08/18/2022] [Indexed: 11/22/2022]
Abstract
Photodynamic therapy (PDT) is proved to be a promising modality for clinical cancer treatment. However, it also suffers from a key obstacle in association with its oxygen-dependent nature which greatly limits its effective application against hypoxic tumors. Herein, on the basis of the unique property of calcium peroxide (CaO2), we propose an O2-self-supply strategy for the promotion of PDT by combining the in situ O2-generation characteristic of calcium peroxide with the photosensitive nature of porphyrin. A shell of ZIF-8 was synthesized surround the CaO2 core to prevent the CaO2 from premature decomposition and increased the loading of THPP efficiently. Depending on the in situ self-supply of O2, the photosensitizer was able to exhibit an enhanced PDT effect that significantly inhibit the growth of tumor. Moreover, the enrichment of free calcium ions derived from the decomposition of CaO2 under acidic tumor microenvironment also shows the unique ion-interference effect and contributes to the obvious inhibition against tumor growth. This work presents a synergistic strategy for the construction of a photodynamic promotion/ion-interference combined nano-platform which can also serve as an inspiration for the future design of effective nanocomposites in tumor treatment.
Collapse
|
19
|
Xu XX, Chen SY, Yi NB, Li X, Chen SL, Lei Z, Cheng DB, Sun T. Research progress on tumor hypoxia-associative nanomedicine. J Control Release 2022; 350:829-840. [PMID: 36100192 DOI: 10.1016/j.jconrel.2022.09.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 12/17/2022]
Abstract
Hypoxia at the solid tumor site is generally related to the unrestricted proliferation and metabolism of cancerous cells, which can cause tumor metastasis and aggravate tumor progression. Besides, hypoxia plays a substantial role in tumor treatment, and it is one of the main reasons that malignant tumors are difficult to cure and have a poor prognosis. On account of the tumor specific hypoxic environment, many hypoxia-associative nanomedicine have been proposed for tumor treatment. Considering the enhanced targeting effect, designing hypoxia-associative nanomedicine can not only minimize the adverse effects of drugs on normal tissues, but also achieve targeted therapy at the lesion site. Mostly, there can be three strategies for the treatment of hypoxic tumor, including improvement of hypoxic environment, hypoxia responsive drug release and hypoxia activated prodrug. The review describes the design principle and applications of tumor hypoxia-associative nanomedicine in recent years, and also explores its development trends in solid tumor treatment. Moreover, this review presents the current limitations of tumor hypoxia-associative nanomedicine in chemotherapy, radiotherapy, photodynamic therapy, sonodynamic therapy and immunotherapy, which may provide a reference for clinic translation of tumor hypoxia-associative nanomedicine.
Collapse
Affiliation(s)
- Xiao-Xue Xu
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Si-Yi Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Ning-Bo Yi
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Xin Li
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Si-Lin Chen
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China
| | - Zhixin Lei
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| | - Dong-Bing Cheng
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| | - Taolei Sun
- School of Chemistry, Chemical Engineering & Life Science, Wuhan University of Technology, No. 122 Luoshi Road, Wuhan 430070, PR China.
| |
Collapse
|
20
|
Qian J, Xu Z, Meng C, Liu Y, Wu H, Wang Y, Yang J, Zheng H, Ran F, Liu GQ, Ling Y. Redox-Activatable Theranostic Co-Prodrug for Precise Tumor Diagnosis and Selective Combination Chemotherapy. J Med Chem 2022; 65:10393-10407. [PMID: 35877176 DOI: 10.1021/acs.jmedchem.2c00130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
A novel theranostic co-prodrug SCB has been designed by combining a co-prodrug from CDDO-Me and SAHA with a biotin-coupled near-infrared (NIR) probe hemicyanine via redox-responsive linker thiolactate to enhance the tumor theranostic efficacy and reduce the toxic side effects using both active and passive targeting strategies. SCB displayed reactive oxygen species (ROS)- and glutathione (GSH)-dependent release of NIR fluorescence and two parent drugs. Furthermore, the administration of SCB caused selective illumination of the tumor tissues for >24 h, thereby guiding precise removal of a tumor from intraoperative mice. Importantly, SCB exhibited highly efficient tumor inhibition, exerted selective combination therapy through prodrug mode, and minimized the adverse effects. Finally, SCB induced mitochondrial depolarization, DNA damage, and cell apoptosis through ROS generation and downregulation of HDAC6 protein, as verified by H2AX, Bax, cleaved-PARP, and Mcl-1 proteins. Thus, we suggest that SCB can provide a new platform for both precise diagnosis-guided tumor removal and selective combination therapy with high safety.
Collapse
Affiliation(s)
- Jianqiang Qian
- School of Pharmacy, Nantong University, Nantong 226001, P. R. China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, P. R. China
| | - Zhongyuan Xu
- School of Pharmacy, Nantong University, Nantong 226001, P. R. China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, P. R. China
| | - Chi Meng
- School of Pharmacy, Nantong University, Nantong 226001, P. R. China
| | - Yun Liu
- School of Pharmacy, Nantong University, Nantong 226001, P. R. China
| | - Hongmei Wu
- School of Pharmacy, Nantong University, Nantong 226001, P. R. China
| | - Yunyun Wang
- School of Pharmacy, Nantong University, Nantong 226001, P. R. China
| | - Jinxian Yang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, P. R. China
| | - Hongwei Zheng
- School of Pharmacy, Nantong University, Nantong 226001, P. R. China
| | - Fansheng Ran
- School of Pharmacy, Nantong University, Nantong 226001, P. R. China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, P. R. China
| | - Gong-Qing Liu
- School of Pharmacy, Nantong University, Nantong 226001, P. R. China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, P. R. China
| | - Yong Ling
- School of Pharmacy, Nantong University, Nantong 226001, P. R. China.,Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Nantong University, Nantong 226001, P. R. China
| |
Collapse
|
21
|
Effects of Surgery Combined with Different Chemotherapy on Matrix Metalloproteinase-9 and Tissue Inhibitors of Metalloproteinase-1 in Children with Neuroblastoma. JOURNAL OF ONCOLOGY 2022; 2022:8319221. [PMID: 35847358 PMCID: PMC9277205 DOI: 10.1155/2022/8319221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/14/2022] [Accepted: 05/18/2022] [Indexed: 12/03/2022]
Abstract
Background Neuroblastoma (NB) is a common extracranial malignancy in children and accounts for 15% of all cancer-related deaths in children, with the 5-year survival of patients in an advanced stage being lower than 40%. Preoperative adjuvant chemotherapy has been reported to facilitate surgical resection and improve the 2-year survival of patients. Objective To analyze the efficacy of surgery plus different chemotherapy on children with NB and to investigate the correlation of matrix metalloproteinase-9 (MMP-9) and tissue inhibitors of metalloproteinase-1 (TIMP-1) with chemotherapy efficacy. Methods From April 2005 to May 2017, a total of 92 cases of NB treated in our hospital were assessed for eligibility and recruited. They were assigned at a ratio of 1: 1 to receive either CAV (cyclophosphamide + vincristine + adriamycin) (group A) and EP (etoposide + cisplatin) alternately or TOPO (topotecan) + CTX (cytoxan) + CiE (etoposide + cisplatin) + CPV (cyclophosphamide + pirarubicin + vincristine) (group B). The outcome measures include chemotherapy efficacy, surgical resection rates, complications, 2-year recurrence, and 2-year survival. The levels of NK cells, CD4+/CD8+ cells, MMP-9, TIMP-1, and urine catecholamine (VMA) in peripheral blood of patients before and after initial chemotherapy were determined to analyze the correlation of MMP-9, TIMP-1, and VMA with the efficacy of chemotherapy. Results The two groups had similar efficacy (84.00% vs. 95.24%) and surgical resection rates (60.00% vs. 61.90%) after the initial chemotherapy (P > 0.05). Surgery for all eligible patients was successful after second chemotherapy. All eligible patients showed myelosuppression after chemotherapy, including 48 cases with stages I-II (52.17%) and 44 cases with stages III-IV (47.83%). The ratio of CD4+/CD8+ cells, MMP-9, TIMP-1, and VMA expression levels in peripheral blood of patients decreased (P < 0.05) after chemotherapy, and the ratio of CD4+/CD8+ cells was further reduced after surgery (P < 0.05), while natural killer (NK) cells levels increased (P < 0.05). However, intergroup differences were absent in the incidence of myelosuppression, CD4+/CD8+ cell ratio, NK cells, MMP-9, TIMP-1, and VMA expression levels (P > 0.05). MMP-9 and TIMP-1 were positively correlated with VMA (P < 0.05), and the expression levels of MMP-9 and TIMP-1 and VMA after chemotherapy were negatively correlated with chemotherapy efficiency (P < 0.05). Patients with high expressions of MMP-9, TIMP-1, and VMA were associated with lower 2-year survival versus those with low expressions (P < 0.05). Conclusion Surgery plus chemotherapy for children with NB yields a promising clinical efficacy and a favorable surgical resection outcome. MMP-9 and TIMP-1 may be the potential biological indicators for chemotherapy efficiency and have a reference value for following surgical treatment of patients.
Collapse
|
22
|
Zhao P, Liu D, Hu H, Qiu Z, Liang Y, Chen Z. Anticancer activity of four trinuclear cobalt complexes bearing bis(salicylidene)-1,3-propanediamine derivatives. J Inorg Biochem 2022; 233:111860. [DOI: 10.1016/j.jinorgbio.2022.111860] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/17/2022] [Accepted: 05/08/2022] [Indexed: 10/18/2022]
|
23
|
Jing X, Hu H, Sun Y, Yu B, Cong H, Shen Y. The Intracellular and Extracellular Microenvironment of Tumor Site: The Trigger of Stimuli-Responsive Drug Delivery Systems. SMALL METHODS 2022; 6:e2101437. [PMID: 35048560 DOI: 10.1002/smtd.202101437] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/29/2021] [Indexed: 06/14/2023]
Abstract
The tumor microenvironment (TME), including intracellular and extracellular microenvironment, contains many biochemical indicators (such as acidity/alkalinity, oxygen content, and enzymatic activity) that are different from the normal physiological environment. These abnormal biochemical indicators can accelerate the heterogeneity of tumors, but on the other hand, they also provide opportunities for the design of intelligent drug delivery systems (DDSs). The TME-responsive DDSs have shown great potential in reducing the side effects of chemotherapy and improving the curative effect of tumors. In this review, the abnormal biochemical indicators of TME are introduced in detail from both the extracellular and intracellular aspects. In view of the various physiological barriers encountered during drug delivery, the strategy of constructing TME-responsive DDSs is discussed. By summarizing the typical research progress, the authors prospect the development of TME-responsive DDS in the future.
Collapse
Affiliation(s)
- Xiaodong Jing
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Hao Hu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Yanzhen Sun
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
| | - Bing Yu
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Hailin Cong
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- State Key Laboratory of Bio-Fibers and Eco-Textiles, Qingdao University, Qingdao, 266071, China
| | - Youqing Shen
- Institute of Biomedical Materials and Engineering, College of Materials Science and Engineering, College of Chemistry and Chemical Engineering, Qingdao University, Qingdao, 266071, China
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, and Department of Chemical and Biological Engineering, Zhejiang University, Hangzhou, Zhejiang, 310027, China
| |
Collapse
|
24
|
Ma Z, Foda MF, Zhao Y, Han H. Multifunctional Nanosystems with Enhanced Cellular Uptake for Tumor Therapy. Adv Healthc Mater 2022; 11:e2101703. [PMID: 34626528 DOI: 10.1002/adhm.202101703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 10/01/2021] [Indexed: 11/10/2022]
Abstract
Rapid development of nanotechnology provides promising strategies in biomedicine, especially in tumor therapy. In particular, the cellular uptake of nanosystems is not only a basic premise to realize various biomedical applications, but also a fatal factor for determining the final therapeutic effect. Thus, a systematic and comprehensive summary is necessary to overview the recent research progress on the improvement of nanosystem cellular uptake for cancer treatment. According to the process of nanosystems entering the body, they can be classified into three categories. The first segment is to enhance the accumulation and permeation of nanosystems to tumor cells through extracellular microenvironment stimulation. The second segment is to improve cellular internalization from extracellular to intracellular via active targeting. The third segment is to enhance the intracellular retention of therapeutics by subcellular localization. The major factors in the delivery can be utilized to develop multifunctional nanosystems for strengthening the tumor therapy. Ultimately, the key challenges and prospective in the emerging research frontier are thoroughly outlined. This review is expected to provide inspiring ideas, promising strategies and potential pathways for developing advanced anticancer nanosystems in clinical practice.
Collapse
Affiliation(s)
- Zhaoyu Ma
- State Key Laboratory of Agricultural Microbiology College of Life Science and Technology Huazhong Agricultural University Wuhan Hubei 430070 P. R. China
- Division of Chemistry and Biological Chemistry School of Physical and Mathematical Sciences Nanyang Technological University 21 Nanyang Link Singapore 637371 Singapore
| | - Mohamed F. Foda
- State Key Laboratory of Agricultural Microbiology College of Life Science and Technology Huazhong Agricultural University Wuhan Hubei 430070 P. R. China
- Department of Biochemistry Faculty of Agriculture Benha University Moshtohor Toukh 13736 Egypt
| | - Yanli Zhao
- State Key Laboratory of Agricultural Microbiology College of Life Science and Technology Huazhong Agricultural University Wuhan Hubei 430070 P. R. China
- Division of Chemistry and Biological Chemistry School of Physical and Mathematical Sciences Nanyang Technological University 21 Nanyang Link Singapore 637371 Singapore
| | - Heyou Han
- State Key Laboratory of Agricultural Microbiology College of Life Science and Technology Huazhong Agricultural University Wuhan Hubei 430070 P. R. China
| |
Collapse
|
25
|
Ren L, Nie J, Wei J, Li Y, Yin J, Yang X, Chen G. RGD-targeted redox responsive nano micelle: co-loading docetaxel and indocyanine green to treat the tumor. Drug Deliv 2021; 28:2024-2032. [PMID: 34569890 PMCID: PMC8477929 DOI: 10.1080/10717544.2021.1977425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Cancer, also known as a malignant tumor, has developed into a type of disease with the highest fatality rate, seriously threatening the lives and health of people. Chemotherapy is one of the most important methods for the treatment of cancer. However, chemotherapy drugs have some problems, such as low solubility and lack of targeting, which severely limit their clinical applications. To solve these problems, we designed a block copolymer that has a disulfide bond response. The polymer uses RGD peptide (arginine-glycine-aspartic acid) as the active targeting group, PEG (polyethylene glycol) as the hydrophilic end, and PCL (polycaprolactone) as the hydrophobic end. Then we utilized the amphiphilic polymer as a carrier to simultaneously deliver DOC (docetaxel) and ICG (indocyanine green), to realize the combined application of chemotherapy and photothermal therapy. The antitumor efficacy in vivo and histology analysis showed that the DOC/ICG-loaded micelle exhibited higher antitumor activity. The drug delivery system improved the solubility of DOC and the stability of ICG, realized NIR-guided photothermal therapy, and achieved an ideal therapeutic effect.
Collapse
Affiliation(s)
- Lili Ren
- School of Pharmacy, Nanjing Tech University, Nanjing, China.,Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA
| | - Junfang Nie
- School of Pharmacy, Nanjing Tech University, Nanjing, China
| | - Jie Wei
- School of Pharmacy, Nanjing Tech University, Nanjing, China
| | - Yaning Li
- School of Pharmacy, Nanjing Tech University, Nanjing, China
| | - Jun Yin
- School of Pharmacy, Nanjing Tech University, Nanjing, China
| | - Xiaolong Yang
- School of Pharmacy, Nanjing Tech University, Nanjing, China
| | - Guoguang Chen
- School of Pharmacy, Nanjing Tech University, Nanjing, China
| |
Collapse
|
26
|
Zheng J, Wang Q, Shi L, Peng P, Shi L, Li T. Logic-Gated Proximity Aptasensing for Cell-Surface Real-Time Monitoring of Apoptosis. Angew Chem Int Ed Engl 2021; 60:20858-20864. [PMID: 34309152 DOI: 10.1002/anie.202106651] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Indexed: 12/15/2022]
Abstract
In nature, intact apoptotic cells release ATP as a signaling molecule to trigger prompt phagocytic clearance, even at the earliest stage of apoptosis. Inspired by this, here we introduce a straightforward strategy for real-time monitoring ATP exocytosis and drug-stimulated apoptosis in the cancer cell surroundings. Triplex-boosted G-quadruplexes (tb-G4s) responding to cell environmental factors (H+ and K+ ) are engineered to construct a DNA logic-gated nanoplatform for proximity ATP aptasensing on the cell surface. It enables the real-time monitoring of cell apoptosis by capturing released endogenous ATP during chemotherapy drug stimulation, providing a sensitive approach for dynamically evaluating drug-induced apoptosis and therapeutic efficacy.
Collapse
Affiliation(s)
- Jiao Zheng
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, China
| | - Qiwei Wang
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, China
| | - Lin Shi
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, China
| | - Pai Peng
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, China
| | - Lili Shi
- Department of Chemistry, Anhui University, 111 Jiulong Road, Hefei, Anhui, 230601, China
| | - Tao Li
- Department of Chemistry, University of Science and Technology of China, 96 Jinzhai Road, Hefei, Anhui, 230026, China
| |
Collapse
|
27
|
Zheng J, Wang Q, Shi L, Peng P, Shi L, Li T. Logic‐Gated Proximity Aptasensing for Cell‐Surface Real‐Time Monitoring of Apoptosis. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202106651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Jiao Zheng
- Department of Chemistry University of Science and Technology of China 96 Jinzhai Road Hefei Anhui 230026 China
| | - Qiwei Wang
- Department of Chemistry University of Science and Technology of China 96 Jinzhai Road Hefei Anhui 230026 China
| | - Lin Shi
- Department of Chemistry University of Science and Technology of China 96 Jinzhai Road Hefei Anhui 230026 China
| | - Pai Peng
- Department of Chemistry University of Science and Technology of China 96 Jinzhai Road Hefei Anhui 230026 China
| | - Lili Shi
- Department of Chemistry Anhui University 111 Jiulong Road Hefei Anhui 230601 China
| | - Tao Li
- Department of Chemistry University of Science and Technology of China 96 Jinzhai Road Hefei Anhui 230026 China
| |
Collapse
|
28
|
Huang X, Li Y, Li D, Zhou X, Qiao H, Yang L, Ji Y, Zhang X, Huang D, Chen W. Black phosphorus assisted polyionic micelles with efficient PTX loading for remotely controlled release and synergistic treatment of drug-resistant tumors. Biomater Sci 2021; 9:6108-6115. [PMID: 34369491 DOI: 10.1039/d1bm01033f] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nanomedicines have been widely used in the effective delivery of chemotherapeutic drugs due to their advantages such as increasing the half-life of drugs, selectively targeting tumor tissues, and thus reducing systemic toxicity. However, the low drug entrapment rate and the difficulty of real-controlled release at tumor sites hinder their further clinical translations. Here we have developed biodegradable polyionic micelles (PD-M) to facilitate black phosphorus (BP) encapsulation (PD-M@BP) for improved drug loading. With the introduction of BP, PTX-loaded PD-M@BP (PD-M@BP/PTX) with sizes of 124-162 nm exhibited superior encapsulation efficiency over 94% and excellent colloidal stability. Meanwhile, PD-M well protected BP from fast degradation to show the good photothermal performance under near-infrared (NIR) irradiation, thus achieving the remotely controlled fast PTX release due to micelle core melting and dissociation, accompanied by the synergistic photothermal tumor therapy. The in vivo results demonstrated that the PD-M@BP/PTX nanosystem not only realized significant inhibition of multi-drug resistant (MDR) cervical tumors (HeLa/PTXR tumor) by remote NIR-regulation, but also reduced the potential damage of chemotherapeutic drugs to the whole body, rendering these hybrid nanosystems as great tools to treat MDR tumors synergistically.
Collapse
Affiliation(s)
- Xin Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Yanfei Li
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Dengyu Li
- Key Laboratory of Pollution Ecology and Environmental Engineering, Shenyang Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, China.
| | - Xiang Zhou
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Haishi Qiao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Lifen Yang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Yicheng Ji
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Xuejiao Zhang
- Key Laboratory of Pollution Ecology and Environmental Engineering, Shenyang Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang, 110016, China.
| | - Dechun Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China. and Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, PR China. and Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| |
Collapse
|
29
|
Meireson A, Tavernier SJ, Van Gassen S, Sundahl N, Demeyer A, Spaas M, Kruse V, Ferdinande L, Van Dorpe J, Hennart B, Allorge D, Haerynck F, Decaestecker K, Rottey S, Saeys Y, Ost P, Brochez L. Immune Monitoring in Melanoma and Urothelial Cancer Patients Treated with Anti-PD-1 Immunotherapy and SBRT Discloses Tumor Specific Immune Signatures. Cancers (Basel) 2021; 13:cancers13112630. [PMID: 34071888 PMCID: PMC8198315 DOI: 10.3390/cancers13112630] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/21/2021] [Accepted: 05/25/2021] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Currently available biomarkers for response to checkpoint inhibitors are incomplete and predominantly focus on tumor tissue analysis e.g., tumor mutational burden, programmed cell death-ligand 1 (PD-L1) expression. Biomarkers in peripheral blood would allow a more dynamic monitoring and could offer a way for sequential adaptation of treatment strategy. We conducted an in-depth analysis of baseline and on-treatment systemic immune features in a cohort of stage III/IV melanoma and stage IV urothelial cancer (UC) patients treated with anti-programmed cell death-1 (anti-PD-1) therapy combined with stereotactic body radiotherapy (SBRT) in a similar regimen/schedule. Baseline immunity was clearly different between these two cohorts, indicating a less active immune landscape in UC patients. This study also detected signatures of proliferation in the CD8+ T-cell compartment pre-treatment and early after anti-PD-1 initiation that were positively correlated with clinical outcome in both tumor types. In addition our data support the biological relevance of PD-1/PD-L1 expression on circulating immune cell subsets, especially in melanoma. Abstract (1) Background: Blockade of the PD-1/PD-L1 pathway has revolutionized the oncology field in the last decade. However, the proportion of patients experiencing a durable response is still limited. In the current study, we performed an extensive immune monitoring in patients with stage III/IV melanoma and stage IV UC who received anti-PD-1 immunotherapy with SBRT. (2) Methods: In total 145 blood samples from 38 patients, collected at fixed time points before and during treatment, were phenotyped via high-parameter flow cytometry, luminex assay and UPLC-MS/MS. (3) Results: Baseline systemic immunity in melanoma and UC patients was different with a more prominent myeloid compartment and a higher neutrophil to lymphocyte ratio in UC. Proliferation (Ki67+) of CD8+ T-cells and of the PD-1+/PD-L1+ CD8+ subset at baseline correlated with progression free survival in melanoma. In contrast a higher frequency of PD-1/PD-L1 expressing non-proliferating (Ki67−) CD8+ and CD4+ T-cells before treatment was associated with worse outcome in melanoma. In UC, the expansion of Ki67+ CD8+ T-cells and of the PD-L1+ subset relative to tumor burden correlated with clinical outcome. (4) Conclusion: This study reveals a clearly different immune landscape in melanoma and UC at baseline, which may impact immunotherapy response. Signatures of proliferation in the CD8+ T-cell compartment prior to and early after anti-PD-1 initiation were positively correlated with clinical outcome in both cohorts. PD-1/PD-L1 expression on circulating immune cell subsets seems of clinical relevance in the melanoma cohort.
Collapse
Affiliation(s)
- Annabel Meireson
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Dermatology Research Unit, Ghent University Hospital, 9000 Ghent, Belgium
| | - Simon J. Tavernier
- Centre for Primary Immunodeficiency Ghent, Primary Immune Deficiency Research Lab, Department of Internal Medicine and Pediatrics, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, 9000 Ghent, Belgium; (S.J.T.); (F.H.)
- VIB Center for Inflammation Research, Unit of Molecular Signal Transduction in Inflammation, 9000 Ghent, Belgium
| | - Sofie Van Gassen
- VIB Center for Inflammation Research, Unit of Data Mining and Modeling for Biomedicine, 9000 Ghent, Belgium;
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000 Ghent, Belgium
| | - Nora Sundahl
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, 9000 Ghent, Belgium
| | - Annelies Demeyer
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Dermatology Research Unit, Ghent University Hospital, 9000 Ghent, Belgium
| | - Mathieu Spaas
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, 9000 Ghent, Belgium
| | - Vibeke Kruse
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
| | | | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Pathology, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Benjamin Hennart
- Unité Fonctionnelle de Toxicologie, CHU Lille, F-59000 Lille, France; (B.H.); (D.A.)
- ULR 4483-IMPact de l’Environnement Chimique sur la Santé Humaine (IMPECS), Université de Lille, F-59000 Lille, France
| | - Delphine Allorge
- Unité Fonctionnelle de Toxicologie, CHU Lille, F-59000 Lille, France; (B.H.); (D.A.)
- ULR 4483-IMPact de l’Environnement Chimique sur la Santé Humaine (IMPECS), Université de Lille, F-59000 Lille, France
| | - Filomeen Haerynck
- Centre for Primary Immunodeficiency Ghent, Primary Immune Deficiency Research Lab, Department of Internal Medicine and Pediatrics, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, 9000 Ghent, Belgium; (S.J.T.); (F.H.)
| | - Karel Decaestecker
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Urology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Sylvie Rottey
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Medical Oncology, Ghent University Hospital, 9000 Ghent, Belgium
| | - Yvan Saeys
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- VIB Center for Inflammation Research, Unit of Data Mining and Modeling for Biomedicine, 9000 Ghent, Belgium;
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000 Ghent, Belgium
| | - Piet Ost
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Department of Radiation Oncology and Experimental Cancer Research, Ghent University Hospital, 9000 Ghent, Belgium
| | - Lieve Brochez
- Cancer Research Institute Ghent (CRIG), Ghent University, 9000 Ghent, Belgium; (A.M.); (N.S.); (A.D.); (M.S.); (V.K.); (J.V.D.); (K.D.); (S.R.); (Y.S.); (P.O.)
- Dermatology Research Unit, Ghent University Hospital, 9000 Ghent, Belgium
- Correspondence:
| |
Collapse
|
30
|
Liu X, Yuan Z, Tang Z, Chen Q, Huang J, He L, Chen T. Selenium-driven enhancement of synergistic cancer chemo-/radiotherapy by targeting nanotherapeutics. Biomater Sci 2021; 9:4691-4700. [PMID: 34019044 DOI: 10.1039/d1bm00348h] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
To overcome drug resistance in hypoxic tumors and the limitations of radiation impedance and radiation dose, we developed a nano-radiosensitizer to improve the efficacy of cancer radiotherapy. We used multifunctional mesoporous silica nanoparticles (MSNs) as the carriers for a novel anticancer selenadiazole derivative (SeD) and modified its surface with folic acid (FA) to enhance its cervical cancer-targeting effects, forming the nanosystem named SeD@MSNs-FA. Upon radiation, SeD@MSNs-FA inhibits the growth of cervical cancer cells by inducing apoptosis through the death receptor-mediated apoptosis pathway and S phase arrest, significantly improving the sensitivity of cervical cancer cells to X-ray radiation. The combined activity of SeD@MSN-FA and radiation can promote excessive production of intracellular reactive oxygen species (ROS) and induce cell apoptosis by affecting p53, protein kinase B (AKT), and mitogen-activated protein kinase (MAPK) pathways. Furthermore, SeD@MSNs-FA can effectively inhibit tumor growth of xenografted HeLa tumors in nude mice. The toxicity analysis of SeD@MSNs-FA nanoparticles in vivo and the histological analysis performed in the mouse model showed that under the current experimental conditions, the nanoparticles induced no significant damage to the heart, liver, spleen, lungs, kidneys, or other major organs. Taken together, this study provides a translational nanomedicine-based strategy for the simultaneous chemo- and radiotherapy of cervical cancer and sheds light on potential mechanisms that can be used to overcome radiotherapeutic resistance.
Collapse
Affiliation(s)
- Xinxin Liu
- Department of Neurology and Stroke Center of The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Zhongwen Yuan
- Department of Neurology and Stroke Center of The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Zheng Tang
- Department of Neurology and Stroke Center of The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Qi Chen
- Department of Neurology and Stroke Center of The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Jiarun Huang
- Department of Neurology and Stroke Center of The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China.
| | - Lizhen He
- Department of Neurology and Stroke Center of The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China. and The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Guangzhou 510632, China
| | - Tianfeng Chen
- Department of Neurology and Stroke Center of The First Affiliated Hospital, and Department of Chemistry, Jinan University, Guangzhou 510632, China. and The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, Guangzhou 510632, China
| |
Collapse
|
31
|
Construction of cancer-on-a-chip for drug screening. Drug Discov Today 2021; 26:1875-1890. [PMID: 33731317 DOI: 10.1016/j.drudis.2021.03.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 10/16/2020] [Accepted: 03/09/2021] [Indexed: 12/13/2022]
Abstract
Cancer-on-a-chip has effectively contributed to the development of drug screening, holding great promise for more convenient and reliable drug development as well as personalized drug administration.
Collapse
|
32
|
Badia-Ramentol J, Linares J, Gómez-Llonin A, Calon A. Minimal Residual Disease, Metastasis and Immunity. Biomolecules 2021; 11:130. [PMID: 33498251 PMCID: PMC7909268 DOI: 10.3390/biom11020130] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Progression from localized to metastatic disease requires cancer cells spreading to distant organs through the bloodstream. Only a small proportion of these circulating tumor cells (CTCs) survives dissemination due to anoikis, shear forces and elimination by the immune system. However, all metastases originate from CTCs capable of surviving and extravasating into distant tissue to re-initiate a tumor. Metastasis initiation is not always immediate as disseminated tumor cells (DTCs) may enter a non-dividing state of cell dormancy. Cancer dormancy is a reversible condition that can be maintained for many years without being clinically detectable. Subsequently, late disease relapses are thought to be due to cancer cells ultimately escaping from dormant state. Cancer dormancy is usually associated with minimal residual disease (MRD), where DTCs persist after intended curative therapy. Thus, MRD is commonly regarded as an indicator of poor prognosis in all cancers. In this review, we examine the current understanding of MRD and immunity during cancer progression to metastasis and discuss clinical perspectives for oncology.
Collapse
Affiliation(s)
| | | | | | - Alexandre Calon
- Cancer Research Program, Hospital del Mar Medical Research Institute (IMIM), 08003 Barcelona, Spain; (J.B.-R.); (J.L.); (A.G.-L.)
| |
Collapse
|
33
|
Zhou M, Wang X, Lin S, Cheng Y, Zhao S, Lin J, Fang Z, Lou Z, Qin L, Wei H. Multifunctional STING-Activating Mn 3 O 4 @Au-dsDNA/DOX Nanoparticle for Antitumor Immunotherapy. Adv Healthc Mater 2020; 9:e2000064. [PMID: 32484320 DOI: 10.1002/adhm.202000064] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 05/08/2020] [Indexed: 12/18/2022]
Abstract
The promise of immunotherapy for cancer therapy has not been fully fulfilled because portions of tumors are immunosuppressive. To tackle this challenge, the initiation of immune system by stimulator of interferon genes (STING) pathway is explored and multifunctional STING-activating nanoparticles are rationally designed for synergistic antitumor therapy. The STING-activating nanoparticles have a formulation of Mn3 O4 @Au-dsDNA/DOX, where dsDNA is used to activate STING for immunotherapy and doxorubicin (DOX) is chosen as a model drug for chemotherapy. The STING-mediated immunity is activated, inducing interferon-β (IFN-β) production, increasing T cell priming, and enhancing effector T cell infiltration. Combined with chemotherapy, STING-mediated immunotherapy shows good antitumor efficacy by inhibiting tumor growth and prolonging survival rate in vivo. The promise of cancer immunotherapy can be fulfilled by combining novel antitumor immunity with innovative nanotechnology, and chemotherapy and targeted therapies.
Collapse
Affiliation(s)
- Min Zhou
- College of Engineering and Applied SciencesNanjing National Laboratory of MicrostructuresJiangsu Key Laboratory of Artificial Functional MaterialsNanjing University Nanjing Jiangsu 210093 China
| | - Xiaoyu Wang
- College of Engineering and Applied SciencesNanjing National Laboratory of MicrostructuresJiangsu Key Laboratory of Artificial Functional MaterialsNanjing University Nanjing Jiangsu 210093 China
| | - Shichao Lin
- College of Engineering and Applied SciencesNanjing National Laboratory of MicrostructuresJiangsu Key Laboratory of Artificial Functional MaterialsNanjing University Nanjing Jiangsu 210093 China
| | - Yuan Cheng
- College of Engineering and Applied SciencesNanjing National Laboratory of MicrostructuresJiangsu Key Laboratory of Artificial Functional MaterialsNanjing University Nanjing Jiangsu 210093 China
| | - Sheng Zhao
- College of Engineering and Applied SciencesNanjing National Laboratory of MicrostructuresJiangsu Key Laboratory of Artificial Functional MaterialsNanjing University Nanjing Jiangsu 210093 China
| | - Junshu Lin
- College of Engineering and Applied SciencesNanjing National Laboratory of MicrostructuresJiangsu Key Laboratory of Artificial Functional MaterialsNanjing University Nanjing Jiangsu 210093 China
- Department of BiomaterialsCollege of MaterialsXiamen University Xiamen Fujian 361005 China
| | - Zhuoyao Fang
- State Key Laboratory of Pollution Control and Resource ReuseSchool of the EnvironmentNanjing University Nanjing Jiangsu 210023 China
| | - Zhangping Lou
- College of Engineering and Applied SciencesNanjing National Laboratory of MicrostructuresJiangsu Key Laboratory of Artificial Functional MaterialsNanjing University Nanjing Jiangsu 210093 China
| | - Li Qin
- College of Engineering and Applied SciencesNanjing National Laboratory of MicrostructuresJiangsu Key Laboratory of Artificial Functional MaterialsNanjing University Nanjing Jiangsu 210093 China
| | - Hui Wei
- College of Engineering and Applied SciencesNanjing National Laboratory of MicrostructuresJiangsu Key Laboratory of Artificial Functional MaterialsNanjing University Nanjing Jiangsu 210093 China
- State Key Laboratory of Analytical Chemistry for Life ScienceState Key Laboratory of Coordination ChemistrySchool of Chemistry and Chemical EngineeringNanjing University Nanjing Jiangsu 210023 China
| |
Collapse
|
34
|
Ye M, Hu W, He M, Li C, Zhai S, Liu Z, Wang Y, Zhang H, Li C. Deep imaging for visualizing nitric oxide in lipid droplets: discovering the relationship between nitric oxide and resistance to cancer chemotherapy drugs. Chem Commun (Camb) 2020; 56:6233-6236. [PMID: 32373871 DOI: 10.1039/d0cc01856b] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A near-infrared two-photon fluorescent probe (TAN) was synthesized for selective detection and deep-depth imaging of NO in lipid droplets. All results demonstrated that NO production in lipid droplets is closely correlated with the resistance to anti-tumor drugs, and NO inhibitors can effectively improve the efficacy of chemotherapeutic agents.
Collapse
Affiliation(s)
- Miantai Ye
- Key Laboratory of Analytical Chemistry of the State Ethnic Affairs Commission, College of Chemistry and Material Science, South-central University for Nationalities, Wuhan 430074, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Yang Q, Xiao Y, Yin Y, Li G, Peng J. Erythrocyte Membrane-Camouflaged IR780 and DTX Coloading Polymeric Nanoparticles for Imaging-Guided Cancer Photo-Chemo Combination Therapy. Mol Pharm 2019; 16:3208-3220. [PMID: 31145853 DOI: 10.1021/acs.molpharmaceut.9b00413] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Conventional systemic chemotherapy leads to poor therapeutic outcomes at moments in cancer therapy because the nontargeting anticancer drug release results in adverse effects and consequently drug resistance. The combination therapeutic strategy provides an alternative way to solve the conundrums. Herein, drug delivery systems with a rational design and tumor-targeting abilities become the ideal carriers for combinatorial therapy. IR780 iodide possesses near-infrared fluorescence intensity for fluorescence imaging (FI) and photothermal conversion for photoacoustic imaging (PAI), which also can be employed for tumor phototherapy (including photothermal therapy and photodynamic therapy). However, hydrophobicity and rapid elimination in vivo limit its biomedical applications. Furthermore, the hydrophobicity and high crystallization of IR780 result in poor drug-loading capacity and low stability. In this study, the high-pressure homogenization method was utilized for hydrophobic molecular IR780 and DTX coloading to construct IR780/DTX-PCEC nanoparticles which exhibit narrow size distribution and satisfactory drug-loading capacity. With further erythrocyte membrane [red blood cell (RBC)] camouflaging, the obtained IR780/DTX-PCEC@RBC nanoparticles present desired stability and prolonged circulation time in vivo. Additionally, the IR780/DTX-PCEC@RBC nanoparticles not only can be employed as a FI/PAI dual model imaging probe but also exhibit the property for phototherapy and chemotherapy of tumors. Based on the therapeutic outcome of combination therapy, the IR780/DTX-PCEC@RBC nanoparticles can serve as promising FI- and PAI-guided photo-chemo combination therapy agents for the future treatment of breast cancer.
Collapse
Affiliation(s)
- Qian Yang
- School of Pharmacy , Chengdu Medical College , No. 783, Xindu Avenue , Xindu District, Chengdu 610500 , Sichuan , P. R. China
| | - Yao Xiao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University and Collaborative Innovation Center of Biotherapy , Chengdu , Sichuan 610041 , P. R. China
| | - Yanlong Yin
- School of Pharmacy , Chengdu Medical College , No. 783, Xindu Avenue , Xindu District, Chengdu 610500 , Sichuan , P. R. China
| | - Gaoyin Li
- School of Pharmacy , Chengdu Medical College , No. 783, Xindu Avenue , Xindu District, Chengdu 610500 , Sichuan , P. R. China
| | - Jinrong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital , Sichuan University and Collaborative Innovation Center of Biotherapy , Chengdu , Sichuan 610041 , P. R. China
| |
Collapse
|
36
|
Antonangeli F, Zingoni A, Soriani A, Santoni A. Senescent cells: Living or dying is a matter of NK cells. J Leukoc Biol 2019; 105:1275-1283. [DOI: 10.1002/jlb.mr0718-299r] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 01/30/2019] [Accepted: 02/03/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Fabrizio Antonangeli
- Department of Molecular MedicineSapienza University of RomeLaboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti Rome Italy
| | - Alessandra Zingoni
- Department of Molecular MedicineSapienza University of RomeLaboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti Rome Italy
| | - Alessandra Soriani
- Department of Molecular MedicineSapienza University of RomeLaboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti Rome Italy
| | - Angela Santoni
- Department of Molecular MedicineSapienza University of RomeLaboratory affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti Rome Italy
- Neuromed I.R.C.C.S. Pozzilli (IS) Italy
| |
Collapse
|